PHARMACEUTICAL MANUFACTURING HANDBOOK Production and Processes
SHAYNE COX GAD, PH.D., D.A.B.T. 
Gad Consulting Services 
Cary, North Carolina 



CONTRIBUTORS 
Susanna Abrahms e n - Alami, AstraZeneca R & D Lund, Lund, Sweden, Oral 
Extended - Release Formulations 
James Agalloco, Agalloco & Associates, Belle Mead, New Jersey, Sterile Product 
Manufacturing 
Fakhrul Ahsan, Texas Tech University, Amarillo, Texas, Nasal Delivery of Peptide 
and Nonpeptide Drugs 
James Akers, Akers Kennedy & Associates, Kansas City, Missouri, Sterile Product 
Manufacturing 
Raid G. Alany, The University of Auckland, Auckland, New Zealand, Ocular Drug 
Delivery; Microemulsions as Drug Delivery Systems 
Monique Alric, Universit e d ’ Auvergne, Clermont - Ferrand, France, Recombinant 
Saccharomyces Cerevisiae as New Drug Delivery System to Gut: In Vitro Validation 
and Oral Formulation 
Sacide Alsoy Altinkaya, Izmir Institute of Technology, Urla - Izmir, Turkey, Controlled 
Release of Drugs from Tablet Coatings 
Maria Helena Amaral, University of Porto, Porto, Portugal, Vaginal Drug 
Delivery 
Anil Kumar Anal, Living Cell Technologies (Global) Limited, Auckland, New 
Zealand, Controlled - Release Dosage Forms 
Gavin Andrews, Queen ’ s University Belfast, Belfast, Northern Ireland, Effects of 
Grinding in Pharmaceutical Tablet Production 
Sophia G. Antimisiaris, School of Pharmacy, University of Patras, Rio, Greece, 
Liposomes and Drug Delivery 

vi CONTRIBUTORS 
Robert D. Arnold, The University of Georgia, Athens, Georgia, Biotechnology - 
Derived Drug Product Development 
C. Scott Asbill, Samford University, Birmingham, Alabama, Transdermal Drug 
Delivery 
Maria Fernanda Bahia, University of Porto, Porto, Portugal, Vaginal Drug 
Delivery 
Bernard Bataille, University of Montpelier 1, Montpellier, France, Tablet Design 
Gerald W. Becker, SSCI, West Lafayette, Indiana, Biotechnology - Derived Drug 
Product Development; Regulatory Considerations in Approval of Follow - On 
Protein Drug Products 
B. Wayne Bequette, Rensselaer Polytechnic Institute, Troy, New York, From Pilot 
Plant to Manufacturing: Effect of Scale - Up on Operation of Jacketed Reactors 
Erem Bilensoy, Hacettepe University Faculty of Pharmacy, Ankara, Turkey, Cyclodextrin 
- Based Nanomaterials in Pharmaceutical Field 
St e phanie Blanquet, Universit e d ’ Auvergne, Clermont - Ferrand, France, Recombinant 
Saccharomyces Cerevisiae as New Drug Delivery System to Gut: In Vitro 
Validation and Oral Formulation 
Gary W. Bumgarner, Samford University, Birmingham, Alabama, Transdermal 
Drug Delivery 
Isidoro Caraballo, University of Sevilla, Seville, Spain, Tablet Design 
Stephen M. Carl, Purdue University, West Lafayette, Indiana, Biotechnology - 
Derived Drug Product Development; Regulatory Considerations in Approval of 
Follow - On Protein Drug Products 
Sudhir S. Chakravarthi, University of Nebraska Medical Center, College of Pharmacy, 
Omaha, Nebraska, Biodegradable Nanoparticles 
D.F. Chowdhury, University of Oxford, Oxford, United Kingdom, Pharmaceutical 
Nanosystems: Manufacture, Characterization, and Safety 
Barbara R. Conway, Aston University, Birmingham, United Kingdom, Solid Dosage 
Forms 
Jos e das Neves, University of Porto, Porto, Portugal, Vaginal Drug Delivery 
Osama Abu Diak, Queen ’ s University Belfast, Belfast, Northern Ireland, Effects 
of Grinding in Pharmaceutical Tablet Production 
Brit S. Farstad, Instititue for Energy Technology, Isotope Laboratories, Kjeller, 
Norway, Radiopharmaceutical Manufacturing 
Dimitrios G. Fatouros, School of Pharmacy and Biomedical Sciences, Portsmouth, 
England, Liposomes and Drug Delivery 
Jelena Filipovi c - Gr i , Faculty of Pharmacy and Biochemistry, University of 
Zagreb, Zagreb, Croatia, Nasal Powder Drug Delivery 
c c

CONTRIBUTORS vii 
Eddy Castellanos Gil, Center of Pharmaceutical Chemistry and University of 
Havana, Havana, Cuba; University of Sevilla, Seville, Spain; University of Montpelier 
1, Montpellier, France, Tablet Design 
Anita Hafner, Faculty of Pharmacy and Biochemistry, University of Zagreb, 
Zagreb, Croatia, Nasal Powder Drug Delivery 
A. Atilla Hincal, Hacettepe University Faculty of Pharmacy, Ankara, Turkey, 
Cyclodextrin - Based Nanomaterials in Pharmaceutical Field 
Michael Hindle, Virginia Commonwealth University, Richmond, Virginia, Aerosol 
Drug Delviery 
Bhaskara R. Jasti, University of the Pacifi c, Stockton, California, Semisolid Dosages: 
Ointments, Creams, and Gels 
Yiguang Jin, Beijing Institute of Radiation Medicine, Beijing, China, Nanotechnology 
in Pharmaceutical Manufacturing 
David Jones, Queen ’ s University Belfast, Belfast, Northern Ireland, Effects of 
Grinding in Pharmaceutical Tablet Production 
Anne Juppo, University of Helsinki, Helsinki, Finland, Oral Extended - Release 
Formulations 
Paraskevi Kallinteri, Medway School of Pharmacy, Universities of Greenwich and 
Kent, England, Liposomes and Drug Delivery 
Gregory T. Knipp, Purdue University, West Lafayette, Indiana, Biotechnology - 
Derived Drug Product Development; Regulatory Considerations in Approval of 
Follow - On Protein Drug Products 
Anette Larsson, Chalmers University of Technology, G o teborg, Sweden, Oral 
Extended - Release Formulations 
Beom - Jin Lee, Kangwon National University, Chuncheon, Korea, Pharmaceutical 
Preformulation: Physiochemical Properties of Excipients and Powders and Tablet 
Characterization 
Xiaoling Li, University of the Pacifi c, Stockton, California, Semisolid Dosages: 
Ointments, Creams, and Gels 
David J. Lindley, Purdue University, West Lafayette, Indiana, Biotechnology - 
Derived Drug Product Development 
Roberto Londono, Washington State University, Pullman, Washington, Liquid 
Dosage Forms 
Ravichandran Mahalingam, University of the Pacifi c, Stockton, California, Semisolid 
Dosages: Ointments, Creams, and Gels 
Kenneth R. Morris, Purdue University, West Lafayette, Indiana, Biotechnology - 
Derived Drug Product Development; Regulatory Considerations in Approval of 
Follow - On Protein Drug Products 
Erin Oliver, Rutgers, The State University of New Jersey, Piscataway, New Jersey, 
Biotechnology - Derived Drug Product Development; Regulatory Considerations 
in Approval of Follow - On Protein Drug Products 

viii CONTRIBUTORS 
Iv a n Pe n uelas, University of Navarra, Pamplona, Spain, Radiopharmaceutical 
Manufacturing 
Omanthanu P. Perumal, South Dakota State University, Brookings, South Dakota, 
Role of Preformulation in Development of Solid Dosage Forms 
Katharina M. Picker - Freyer, Martin - Luther - University Halle - Wittenberg, Institute 
of Pharmaceutics and Biopharmaceutics, Halle/Saale, Germany, Tablet Production 
Systems 
Satheesh K. Podaralla, South Dakota State University, Brookings, South Dakota, 
Role of Preformulation in Development of Solid Dosage Forms 
Dennis H. Robinson, University of Nebraska Medical Center, College of Pharmacy, 
Omaha, Nebraska, Biodegradable Nanoparticles 
Arcesio Rubio, Caracas, Venezuela, Liquid Dosage Forms 
Maria V. Rubio - Bonilla, Research Associate, College of Pharmacy, Washington 
State University, Pullman, Washington, Liquid Dosage Forms 
Ilva D. Rupenthal, The University of Auckland, Auckland, New Zealand, Ocular 
Drug Delivery 
Maria In e s Rocha Miritello Santoro, Department of Pharmacy, Faculty of Pharmaceutical 
Sciences, University of S a o Paulo, S a o Paulo, Brazil, Packaging and 
Labeling 
Helton M.M. Santos, University of Coimbra, Coimbra, Portugal, Tablet 
Compression 
Raymond K. Schneider, Clemson University, Clemson, South Carolina, Clean - 
Facility Design, Construction, and Maintenance Issues 
Anil Kumar Singh, Department of Pharmacy, Faculty of Pharmaceutical Sciences, 
University of S a o Paulo, S a o Paulo, Brazil, Packaging and Labeling 
Jo a o J.M.S. Sousa, University of Coimbra, Coimbra, Portugal, Tablet 
Compression 
Shunmugaperumal Tamilvanan, University of Antwerp, Antwerp, Belgium, Progress 
in Design of Biodegradable Polymer - Based Microspheres for Parenteral 
Controlled Delivery of Therapeutic Peptide/Protein; Oil - in - Water Nanosized 
Emulsions: Medical Applications 
Chandan Thomas, Texas Tech University, Amarillo, Texas, Nasal Delivery of 
Peptide and Nonpeptide Drugs 
Gavin Walker, Queen ’ s University Belfast, Belfast, Northern Ireland, Effects of 
Grinding in Pharmaceutical Tablet Production 
Jingyuan Wen, The University of Auckland, Auckland, New Zealand, Microemulsions 
as Drug Delivery Systems 
Hui Zhai, Queen ’ s University Belfast, Belfast, Northern Ireland, Effects of Grinding 
in Pharmaceutical Tablet Production 

ix 
CONTENTS 
PREFACE xiii 
SECTION 1 MANUFACTURING SPECIALTIES 1 
1.1 Biotechnology-Derived Drug Product Development 3 
Stephen M. Carl, David J. Lindley, Gregory T. Knipp, Kenneth R. Morris, 
Erin Oliver, Gerald W. Becker, and Robert D. Arnold 
1.2 Regulatory Considerations in Approval on Follow-On Protein 
Drug Products 33 
Erin Oliver, Stephen M. Carl, Kenneth R. Morris, Gerald W. Becker, and 
Gregory T. Knipp 
1.3 Radiopharmaceutical Manufacturing 59 
Brit S. Farstad and Ivan Penuelas 
SECTION 2 ASEPTIC PROCESSING 97 
2.1 Sterile Product Manufacturing 99 
James Agalloco and James Akers 
SECTION 3 FACILITY 137 
3.1 From Pilot Plant to Manufacturing: Effect of Scale-Up on 
Operation of Jacketed Reactors 139 
B. Wayne Bequette

x CONTENTS 
3.2 Packaging and Labeling 159 
Maria Ines Rocha Miritello Santoro and Anil Kumar Singh 
3.3 Clean-Facility Design, Construction, and Maintenance Issues 201 
Raymond K. Schneider 
SECTION 4 NORMAL DOSAGE FORMS 233 
4.1 Solid Dosage Forms 235 
Barbara R. Conway 
4.2 Semisolid Dosages: Ointments, Creams, and Gels 267 
Ravichandran Mahalingam, Xiaoling Li, and Bhaskara R. Jasti 
4.3 Liquid Dosage Forms 313 
Maria V. Rubio-Bonilla, Roberto Londono, and Arcesio Rubio 
SECTION 5 NEW DOSAGE FORMS 345 
5.1 Controlled-Release Dosage Forms 347 
Anil Kumar Anal 
5.2 Progress in the Design of Biodegradable Polymer-Based 
Microspheres for Parenteral Controlled Delivery of Therapeutic 
Peptide/Protein 393 
Shunmugaperumal Tamilvanan 
5.3 Liposomes and Drug Delivery 443 
Sophia G. Antimisiaris, Paraskevi Kallinteri, and Dimitrios G. Fatouros 
5.4 Biodegradable Nanoparticles 535 
Sudhir S. Chakravarthi and Dennis H. Robinson 
5.5 Recombinant Saccharomyces cerevisiae as New Drug Delivery 
System to Gut: In Vitro Validation and Oral Formulation 565 
Stephanie Blanquet and Monique Alric 
5.6 Nasal Delivery of Peptide and Nonpeptide Drugs 591 
Chandan Thomas and Fakhrul Ahsan 
5.7 Nasal Powder Drug Delivery 651 
Jelena Filipovi -Gr i and Anita Hafner 
5.8 Aerosol Drug Delivery 683 
Michael Hindle 
5.9 Ocular Drug Delivery 729 
Ilva D. Rupenthal and Raid G. Alany 
5.10 Microemulsions as Drug Delivery Systems 769 
Raid G. Alany and Jingyuan Wen 
c c c

CONTENTS xi 
5.11 Transdermal Drug Delivery 793 
C. Scott Asbill and Gary W. Bumgarner 
5.12 Vaginal Drug Delivery 809 
Jose das Neves, Maria Helena Amaral, and Maria Fernanda Bahia 
SECTION 6 TABLET PRODUCTION 879 
6.1 Pharmaceutical Preformulation: Physicochemical Properties of 
Excipients and Powers and Tablet Characterization 881 
Beom-Jin Lee 
6.2 Role of Preformulation in Development of Solid Dosage Forms 933 
Omathanu P. Perumal and Satheesh K. Podaralla 
6.3 Tablet Design 977 
Eddy Castellanos Gil, Isidoro Caraballo, and Bernard Bataille 
6.4 Tablet Production Systems 1053 
Katharina M. Picker-Freyer 
6.5 Controlled Release of Drugs from Tablet Coatings 1099 
Sacide Alsoy Altinkaya 
6.6 Tablet Compression 1133 
Helton M. M. Santos and Joao J. M. S. Sousa 
6.7 Effects of Grinding in Pharmaceutical Tablet Production 1165 
Gavin Andrews, David Jones, Hui Zhai, Osama Abu Diak, and 
Gavin Walker 
6.8 Oral Extended-Release Formulations 1191 
Anette Larsson, Susanna Abrahmsen-Alami, and Anne Juppo 
SECTION 7 ROLE OF NANOTECHNOLOGY 1223 
7.1 Cyclodextrin-Based Nanomaterials in Pharmaceutical Field 1225 
Erem Bilensoy and A. Attila Hincal 
7.2 Nanotechnology in Pharmaceutical Manufacturing 1249 
Yiguang Jin 
7.3 Pharmaceutical Nanosystems: Manufacture, Characterization, 
and Safety 1289 
D. F. Chowdhury 
7.4 Oil-in-Water Nanosized Emulsions: Medical Applications 1327 
Shunmugaperumal Tamilvanan 
INDEX 1367


xiii 
PREFACE 
This Handbook of Manufacturing Techniques focuses on a new aspect of the drug 
development challenge: producing and administering the physical drug products 
that we hope are going to provide valuable new pharmacotherapeutic tools in medicine. 
These 34 chapters cover the full range of approaches to developing and producing 
new formulations and new approaches to drug delivery. Also addressed are 
approaches to the issues of producing and packaging these drug products (that is, 
formulations). The area where the most progress is possible in improving therapeutic 
success with new drugs is that of better delivery of active drug molecules to the 
therapeutic target tissue. In this volume, we explore current and new approaches to 
just this issue across the full range of routes (oral, parenteral, topical, anal, nasal, 
aerosol. ocular, vaginal, and transdermal) using all sorts of forms of formulation. 
The current metrics for success of new drugs in development once they enter the 
clinic (estimated at ranging from as low as 2% for oncology drugs to as high as 10% 
for oral drugs) can likely be leveraged in the desired direction most readily by 
improvements in this area of drug delivery. 
The Handbook of Manufacturing Techniques seeks to cover the entire range of 
available approaches to getting a pure drug (as opposed to a combination product) 
into the body and to its therapeutic tissue target. Thanks to the persistent efforts of 
Michael Leventhal, these 34 chapters, which are written by leading practitioners in 
each of these areas, provide coverage of the primary approaches to these fundamental 
problems that stand in the way of so many potentially successful pharmacotherapeutic 
interventions. 


MANUFACTURING SPECIALTIES 
SECTION 1


3 
1.1 
BIOTECHNOLOGY - DERIVED DRUG 
PRODUCT DEVELOPMENT 
Stephen M. Carl, 1 David J. Lindley, 1 Gregory T. Knipp, 1 
Kenneth R. Morris, 1 Erin Oliver, 2 Gerald W. Becker, 3 and 
Robert D. Arnold 4 
1 Purdue University, West Lafayette, Indiana 
2 Rutgers, The State University of New Jersey, Piscataway, New Jersey 
3 SSCI, West Lafayette, Indiana 
4 The University of Georgia, Athens, Georgia 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc. 
Contents 
1.1.1 Introduction 
1.1.2 Formulation Assessment 
1.1.2.1 Route of Administration and Dosage 
1.1.2.2 Pharmacokinetic Implications to Dosage Form Design 
1.1.2.3 Controlled - Release Delivery Systems 
1.1.3 Analytical Method Development 
1.1.3.1 Traditional and Biophysical Analytical Methodologies 
1.1.3.2 Stability - Indicating Methodologies 
1.1.3.3 Method Validation and Transfer 
1.1.4 Formulation Development 
1.1.4.1 Processing Materials and Equipment 
1.1.4.2 Container Closure Systems 
1.1.4.3 Sterility Assurance 
1.1.4.4 Excipient Selection 
1.1.5 Drug Product Stability 
1.1.5.1 Defi ning Drug Product Storage Conditions 
1.1.5.2 Mechanisms of Protein and Peptide Degradation 
1.1.5.3 Photostability 
1.1.5.4 Mechanical Stress 
1.1.5.5 Freeze – Thaw Considerations and Cryopreservation 
1.1.5.6 Use Studies 
1.1.5.7 Container Closure Integrity and Microbiological Assessment 
1.1.5.8 Data Interpretation and Assessment 

4 BIOTECHNOLOGY-DERIVED DRUG PRODUCT DEVELOPMENT 
1.1.6 Quality by Design and Scale - Up 
1.1.6.1 Unit Operations 
1.1.6.2 Bioburden Considerations 
1.1.6.3 Scale - Up and Process Changes 
1.1.7 Concluding Remarks 
References 
1.1.1 INTRODUCTION 
Although the origins of the fi rst biological and/or protein therapeutics can be 
traced to insulin in 1922, the fi rst biotechnology - derived pharmaceutical drug 
product approved in the United States was Humulin in 1982. In the early stages 
of pharmaceutical biotechnology, companies that specialized primarily in the development 
of biologicals were the greatest source of research and development in 
this area. Recent advances in molecular and cellular biological techniques and 
the potential clinical benefi ts of biotechnology drug products have led to a substantial 
increase in their development by biotechnology and traditional pharmaceutical 
companies . In terms of pharmaceuticals, the International Conference on 
Harmonization (ICH) loosely defi nes biotechnology - derived products with biological 
origin products as those that are “ well - characterized proteins and polypeptides, 
their derivatives and products of which they are components, and which are 
isolated from tissues, body fl uids, cell cultures, or produced using rDNA technology 
” [1] . In practical terms, biological and biotechnology - derived pharmaceutical 
agents encompass a number of therapeutic classes, including cytokines, 
erythropoietins, plasminogen activators, blood plasma factors, growth hormones 
and growth factors, insulins, monoclonal antibodies, and vaccines [1] . Additionally, 
short interfering and short hairpin ribonucleic acids (siRNA, shRNA) and antisense 
oligonucleotide therapies are generally characterized as biotechnology - 
derived products. 
According to the biotechnology advocacy group, The Biotechnology Industry 
Organization (BIO), pharmaceutical - based biotechnology represents over a $ 30 
billion dollar a year industry and is directly responsible for the production of 
greater than 160 drug therapeutics and vaccines [2] . Furthermore, there are more 
than 370 biotechnology - derived drug products and vaccines currently in clinical 
trials around the world, targeting more than 200 diseases, including various cancers, 
Alzheimer ’ s disease, heart disease, diabetes, multiple sclerosis, acquired immunodefi 
ciency syndrome (AIDS), and arthritis. While the clinical value of these 
products is well recognized, a far greater number of biotechnology - derived drug 
products with therapeutic potential for life - altering diseases have failed in 
development. 
As the appreciation of the clinical importance and commercial potential for biological 
products grows, new challenges are arising based on the many technological 
limitations related to the development and marketing of these complex agents. 
Additionally, the intellectual property protection of an associated agent might not 

provide a suffi cient window to market and regain the costs associated with the discovery, 
research, development, and scale - up of these products. Therefore, to properly 
estimate the potential return on investment, a clear assessment of potential 
therapeutic advantages and disadvantages, such as the technological limitations in 
the rigorous characterization required of these complex therapeutic agents to gain 
Food and Drug Administration (FDA) approval, is needed prior to initiating 
research. Clearly, research focused on developing methodologies to minimize these 
technological limitations is needed. In doing so we hypothesize the attrition rate 
can be reduced and the number of companies engaged in the development of biotechnology 
- derived products and diversity of products will continue to expand. 
Technological limitations have limited the development of follow - on, or generic 
biopharmaceutical products that have lost patent protection. In fact, the potential 
pitfalls associated with developing these compounds are so diverse that regulatory 
guidance concerning follow - on biologics is relatively obscure and essentially notes 
that products will be assessed on a case - by - case basis. The reader is encouraged to 
see Chapter 1.2 for a more detailed discussion concerning regulatory perspectives 
pertaining to follow - on biologics. 
Many of the greatest challenges in producing biotechnology - derived pharmaceuticals 
are encountered in evaluating and validating the chemical and physical nature 
of the host expression system and the subsequent active pharmaceutical ingredient 
(API) as they are transferred from discovery through to the development and marketing 
stages. Although this area is currently a hotbed of research and is progressing 
steadily, limitations in analytical technologies are responsible for a high degree of 
attrition of these compounds. The problem is primarily associated with limited 
resolution of the analytical technologies utilized for product characterization. For 
example, without the ability to resolve small differences in secondary or tertiary 
structure, linking changes to product performance or clinical response is impossible. 
The biological activity of traditional small molecules is related directly to their 
structure and can be determined readily by nuclear magnetic resonance (NMR), 
X - Ray crystallography (X - ray), mass spectrometry (MS), and/or a combination of 
other spectroscopic techniques. However, methodologies utilized for characterizing 
biological agents are limited by resolution and reproducibility. For instance, circular 
dichroism (CD) is generally considered a good method to determine secondary 
structural elements and provides some information on the folding patterns (tertiary 
structure) of proteins. However, CD suffers from several limitations, including a 
lower resolution that is due in part to the sequence libraries used to deconvolute 
the spectra. To improve the reliability of determining the secondary and tertiary 
structural elements, these databases need to be developed further. An additional 
example is the utility of two - dimensional NMR (2D - NMR) for structural determination. 
While combining homonuclear and heteronuclear experimental techniques 
can prove useful in structural determination, there are challenges in that 2D - NMR 
for a protein could potentially generate thousands of signals. The ability to assign 
specifi c signals to each atom and their respective interactions is a daunting task. 
Resolution between the different amino acids in the primary sequence and their 
positioning in the covalent and folded structures become limited with increasing 
molecular weight. Higher dimensional techniques can be used to improve resolution; 
however, the resolution of these methods remains limited as the number of 
amino acids is increased. 
INTRODUCTION 5

6 BIOTECHNOLOGY-DERIVED DRUG PRODUCT DEVELOPMENT 
Understanding the limitations of the analytical methodologies utilized for product 
characterization has led to the development of new experimental techniques as well 
as the refi ned application of well - established techniques to this emerging fi eld. Only 
through application of a number of complementary techniques will development 
scientists be able to accurately characterize and develop clinically useful products. 
Unfortunately, much of the technology is still in its infancy and does not allow for 
a more in - depth understanding of the subtleties of peptide and protein processing 
and manufacturing. For instance, many of the analytical techniques utilized for 
characterization will evaluate changes to product conformation on the macroscopic 
level, such as potential denaturation or changes in folding, as observed with CD. 
However, these techniques do not afford the resolution to identify subtle changes 
in conformation that may either induce chemical or physical instabilities or unmask 
antigenic epitopes. 
Further limiting successful product development is a lack of basic understanding 
as to critical manufacturing processes that have the potential to affect the structural 
integrity and activity of biopharmaceuticals. As with traditional small molecules, 
stresses associated with the different unit operations may affect biopharmaceutical 
products differently. In contrast to traditional small molecules, there is considerable 
diffi culty in identifying potentially adverse affects, if any, that a particular unit operation 
may have on the clinically critical structural elements of a drug. Considering 
that many proteins exhibit a greater potential for degradation from shear stress, it 
is particularly important to assess any negative effects of mixing, transport through 
tubing, fi ltration, and fi lling operations. Essentially all unit operations for a given 
manufacturing process could create enough shear stress to induce minor structural 
changes that could lead to product failure. The diffi culty is establishing what degree 
of change will have an impact on the stability, bioactivity, or immunogenic potential 
of the compound. Unfortunately, unless exhaustive formulation development studies 
are conducted, coupled with a comprehensive spectrum of analytical methodologies, 
these effects may not be readily evident until after scale - up of the manufacturing 
process or, worse yet, in the clinical setting. Moreover, modeling shear and stress 
using fl uid dynamic structurally diverse molecules is a foreboding task. Extending 
these models to validate process analytical technologies (PAT) and incorporate 
critical quality by design (QbD) elements in the development process for a collection 
of biopharmaceuticals would be largely hindered by the daunting nature of the 
task at hand. 
The use of biological systems to produce these agents results in additional variability. 
Slight changes in nutrient profi le could affect growth patterns and protein 
expression of cultured cells. Furthermore, microbial contamination in the form of 
viruses, bacteria, fungi, and mycoplasma can be introduced during establishment of 
cell lines, cell culture/fermentation, capture and downstream processing steps, formulation 
and fi lling operations, or drug delivery [3] . Therefore, establishing the 
useful life span of purifi cation media and separation columns remains a critical issue 
for consistently producing intermediates and fi nal products that meet the defi ned 
quality and safety attributes of the product [4] . In short, understanding the proper 
processability and manufacturing controls needed has been a major hurdle that has 
kept broader development of biopharmaceutical products relatively limited. 
Notwithstanding the many technological hurdles to successfully develop a pharmaceutically 
active biotechnology product, they offer many advantages in terms of 

therapeutic potency, specifi city, and target design (not generally limited to a particular 
class or series of compounds). This is an iterative approach, whereby every new 
approved compound, new lessons, and applications to ensure successful product 
development are realized, thereby adding to our knowledge base and facilitating 
the development of future products. This chapter will discuss some of the fundamental 
issues associated with successful biopharmaceutical drug product development 
and aims to provide an understanding of the subtleties associated with their 
characterization, processing, and manufacturing. 
1.1.2 FORMULATION ASSESSMENT 
In order to select the most appropriate formulation and route of administration for 
a drug product, one must fi rst assess the properties of the API, the proposed therapeutic 
indication, and the requirements/limitations of the drug and the target patient 
population. Development teams are interdisciplinary comprised of individuals with 
broad expertise, for example, chemistry, biochemistry, bioengineering, and pharmaceutics, 
that can provide insight into the challenges facing successful product development. 
As such, knowledge gained through refi nement of the API manufacturing 
process through to lead optimization is vital to providing an initial starting point 
for success. Information acquired, for example, in the way of analytical development 
and API characterization, during drug discovery or early preclinical development 
that can be applied to fi nal drug product development may contribute to shorter 
development times of successful products. 
The host system utilized for API production is critical to the production of the 
fi nal product and will determine the basic and higher order physicochemical characteristics 
of the drug. Typically biopharmaceuticals are manufactured in Escherichia 
coli as prokaryotic and yeast and Chinese hamster ovary (CHO) cells as eukaryotic 
expression systems [5] . While general procedures for growth condition optimization 
and processing and purifi cation paradigms have emerged, differences in posttranslational 
modifi cations and host – system related impurities can exist even with relatively 
minor processing changes within a single production cell line [5] . Such changes 
have the potential to alter the biopharmaceutical properties of the active compound, 
its bioactivity , and its potential to elicit adverse events such as immunogenic reactions. 
These properties will be a common theme as they could potentially play a 
major role in both analytical and formulation development activities. 
During the process of lead optimization, characterization work is performed that 
would include a number of parameters that are critical to formulation and analytical 
development scientists. The following information is a minimalist look at what 
information should be available to support product development scientists: 
• Color 
• Particle size and morphology (for solid isolates) 
• Thermoanalytical profi le (e.g., Tg for lyophiles) 
• Hygroscopicity 
• Solubility with respect to pH 
• Apparent solution pH 
FORMULATION ASSESSMENT 7

8 BIOTECHNOLOGY-DERIVED DRUG PRODUCT DEVELOPMENT 
• Number and p Ka of ionizable groups 
• Amino acid sequence 
• Secondary and tertiary structural characteristics 
• Some stability parameters with respect to 
 pH 
 Temperature 
 Humidity 
 Light 
 Mechanical stress 
 Oxygen sensitivity 
• Impurity profi le 
 Misfolded/misaligned active 
 Potential isoforms 
 Expression system impurities 
• Potency [median inhibitory concentration (IC 50 )] 
• Animal Pharmacokinetic/Pharmacodynamic (PK/PD) and Tox profi les 
All of the above information will prove invaluable in determining the potential 
methods for rational drug delivery. Particular attention should be paid to the relative 
hygroscopicity of the API, of course, any stability information, as well as the 
impurity profi le and ADMET (absorption, distribution, metabolism, excretion, and 
toxicity) information. In short, the more information that is available when development 
activities are initiated, the easier it is to avoid common pitfalls and make 
development decisions more rationally. 
1.1.2.1 Route of Administration and Dosage 
Biologics are traditionally very potent molecules that may require only picomolar 
blood concentrations to elicit a therapeutic effect. Given that the amount of drug 
required per dosage will be commensurate with the relative potency of the molecule, 
small concentrations are generally required for any unit dose. Biopharmaceuticals 
typically have large molecular weights relative to conventional pharmaceutical 
agents, which may be increased further by posttranslational modifi cations. The pharmacokinetics 
(ADMET) of biotechnology products have been reviewed elsewhere 
[6] , but generally they have short circulating half - lives [7] . As such, biological products 
are most often delivered parenterally and formulated as solutions, suspensions, 
or lyophilized products for reconstitution [8, 9] . However, one must fi rst ascertain 
the potential physiological barriers to drug delivery and effi cacy before assessing 
potential routes of administration. These barriers may include actual physical barriers, 
such as a cell membrane, that could restrict the drug from reaching its site of 
action or chemical barriers, including pH or enzymatic degradation. Based on 
current drug delivery approaches, the proteinaceous nature of biological products 
limits their peroral delivery due to their susceptibility to proteases and peptidases 
present in the gastrointestinal tract as well as size limitations for permeating through 
absorptive enterocytes [10] . 

Diffi culties in peroral delivery have stimulated researchers to explore alternate 
delivery mechanisms for biologics, such as through the lungs or nasal mucosa [11, 
12] . Further, advances in technology and our understanding of the mechanisms 
limiting oral delivery of biotechnology products have led to innovative drug delivery 
approaches to achieve suffi cient oral bioavailability. However, no viable products 
have successfully reached the market [13] . As a result of the technological limitations 
inherent in biopharmaceutical delivery, these compounds are largely delivered 
parenterally through an injection or implant. 
When assessing the potential routes of administration, one must consider the 
physicochemical properties of the drug, its ADMET properties, the therapeutic 
indication, and the patient population, some of which are discussed below. Table 1 
provides a list of some of those factors that must be addressed when determining 
the most favorable route of administration and the subsequent formulation for 
delivery. Ideally the route of administration and subsequent formulation will be 
optimized after identifying critical design parameters to satisfy the needs of patients 
and health care professionals alike while maintaining the safety and effi cacy of the 
product. 
Parenteral administration is the primary route of delivering biopharmaceutical 
agents (e.g., insulin); however, issues associated with patient compliance with administration 
of short - acting molecules are a challenge. Yet, the risk - to - benefi t ratio must 
be weighed when determining such fundamental characteristics of the fi nal dosage 
form. For instance, a number of biopharmaceutical compounds are administered 
subcutaneously, but this route of parenteral administration exhibits the highest 
potential for immunogenic adverse events due to the presence of Langerhans cells 
[14] . A compound ’ s immunogenic potential is related to a host of factors, both 
TABLE 1 Factors That Determine Route of 
Administration 
Site of action 
Therapeutic indication 
Dosage 
Potency/biological activity 
Pharmacokinetic profi le 
Absorption time from tissue vs. IV 
Circulating half - life 
Distribution and elimination kinetics 
Toxicological profi le 
Immunogenic potential 
Patient population characteristics 
Disease state 
Pathophysiology 
Age 
Pharmacodynamic profi le 
Onset and duration of action 
Required clinical effect 
Formulation considerations 
Stability 
Impurity profi le 
FORMULATION ASSESSMENT 9

10 BIOTECHNOLOGY-DERIVED DRUG PRODUCT DEVELOPMENT 
patient and treatment related; however, if an alternate, potentially safer route of 
administration is available, it may be prudent to consider it. Other factors, such as 
the frequency of dosing (especially into an immune organ such as the skin) and the 
duration of treatment, can also dramatically increase the potential for immunogenic 
reactions [14] . Many of the factors that contribute to the immunogenic potential of 
biopharmaceuticals, such as impurities, degradation products, and native antigenic 
epitopes, can be mitigated through altering the physicochemical properties of the 
drug (e.g., pegylation [15, 16] , acylation [17, 18] , increased glycosylation to mask 
epitopes [19] ) or changing the characteristics of the formulation [20, 21] . In reality, 
the pharmaceutical industry has done a good job of recognizing the potential implications 
of immunogenic reactions and readily embraced technologies that can either 
mask or eliminate potential antigenic epitopes. However, additional research is 
needed to further identify and remove immunogenic epitopes. 
1.1.2.2 Pharmacokinetic Implications to Dosage Form Design 
Biological agents are generally eliminated by metabolism into di - and tripeptides, 
amino acids, and smaller components for subsequent absorption as nutrients or 
clearance by the kidney, liver, or other routes. Renal elimination of peptides and 
proteins occur primarily via three distinct mechanisms. The fi rst involves the glomerular 
fi ltration of low - molecular - weight proteins followed by reabsorption into 
endocytic vesicles in the proximal tubule and subsequent hydroysis into small peptide 
fragments and amino acids [22] . Interleukin 11 (IL - 11) [23] , IL - 2 [24] , insulin [25] , 
and growth hormone [26] have been shown to be eliminated by this method. The 
second involves hydrolysis of the compound at the brush border of the lumen and 
subsequent reabsorption of the resulting metabolites [6] . This route of elimination 
applies to small linear peptides such as angiotensin I and II, bradykinin, glucagons, 
and leutinizing hormone releasing hormone (LHRH) [6, 27, 28] . The third route of 
renal elimination involves peritubular extraction from postglomerular capillaries 
and intracellular metabolism [6] . Hepatic elimination may also play a major role in 
the metabolism of peptides and proteins; however, reticuloendothelial elimination is 
by far the primary elimination route for large macromolecular compounds [29] . 
Biopharmaceutical drug products are subject to the same principles of pharmacokinetics 
and exposure/response correlations as conventional small molecules [6] . 
However, these products are subject to numerous pitfalls due to their similarity to 
nutrients and endogenous proteins and the evolutionary mechanisms to break them 
down or prevent absorption. The types of pharmacokinetic - related problems that a 
biotechnology drug development team may encounter range from lack of specifi city 
and sensitivity of bioanalytical assays to low bioavailability and rapid drug elimination 
from the system [6] . For example, most peptides have hormone activity and 
usually short elimination half - lives which can be desirable for close regulation of 
their endogenous levels and function. On the other hand, some proteins such as 
albumin or antibodies have half - lives of several days and formulation strategies 
must be designed to account for these extended elimination times [6] . For example, 
the reported terminal half - life for SB209763, a humanized monoclonal antibody 
against respiratory syncytial virus, was reported as 22 – 50 days [30] . Furthermore, 
some peptide and protein products that persist in the bloodstream exhibit the 
potential for idiosyncratic adverse affects as well as increased immunogenic poten

tial. Therefore, the indication and formulation strategy can prove crucial design 
parameters simply based on clearance mechanisms. 
1.1.2.3 Controlled - Release Delivery Systems 
Given that the majority of biopharmaceutical products are indicated for chronic 
conditions and may require repeated administrations, products may be amenable to 
controlled - release drug delivery systems. Examples include Lupron Depot (leuprolide 
acetate), which is delivered subcutaneously in microspheres [31] , and Viadur, 
which is implanted subcutaneously [32] . Various peptide/protein controlled delivery 
systems have been reviewed recently by Degim and Celebi and include biodegradable 
and nondegradable microspheres, microcapsules, nanocapsules, injectable 
implants, diffusion - controlled hydrogels and other hydrophilic systems, microemulsions 
and multiple emulsions, and the use of iontophoresis or electroporation [33] . 
These systems offer specifi c advantages over traditional delivery mechanisms when 
the drug is highly potent and if prolonged administration greater than one week is 
required [5, 33] . However, each of these systems has its own unique processing and 
manufacturing hurdles that must be addressed on a case - by - case basis. These factors, 
coupled with the diffi culties of maintaining product stability, limit the widespread 
application of these technologies. However, the introduction of postapproval 
extended - release formulations may also provide the innovator company extended 
patent/commercial utility life and, as such, remains a viable option for postmarketing 
development. A current example of this is observed in the development of a 
long - acting release formulation of Amylin and Eli Lilly ’ s co - marketed Byetta 
product. 
1.1.3 ANALYTICAL METHOD DEVELOPMENT 
The physical and chemical characterization of any pharmaceutical product is only 
as reliable as the quality of the analytical methodologies utilized to assess it. Without 
question, the role of analytical services to the overall drug product development 
process is invaluable. Good analytical testing with proper controls could mean the 
difference between a marketable product and one that is eliminated from development. 
Analytical methodologies intended for characterization and/or assessment of 
marketed pharmaceutical products must be relevant, validatable, and transferable 
to manufacturing/quality assurance laboratories. 
1.1.3.1 Traditional and Biophysical Analytical Methodologies 
Typically, there are a handful of traditional analytical methodologies that are utilized 
to assess the physical, chemical, and microbiological attributes of small - 
molecule pharmaceutical products. While many of these testing paradigms can still 
be utilized to assess biopharmaceuticals, these molecules require additional biophysical, 
microbiological, and immunogenic characterization as well. In brief, analytical 
methodologies should evaluate the purity and bioactivity of the product and 
must also be suitable to assess potential contaminants from expression systems 
as well as different isoforms and degradation products of the active. Biophysical 
ANALYTICAL METHOD DEVELOPMENT 11

12 BIOTECHNOLOGY-DERIVED DRUG PRODUCT DEVELOPMENT 
methodologies allow for assessment of the structural elements of the product with 
respect to its activity. Such assessments include structural elements, such as the 
folding of the molecule, and also encompass potential posttranslational modifi cations 
and their impact on structure. A list of typical analytical parameters and 
methodologies utilized to assess those parameters can be found in Table 2 . 
The impact of a molecule ’ s biophysical characteristics on its clinical effi cacy 
should be readily quantifi able. With respect to rational drug design, it is also 
extremely important to minimize external factors that may infl uence the formation 
of any adverse response. One such factor is the presence of degradation products 
and drug - related impurities that may be responsible for an immune response. One 
such industrial example is granulocyte - macrophage colony - stimulating factor [GM - 
CSF, or Leukine (sargramostim), by Berlex Co.], which is produced as a recombinant 
protein synthesized and purifi ed from a yeast culture, Saccharomyces cerevisiae . As 
expected, the expression system has an impact on the fi nal product: sargramostim, 
manufactured from S. cerevisiae , yields an O - glycosylated protein, while molgramostim 
(Leucomax), synthesized using an E. coli expression system, is nonglycosylated 
[34] . The E. coli – derived product exhibited a higher incidence of adverse reactions 
in clinical trials and never made it to the market. With respect to the drug product, 
the immunogenic reactions included [34, 35] : 
TABLE 2 Analytical Methodologies and Their Utility for API and Drug Product 
Characterization 
Parameter 
Assessed Methodologies Utility 
Appearance Visual appearance, 
colorimetric assays, 
turbidity 
Simple determination of physical 
stability, i.e., are there particles in 
solution, is the solution the correct 
color/turbidity? Is the container 
closure system seemingly intact? 
Purity, 
degradation 
products and 
related 
substances 
GPC/SEC - HPLC, RP - 
HPLC, gel electrophoresis, 
immunoassays, IEF, MS, 
CD, CE 
Gives a general idea of the relative 
purity of the API and the drug 
product. Are there impurities 
related to the expression system? 
Are there alternate API isoforms 
present? Can degradation products 
be distinguished from the active 
component(s)? 
Molecular 
weight 
determination 
GPC/SEC - HPLC, gel 
electrophoresis, multiangle 
laser light scattering 
(MALLS), laser diffraction 
Is the product a single molecular 
weight or polydisperse? Is the 
molecular weight dependent on 
posttranslational modifi cations? 
Potency Biological activity (direct or 
indirect) 
Does the compound have reproducible 
in vitro activity and can this be 
correlated to in vivo? 
pH Potentiometric assays Is the product pH labile or do pH 
changes affect potency is such ways 
that are not evident in other assays, 
i.e., minimal degradation and/or 
unfolding?

ANALYTICAL METHOD DEVELOPMENT 13 
Parameter 
Assessed Methodologies Utility 
Primary 
structural 
elements 
Protein sequencing, N - term 
degradation (Edman 
degradation), peptide 
mapping, amino acid 
composition, 2D - NMR 
Verifi es primary amino acid sequence 
and gives preliminary insight into 
activity. 
Secondary 
structural 
elements 
CD, 2D - NMR, in silico 
modeling from AA 
sequence 
Secondary structural elements result 
from the primary sequence and help 
defi ne the overall conformation 
(3D folding) of the compound. 
Tertiary 
structural 
elements 
Disulfi de content/position, 
CD 
Determines correct folding and overall 
integrity of the 3D product. 
Qualitative determination for 
denaturation potential. Also 
correlates to immunogenic potential. 
Agglomeration/ 
aggregation 
Subvisual and visual Particle 
size analysis, 
immunogenicity 
Indicator of physical instability. Also 
gives an indication of immunogenic 
potential. 
Carbohydrate 
analysis 
RP - HPLC, gel 
electrophoresis, AE - 
HPLC, CE, MALDI - MS, 
ES - MS, enzyme arrays 
Ensures proper posttranslational 
modifi cations and carbohydrate 
content. 
Water content 
(lyophilized 
products) 
Karl Fischer, TGA, NIR Indicator of hydrolytic potential and 
process effi ciency. 
Immunogenic 
potential 
Surface plasmon resonance, 
ELISA, 
immunoprecipitation 
Methodologies generally only give 
positive/negative indicators of 
immunogenic potential. In vitro 
methodologies do not always 
correlate to in vivo. 
Sterility Membrane fi ltration Indicator of microbial contaminants 
from manufacturing operations. 
Bacterial 
endotoxins 
Limulus amebocyte lysate 
(LAL) 
Gives an idea of processing 
contaminants and potentially host 
organism contaminants. 
Container 
closure 
integrity 
Dye immersion, NIR, 
microbial ingress/sterility 
Demonstrates viability of container 
closure system over the life of the 
product. 
Abbreviations : gel permeation chromatography (GPC), size exclusion chromatography (SEC), high - 
performance, or high - pressure, liquid chromatography (HPLC), reverse phase (RP), isoelectric focusing 
(IEF), mass spectrometry (MS), circular dichroism (CD), capillary electrophoresis (CE), nuclear magnetic 
resonance (NMR), anion exchange (AE), matrix - assisted laser desorption ionization (MALDI), 
electrospray ionization (ES), thermogravimetric analysis (TGA), near infrared (NIR), enzyme - linked 
immunosorbent assay (ELISA) 
TABLE 2 Continued

14 BIOTECHNOLOGY-DERIVED DRUG PRODUCT DEVELOPMENT 
1. Formation of antibodies which bind and neutralize the GM - CSF 
2. Formation of antibodies which bind but do not affect the effi cacy of 
GM - CSF 
3. Antibody formation against proteins not related to GM - CSF, but to proteins 
from the expression system ( E. coli ) 
4. Antibodies formed against both product - and non - product - related proteins 
5. No antibody formation 
This example clearly illustrates not only the range of clinical manifestations with 
respect to antibody formation to drug therapy but also how the choice of an expression 
system can affect the fi nal product. In this example, the expression system was 
responsible for the adverse events reported. This fi nding is certainly clinically relevant 
considering the homologous product, sargramostim, has been on the U.S. 
market for quite some time. 
The above example also gives an indication of the relative importance of carbohydrate 
analysis. Without question, protein glycosylation is the most complex of all 
posttranslational modifi cations made in eukaryotic cells, the importance of which 
cannot be underestimated. For many compounds, glycosylation can readily affect 
protein solubility (as infl uenced by folding), protease resistance, immunogenicity, 
and pharmacokinetic/pharmacodynamic profi les (i.e., clearance and effi cacy) [36] . 
Typical analytical methodologies used to assess carbohydrate content are also listed 
in Table 2 . 
1.1.3.2 Stability - Indicating Methodologies 
Analytical methodologies that are specifi c to the major analyte that are also capable 
of separating and quantifying potential degradation products and impurities, while 
simultaneously maintaining specifi city and accuracy, are deemed stability indicating. 
Traditional stability - indicating high - performance liquid chromatography (HPLC) 
methodologies for small molecules are developed and validated with relative ease. 
Typically, the stability - indicating nature of an analytical method can be demonstrated 
by subjecting the product to forced degradation in the presence of heat, acid, 
alkali, light, or peroxide [37] . If degradation products are suffi ciently well resolved 
from the active while maintaining specifi city and accuracy, the method is suitable. 
In contrast to small molecules, there is no one “ gold standard ” analytical methodology 
that can be utilized to determine the potential degradation products and impurities 
in the milieu that may constitute a biopharmaceutical drug product. Furthermore, 
a one - dimensional structure assessment (e.g., in terms of an absorption spectrum) 
does not give any indication of the overall activity of the product, as is the case with 
traditional small molecules. Thus, the stability assessment of biopharmaceuticals will 
typically comprise a multitude of methodologies that when taken together give an 
indication of the stability of the product. The overall goal is to assess the structural 
elements of the compound as well as attempt to determine the relative quantities 
of potential degradation products, as well as product isoforms and impurities, that 
are inherent to the expression systems utilized for API manufacture. However, it is 
still advised that bioactivity determinations are made at appropriate intervals 
throughout the stability program, as discussed below. Furthermore, any biopharma

ceutical stability program should also minimally include an evaluation of the in vitro 
immunogenicity profi le of the product with respect to time, temperature, and other 
potential degradative conditions. 
1.1.3.3 Method Validation and Transfer 
Analytical method validation is the process by which scientists prove that the analytical 
method is suitable for its intended use. Guidances available on validation 
procedures for some traditional analytical methodologies [38] can be adapted to 
nontraditional methodologies. The United States Pharmacopeia (USP) and National 
Formulary (NF) do provide some guidance on designing and assessing biological 
assays [39] , as does the U.S. FDA [40] . Essentially, validation determines the acceptable 
working ranges of a method and the limitations of that method. At a minimum 
the robustness, precision, and accuracy of quantitative methodologies should be 
determined during support of API iteration and refi nement, while at the very least 
the robustness of qualitative methodologies should be assessed. Of particular importance 
for successful analytical method validation is ensuring that the proper standards 
and system suitability compounds have been chosen and are representative 
or analogous to the compound to be analyzed and traceable to a known origin 
standard, such as the National Institute of Standards and Technology (NIST) or 
USP/NF. If a reference standard from an “ offi cial ” source is not available, in - house 
standards may be used provided they are of the highest purity that can be reasonably 
obtained and are thoroughly characterized to ensure its identity, strength, 
quality, purity, and potency. 
Methods developed and validated during the product development phase are 
routinely transferred to quality control or contract laboratories to facilitate release 
and in - process testing of production batches. Ensuring that method transfer is executed 
properly, with well - defi ned and reproducible system suitability and acceptance 
criteria, is the responsibility of both laboratories. Experiments should consist 
of all those parameters assessed during method validation and should include an 
evaluation of laboratory - to - laboratory variation. This information will give an idea 
of the reliability of the methodology and equipment used under the rigors of large - 
scale manufacturing. 
1.1.4 FORMULATION DEVELOPMENT 
The previous sections have highlighted some of the limitations and diffi culties in 
developing biotechnology - derived pharmaceuticals. Although there are major technological 
limitations in working with these products, their synthesis and manufacturing 
are signifi cantly more reproducible compared to naturally derived biologics. 
Determining the most appropriate route of administration and subsequent formulation 
is dependent on a number of factors, including the product ’ s indication, duration 
of action, pharmacokinetic parameters, stability profi le, and toxicity. As 
mentioned previously, biopharmaceuticals are typically delivered parenterally, and 
thus we will focus on those studies required to successfully develop a parenteral 
formulation of a biopharmaceutical agent. The goal of formulation development is 
to design a dosage form that ensures the safety and effi cacy of the product through- 
FORMULATION DEVELOPMENT 15

16 BIOTECHNOLOGY-DERIVED DRUG PRODUCT DEVELOPMENT 
out its shelf life while simultaneously addressing the clinical needs of both the 
patient and caregivers to ensure compliance. Formulation development is truly a 
balancing act, attempting to emphasize the benefi ts of the therapy and patient compliance 
while maximizing drug effi cacy and minimizing toxicity. As such, a number 
of studies are required to properly design and develop a formulation, many of which 
are discussed below. 
1.1.4.1 Processing Materials and Equipment 
An important factor in the quality and reproducibility of any formulation development 
activity is the materials utilized for formulating and processing studies. In 
addition, the choice of container closure systems for the API and the formulation 
needs to be considered carefully to provide maximum product protection and 
optimal stability. Variability between small - and larger scale development stages 
may also be signifi cant depending on the API and materials involved during process 
scale - up. It is important to conduct process development studies utilizing equipment 
representative of what will be used for large - scale production, if possible. Implementing 
this design approach will enable at least some limited dimensional analysis, 
allowing for early identifi cation of critical design parameters, thereby facilitating 
scale - up or permitting earlier attrition decisions and cost savings. Regardless, it is 
important to consider the chemical composition and material properties of every 
manufacturing component that may contact the drug product. For instance, processing 
vessels may be made of glass, glass - lined steel, or bare steel, while stir paddles 
used to ensure homogeneity made be manufactured of a number of different materials. 
In short, any manufacturing unit that could potentially come into intimate 
contact with either the formulation or the API should be demonstrated to be compatible 
with the product, including sampling instruments, sample vials, analytical and 
processing tubing, and so forth. Material incompatibility could result in something 
as simple as unexplained analytical variability due to a loss of drug through adsorptive 
mechanisms to something as serious as a loss of bioactivity or an increase in 
immunogenic potential. Therefore, equipment design and materials would ideally 
be consistent from formulation development through to scale - up and process validation; 
however, this may not be readily feasible. As such, determining the chemical 
and physical compatibility of each piece of processing equipment with the API is 
critical to maintaining the physical and chemical attributes of the product. Furthermore, 
such studies help eliminate potential sources of experimental variability and 
give a better indicator as to the relative technological hurdles to successful product 
development. 
Material compatibility protocols must be clearly defi ned and require that analytical 
methodologies be suitable for their intended use. Typically, product purity 
methods and cleaning methodologies utilized to determine residual contaminating 
product on processing equipment are used for compatibility studies as they are suf- 
fi ciently sensitive and rugged to accurately determine product content in the presence 
of a multitude of potential confounding factors. This is particularly important 
when assessing potential metal, glass, and tubing compatibilities. Compatibility is a 
function not only of the product ’ s intimate contact with surrounding materials but 
also of the contact time and surface area with these equipment. As such, protocols 
should be designed to incorporate expected real - world conditions the product will 

see when in contact with the material. For instance, temperature, light, and mechanical 
stimulation should mimic usage conditions, although study duration should 
include time intervals that surpass expectations to estimate a potential worst case. 
These factors should all be considered when examining potential process - related 
stability. 
1.1.4.2 Container Closure Systems 
The ICH guideline for pharmaceutical development outlines requirements for container 
closure systems for drugs and biologics [41] . The concept paper prepared for 
this guidance specifi cally states that “ the choice of materials for primary packaging 
should be justifi ed. The discussion should describe studies performed to demonstrate 
the integrity of the container and closure. A possible interaction between 
product and container or label should be considered ” [42] . In essence, this indicates 
that the container closure system should maintain the integrity of the formulation 
throughout the shelf life of the product. In order to maintain integrity, the container 
closure system should be chosen to afford protection from degradation induced by 
external sources, such as light and oxygen. In addition to the primary container, the 
stability of the product should also be examined in the presence of IV administration 
components if the product could be exposed to these conditions (see Section 
1.1.5.6 ). Understanding the potential impact of product - to - container interactions is 
integral to maintaining stability and ensuring a uniform dosage. For example, adsorption 
of insulin and some small molecules has been demonstrated to readily occur 
in polyvinyl chloride (PVC) bags and tubing when these drugs were present as 
additives in intravenous (IV) admixtures [43] . 
In addition to their use in large - volume parenterals and IV sets, thermoplastic 
polymers have also recently found utility as packaging materials for ophthalmic 
solutions and some small - volume parenterals [43] . However, there are many 
potential issues with using these polymers as primary packaging components that 
are not major concerns with traditional glass container closure systems, including 
[44] : 
1. Permeation of vapors and other molecules in either direction through the wall 
of the plastic container 
2. Leaching of constituents from the plastic into the product 
3. Sorption (absorption and/or adsorption) or drug molecules or ions on the 
plastic material 
These concerns largely preclude the utility of thermoplastic polymers as the primary 
choice of container closure system for protein and peptide therapeutics, although 
the formulation scientist should be aware of the potential advantages of these 
systems, such as the ease of manufacturability and their cost. These systems are also 
fi nding greater utility in intranasal and pulmonary delivery systems. 
Parenterally formulated biopharmaceuticals are typically packaged in glass containers 
with rubber/synthetic elastomeric closures. Pharmaceutical glass is composed 
primarily of silicon dioxide tetrahedron which is modifi ed with oxides such 
as sodium, potassium, calcium, magnesium, aluminum, boron, and iron [45] . The USP 
classifi es glass formulations as follows: 
FORMULATION DEVELOPMENT 17

18 BIOTECHNOLOGY-DERIVED DRUG PRODUCT DEVELOPMENT 
Type I, a borosilicate glass 
Type II, a soda – lime treated glass 
Type III, a soda – lime glass 
NP, a soda – lime glass not suitable for containers for parenterals 
The tendency of peptides to adsorb onto glass surfaces is well known and a major 
concern in the pharmaceutical industry. This is especially important when the dose 
of the active ingredient is relatively small and a signifi cant amount of drug is 
adsorbed to these surfaces. In addition, the leaching of atoms or elements in the 
glass ’ s silicate network into solution is also a potential issue. This is especially important 
for terminally heat sterilized products where oxide additives included in the 
silicate network are relatively free to migrate/leach, resulting in increased solution 
pH, reaction catalysis, and so on [45] . As such, only type 1 treated glass is traditionally 
used for parenterally administered formulations, where these alkaline - rich 
phases in the glass have been eliminated, thus decreasing the potential for container 
closure system interactions. Additional approaches, including surface treatment with 
silicone (siliconization), have also been developed to minimize the interaction of 
biotechnology products with free silanols (Si – OH) [46] . 
Elastomeric closures are typically used for syringe and vial plungers and closures. 
For vials, elastomers provide a soft and elastic material that can permit the entry of 
a hypodermic needle without loss of the integrity [45] . For syringes, the closures not 
only provide a permeation barrier but also allow for a soft gliding surface facilitating 
plunger movement and drug delivery. Elastomeric polymers, however, are very 
complex materials composed of multiple ingredients in addition to the basic polymers, 
such as vulcanizing agents, accelerators, activators, antioxidants, fi llers, lubricating 
agents, and pigments [45] . As leaching of these components into solution 
is a potential issue, the compatibility of the drug formulation with the closures 
must be studied early during the formulation development process. The choice and 
type of elastomeric closure depends on the pH and buffer, if any preservatives 
are present, the sterilization method, moisture vapor/gas protection, and active 
compatibility [47] . In addition, the problem of the additives in rubber leaching 
into the product can be reduced by the coating with specifi c polymers such as 
Tefl on [48] . 
Container closure systems required for implantable devices are further restricted 
by the fact that they are required to be compatible with the formulation over the 
intended shelf life and therapeutic application time as well as being biocompatible. 
This means that the system not only must afford protection to and contain the formulation 
but also cannot cause any potential adverse effects, such as allergy. Typically, 
implantable systems are composed of biocompatible metals, such as titanium 
or polymers such as polyethylene glycol or polylactic - co - glycolic acid. 
1.1.4.3 Sterility Assurance 
Maintaining the sterility of biopharmaceutical products is especially important due 
to the relative potency and their innate potential for immunogenic reactions. Further, 
the biochemical nature of these compounds enables them to serve as potential 
nutrients for invading organisms. Methods for sterilizing small molecules include 

heat terminal sterilization, terminal fi ltration coupled with aseptic processing techniques, 
ultraviolet (UV) and gamma irradiation, ethylene oxide exposure (for containers 
and packaging only), and electron beam irradiation. While terminal heat 
sterilization is by far the most common sterilization technique, it normally cannot 
readily be utilized for peptide or protein formulations due to the potential effects 
of heat and pressure on the compound ’ s structure [48] . Furthermore, irradiation can 
affect protein stability by cross - linking the sulfur - containing and aromatic residues, 
resulting in protein aggregation [49] . 
To overcome these issues, sterile fi ltration coupled with aseptic processing and 
fi lling is the preferred manufacturing procedure for biopharmaceuticals. Garfi nkle 
et al. refer to aseptic processing as “ those operations performed between the sterilization 
of an object or preparation and the fi nal sealing of its package. These operations 
are, by defi nition, carried out in the complete absence of microorganisms ” [50] . 
This highlights the importance of manufacturing controls and bioburden monitoring 
during aseptic processes. Newer technologies such as isolator technology have been 
developed to reduce human intervention, thereby increasing the sterility assurance. 
These technologies have the added benefi t of facilitating aseptic processing without 
construction of large processing areas, sterile suites, or gowning areas [50] . 
Even the most robust monitoring programs do not ensure the sterility of the fi nal 
formulation. As such, aseptically processed formulations are traditionally fi ltered 
through a retentive fi nal fi lter, which ensures sterility. Coupled with proper component 
sterilization, traditionally by autoclaving, these processes ensure product sterility. 
However, fi ltration is a complex unit operation that can adversely affect the drug 
product through increased pressure, shear, or material incompatibility. Therefore, 
fi ltration compatibility must be assessed thoroughly to demonstrate both product 
compatibility, and suffi cient contaminant retention [51] . Parenteral Drug Association 
(PDA) technical report 26 provides a thorough systematic approach to selecting 
and validating the most appropriate fi lter for a sterilizing fi ltration application 
[51] . 
1.1.4.4 Excipient Selection 
Pharmaceutical products are typically formulated to contain selected nonactive 
ingredients (excipients) whose function is to promote product stability and enable 
delivery of the active pharmaceutical ingredient(s) to the target site. These substances 
include but are not limited to solubilizers, antioxidants, chelating agents, 
buffers, tonicity contributors, antibacterial agents, antifungal agents, hydrolysis 
inhibitors, bulking agents, and antifoaming agents [45] . The ICH states that “ the 
excipients chosen, their concentration, and the characteristics that can infl uence the 
drug product performance (e.g. stability, bioavailability) or manufacturability should 
be discussed relative to the respective function of each excipient ” [42] . Excipients 
must be nontoxic and compatible with the formulation while remaining stable 
throughout the life of the product. Excipients require thorough evaluation and 
optimization studies for compatibility with the other formulation constituents as 
well as the container/closure system [52] . Furthermore, excipient purity may be 
required to be greater than that listed in the pharmacopeial monograph if a specifi c 
impurity is implicated in potential degradation reactions (e.g., presence of trace 
metals) [48] . 
FORMULATION DEVELOPMENT 19

20 BIOTECHNOLOGY-DERIVED DRUG PRODUCT DEVELOPMENT 
One of the critical factors in excipient selection and concentration is the effect 
on preferential hydration of the biopharmaceutical product [53, 54] . Preferential 
hydration refers to the hydration layers on the outer surface of the protein and can 
be utilized to thermodynamically explain both stability enhancement and denaturation. 
Typical excipients used in protein formulations include albumin, amino acids, 
carbohydrates, chelating and reducing agents, cyclodextrins, polyhydric alcohols, 
polyethylene glycol, salts, and surfactants. Several of these excipients increase the 
preferential hydration of the protein and thus enhance its stability. Cosolvents need 
to be added in a concentration that will ensure their exclusion from the protein 
surface and enhance stability [54] . A more comprehensive review of excipients utilized 
for biopharmaceutical drug products is available elsewhere [48] . 
Buffer Selection In addition to maintaining solution pH, buffers serve a multitude 
of functions in pharmaceutical formulations, such as contributing toward overall 
isotonicity, preferential hydration of proteins and peptides, and serving as bulking 
agents in lyophilized formulations. The buffer system chosen is especially important 
for peptide and proteins that have sensitive secondary, tertiary, and quaternary 
structures, as the overall mechanisms contributing to conformational stabilization 
are extremely complex [48] . Furthermore, a protein ’ s propensity for deamidation at 
a particular pH can be signifi cant, as illustrated by Wakankar and Borchardt [55] . 
This study illustrated stability concerns with peptides and proteins at physiological 
pH in terms of asparagine (Asn) deamidation and aspartate (Asp) isomerization, 
which can be a major issue with respect to circulating half - life and potential in vivo 
degradation. This study and others also provide insight into predicting potential 
degradative mechanisms based on primary and secondary structural elements allowing 
for formulation design with these pathways in mind. 
Selecting the appropriate buffer primarily depends on the desired pH range and 
buffer capacity required for the individual formulation; however, other factors, 
including concentration, effective range, chemical compatibility, and isotonicity 
contribution, should be considered [56] . Some acceptable buffers include phosphate 
(pH 6.2 – 8.2), acetate (pH 3.8 – 5.8), citrate (pH 2.1 – 6.2, p K 3.15, 4.8, and 6.4), 
succinate (pH 3.2 – 6.6, p K 4.2 and 5.6), histidine (p K 1.8, 6.0, and 9.0), glycine 
(pK 2.35 and 9.8), arginine (p K 2.18 and 9.1), triethanolamine (pH 7.0 – 9.0), tris - 
hydroxymethylaminomethane (THAM, p K 8.1), and maleate buffer [48] . Additionally, 
excipients utilized solely for tonicity adjustment, such as sodium chloride and 
glycerin, may not only differ in ionic strength but also could afford some buffering 
effects that should be considered [52] . 
Preservatives In addition to those processing controls mentioned above (Section 
3.1.4.3 ), the sterility of a product may be maintained through the addition of antimicrobial 
preservatives. Preservation against microbial growth is an important 
aspect of multidose parenteral preparations as well as other formulations that 
require preservatives to minimize the risk of patient infection upon administration, 
such as infusion products [52] . Aqueous liquid products are prone to microbial 
contamination because water in combination with excipients derived from natural 
sources (e.g., polypeptides, carbohydrates) and proteinaceous active ingredients 
may serve as excellent media for the growth [57] . The major criteria for the selection 
of an appropriate preservative include effi ciency against a wide spectrum of micro

organisms, stability (shelf life), toxicity, sensitizing effects, and compatibility with 
other ingredients in the dosage form [57] . Typical antimicrobial preservatives include 
m - cresol, phenol, parabens, thimerosal, sorbic acid, potassium sorbate, benzoic acid, 
chlorocresol, and benzalkonium chloride. Cationic agents such as benzalkonium 
chloride are typically not utilized for peptide and protein formulations because they 
may be inactivated by other formulation components and their respective charges 
may induce conformational changes and lead to physical instability of the API. 
Further, excipients intended for other applications, such as chelating agents, may 
exhibit some antimicrobial activity. For instance, the chelating agent ethylenediaminetetraacetic 
acid (EDTA) may exhibit antimicrobial activity, as calcium is required 
for bacterial growth. 
Identifying an optimal antimicrobial preservative is based largely on the effectiveness 
of that preservative at the concentration chosen. In short, it is not enough to 
assess the compatibility of the preservative of choice with the API and formulation 
and processing components. There also needs to be a determination of whether the 
preservative concentration is suffi cient to kill certain standard test organisms. The 
USP presents standard protocols for assessing the relative effi cacy of a preservative 
in a formulation using the antimicrobial effectiveness test (AET) [58] . Briefl y, by 
comparing the relative kill effi ciency of the formulation containing varying concentrations 
of the preservative, the formulator can determine the minimal concentration 
required for preservative effi cacy and design the formulation accordingly. 
1.1.5 DRUG PRODUCT STABILITY 
1.1.5.1 Defi ning Drug Product Storage Conditions 
From a regulatory standpoint, the primary objective of formulation development is 
to enable the delivery of a safe and effi cacious drug product to treat and/or mitigate 
a disease state throughout its proposed shelf life. The effi cacy and in many cases the 
safety of a product are directly related to the stability of the API, both neat and in 
the proposed formulation under processing, storage, and shipping conditions as well 
as during administration. As such, the concept of drug stability for biotechnology - 
derived products does not change substantially from that of small molecules, 
although the level of complexity increases commensurate with the increased complexity 
of the APIs in question and the formulation systems utilized for their 
delivery. 
Stability study conditions for biotechnology - derived APIs and their respective 
drug products are not substantially different from those studies conducted for small 
molecules. Temperature and humidity conditions under which to conduct said 
studies are outlined in ICH Q1A(R2), which incorporates ICH Q1F, stability study 
conditions for zones III and IV climactic conditions [59] . Additional guidance specifi 
c to conducting stability studies on biopharmaceutical drug products is given in 
ICH Q5C [1] . However, the intention of ICH Q5C is not to outline alternate temperature 
and humidity conditions to conduct primary stability studies; rather it 
provides guidance with respect to the fact that the recommended storage conditions 
and expiration dating for biopharmaceutical products will be different from product 
to product and provides the necessary fl exibility in letting the applicant determine 
DRUG PRODUCT STABILITY 21

22 BIOTECHNOLOGY-DERIVED DRUG PRODUCT DEVELOPMENT 
the proper storage conditions for their respective product. Furthermore, this document 
provides general guidance in directing applicants in the types of analytical 
methodologies that may be used and direction on how to properly assess the stability 
of these complex molecules [1] : 
Assays for biological activity, where applicable, should be part of the pivotal stability 
studies. Appropriate physicochemical, biochemical and immunochemical methods for 
the analysis of the molecular entity and the quantitative detection of degradation 
products should also be part of the stability program whenever purity and molecular 
characteristics of the product permit use of these methodologies. 
One recent approach to aid in defi ning the design space for protein and peptide 
therapeutics has been to create empirical phase diagrams indicating the relative 
stability of compounds based on altering conditions and assessing conformational 
changes via a compilation of analytical techniques (Figure 1 ) [60 – 62] . These empirical 
phase diagrams can be generated based on pH, temperature, salt concentration, 
and so on, and, although seemingly laborious at fi rst glance, could provide invaluable 
information in defi ning the extremes to which a compound may be subjected without 
altering its conformation. For instance, if an empirical phase diagram determines 
the safe temperature range for a compound is up to 35 ° C and an excursion occurs 
to 33 ° C, this information would give the stability scientist a guideline as to the 
appropriate course of action. Under the traditional testing paradigm of ICH Q1A, 
where stability testing is limited to 25, 30, and 40 ° C, one may not know the compound 
’ s upper transition temperature to induce conformational changes. If the 
information is not already available, then additional excursion studies may need to 
be conducted to assimilate this information and take the appropriate course of 
action. 
1.1.5.2 Mechanisms of Protein and Peptide Degradation 
The inherent heterogeneity of peptide and protein drug substances results in their 
relative sensitivity to processing, storage, and handling conditions as well as a mul- 
FIGURE 1 Empirical phase diagram for ricin toxin A - chain generated using CD molar 
ellipticity at 208 nm, ANS fl uorescence, and intrinsic Trp fl uorescence intensity data. Labels 
indicate the state of the protein within the same region of color based on evaluation of a 
compilation of data sets. (Reproduced with permission from ref. 62 .) 
20 
40 
60 
80
T 
4 5 6 7 8 9 
pH

titude of other factors. Most importantly, this heterogeneity results in a whole host 
of potential degradative mechanisms, some of which are compiled in Table 3 and 
include chemical instability pathways such as oxidation, hydrolysis of side chains 
and potentially the peptide backbone, and deamidation of Asn and Gln side chains. 
Also, physical instability manifesting in the form of protein unfolding, formation of 
intermediate structures, aggregation, and adsorption to the surfaces of containers 
and other equipment can be a major technical hurdle in developing any biopharmaceutical 
and may or may not be related to chemical instability [63] . Further 
complicating matters is that instability can potentially manifest in various ways and 
may or may not be detectable by any one method. Taken together, however, the 
compilation of methodologies utilized for stability assessment should give a good 
approximation as to the degradative mechanisms of the compound in its respective 
formulation. Further, bioactivity and immunogenicity assays should play integral 
roles in assessing the relative stability of any biopharmaceutical compound. Briefl y 
stated, the chemical and physical stability of products is extraordinarily diffi cult to 
assess and will not be belabored here as good reviews on this topic are readily available 
in the literature [63, 64] . 
1.1.5.3 Photostability 
In certain cases, exposure of pharmaceutical compounds to UV and visible light 
could result in electronic excitation, termed vertical transition, that could ultimately 
result in light - induced degradation. The ICH guideline Q1B [65] is a reference 
on how to conduct photostability stress testing for pharmaceutical compounds. 
In brief, compounds are exposed to an overall illumination of not less than 1.2 
million lux hours and an integrated near - UV energy of not less than 200 Wh/m 2 
[65] . These requirements are in addition to normal stability stress testing and 
require the additional caveat that analytical methodologies are suitable to also 
detect photolytic degradation products, as discussed above. A comprehensive discussion 
of small - molecule photolytic degradative mechanisms is available for 
further review [66] . 
TABLE 3 Potential Degradative Mechanisms of Peptides and Proteins 
Degradative Mechanism Site of Occurrence 
Chemical 
degradative 
mechanisms 
Oxidation Intrachain disulfi de linkages Met, Trp, Tyr 
Peptide bond hydrolysis AA backbone 
N - to - O migration Ser and Thr 
. - to . - Carboxy migration Asp and Asn 
Deamidation Asn and Gln 
Acylation . - Amino and . - amino group 
Esterifi cation/carboxylation Glu, Asp, and C-term 
Physical 
degradative 
mechanisms 
Unfolding Partial unfolding of tertiary structure 
Aggregation Aggregation of subunits could result in 
precipitation 
Adsorption Adsorption to processing equipment and 
container closure systems 
Source : Modifi ed from Crommelin et al. [5] . 
DRUG PRODUCT STABILITY 23

24 BIOTECHNOLOGY-DERIVED DRUG PRODUCT DEVELOPMENT 
1.1.5.4 Mechanical Stress 
Regulatory guidance on appropriate methods to evaluate the effect of shear stress 
and process - handling stability studies is not available. However, these studies are 
integral in determining the relative stability of the product with respect to mechanical 
stresses introduced during development and manufacturing. Although not typically 
recognized as a major degradative pathway for most small - molecule dosage 
forms, the introduction of mechanical stress is recognized as a major challenge in 
the formulation of semisolids and can potentially induce physical instability of biopharmaceuticals, 
although the extent of this effect is currently unknown. For example, 
processing shear may infl uence the protein ’ s outer hydration shell, altering the stabilizing 
energy provided from preferential hydration and resulting in the exposure 
of internal, nonpolar residues. This may facilitate aggregation if enough shear force 
is provided. Alternately, the shear energy required to force unfolding has been 
studied but has not been related to the fl uid dynamic shear experienced during 
processing. Therefore, stress studies should include meticulous controls in the form 
of temperature, light and humidity, and fl uid dynamic shear as a function of time. 
Data from these studies could be incorporated into empirical phase diagrams, and/ 
or response surfaces, to help further defi ne the design space for the active and fi nished 
drug product. Understanding the effects of stress introduced during the manufacturing 
processing of biopharmaceutical products could facilitate the selection of 
appropriate PAT tools and QbD incorporation in the development of these products. 
Clearly, there is a considerable need for research in this area, and until the 
extent of the possible effects are understood, this lack of knowledge poses an 
unknown risk and prevents adequate risk assessment for biopharmaceutical development 
activities consistent with ICH Q9. 
1.1.5.5 Freeze – Thaw Considerations and Cryopreservation 
The rapid or continuous freezing and thawing of protein products could contribute 
signifi cantly to instability of the API. Such studies are typically designed to assess 
the implications of potential transport and handling conditions. These conditions 
include not only the manufacturing processing, storage, and shipment to warehouses 
and pharmacies but also subsequent pharmacy storage and patient handling [52] . 
Unpredictable and somewhat modest temperature fl uctuations could easily induce 
degradation or conformational changes that may reduce bioactivity or expose antigenic 
epitopes [5] . These effects could also be a result of altered preferential hydration 
at the surface of the peptide or protein through salting - out effects induced by 
rapid freezing, which could easily denature the product [67] . 
1.1.5.6 Use Studies 
Stability of biopharmaceutical compounds should also be determined under conditions 
that mimic their normal usage. For instance, the stability of reconstituted 
lyophilized products should be assessed with respect to time and temperature and, 
if applicable, light and mechanical stimuli. Likewise, the stability of a compound 
included in implantable devices and controlled - release microsphere formulations 
should be determined over the course of its required use, under conditions which 
mimic the heat, moisture, light, and enzymatic physiological conditions to which it 

will be implanted. Such studies should also determine the release profi le of the 
compound over these specifi ed conditions. 
Drug products intended for IV administration are generally dosed as an initial 
bolus followed by a slow infusion. Consequently, admixture studies of the compound 
in potential IV fl uids, such as 0.9% (w/v) saline, 5% (w/v) dextrose, and Ringer ’ s 
solution, should also be assessed to determine the relative stability of the compound 
in this new environment. These studies are critical as the formulation dynamic that 
protected and stabilized the compound has now been altered dramatically with 
dilution. This environmental change could potentially impact the preferential hydration 
of the compound as well as directly induce conformational changes based on 
the diluent chosen and the compound ’ s potential degradative mechanism(s). Additional 
contributing factors to instability in admixture solutions could be due to 
changes in pH, mechanical mixing of the compound in the IV bag, adsorption of 
the compound to the bag itself (which is typically polymeric), or IV sets used for 
administration, as well as an increased potential for oxidative degradation. The suitability 
of analytical methodologies should also be determined in the presence of 
these additional analytes. 
1.1.5.7 Container Closure Integrity and Microbiological Assessment 
Ensuring that parenteral pharmaceuticals maintain their sterility over the course of 
their shelf life is an integral part of any stability assessment [68] . Parenteral dosage 
forms must be free from microbiological contamination, bacterial endotoxins, and 
foreign particulate matter. Selection of the adequate sterile manufacturing process 
has been briefl y discussed above. Determining the microbiological integrity of the 
product over its shelf life also gives an indication of the relative quality of the container 
closure system chosen for the formulation. Compendial sterility and endotoxin 
testing are often used for this purpose; however, sampling is dependent on a 
statistical evaluation of the batch size, unit fi ll volume, and method of product sterilization 
[68] . Additionally, since these tests are destructive, it would be impossible 
to test an entire stability batch to ensure viability of a container closure system. 
Other nondestructive tests have been developed to determine the integrity of a 
container ’ s closure system [69] . These tests could also serve as a surrogate indicator 
of product manufacturing quality over time. 
1.1.5.8 Data Interpretation and Assessment 
Interpretation of primary stability data for determining expiration dating and 
primary storage conditions has been outlined by ICH Q1E [70] . This guidance document 
delineates broad methodologies for interpreting primary and accelerated stability 
data and extrapolation of said data for determining expiry dating. Of course, 
expiry dating cannot be made without reference to specifi cations for those primary 
stability - indicating parameters assessed, which is discussed below. Traditionally, stability 
assessments performed during preformulation will give an indication of the 
potential storage conditions as well as allow for extrapolation of accelerated stability 
studies to kinetic degradation rates. Typically this is done through Arrhenius 
manipulations. However, as one would expect, these analyses are not readily 
useful for biopharmaceutical products, as there is rarely a linear correlation between 
QUALITY BY DESIGN AND SCALE-UP 25

26 BIOTECHNOLOGY-DERIVED DRUG PRODUCT DEVELOPMENT 
temperature and the compound ’ s degradative rate. This is primarily due to the 
complex and often competing degradative mechanisms as well as the potential for 
so - called molten globule intermediate phases. In spite of these limitations, ICH Q5C 
does provide relevant guidance in illustrating the fl exibility required for determining 
storage conditions, as these products usually require a very narrow temperature 
condition to maintain optimal stability. Further guidances may be needed to enhance 
uniformity in testing methodology and enable the utilization of validated PAT 
methodologies. 
1.1.6 QUALITY BY DESIGN AND SCALE - UP 
1.1.6.1 Unit Operations 
Unit operations are defi ned as the individual basic steps in a process that when 
linked together defi ne the process train and result in the fi nal product. In practical 
terms, a unit operation is often defi ned as an individual step that is carried out on 
one piece of equipment. Typical biopharmaceutical API unit operations may include 
fermentation or bioreactor processes, cell separation through centrifugation or 
microfi ltration, virus removal or inactivation, cell lysis and inclusion body precipitation, 
product refolding, and purifi cation steps [71] . Conversely, those unit operations 
for drug product manufacturing procedures would be similar to those seen in the 
manufacture of a small molecule of comparable dosage form, namely mixing, fl uid 
transfer, sterile fi ltration, dose fi lling, lyophilization, and so on. Of course, unit operations 
will be dependent on the manufacturing process for the specifi c dosage form, 
but careful preformulation and characterization studies will enable relatively 
straightforward process design and ease subsequent scale - up activities. Modeling of 
unit operations for both small and large molecules is a recognized gap in our ability 
to achieve QbD. The application of accepted engineering methods to the problem 
is the subject of active research. 
1.1.6.2 Bioburden Considerations 
Bioburden refers to the amount of microbial fl ora that can be detected on an item, 
on a surface, or in a solution [68] . As mentioned previously, microbial contamination 
and bioburden are especially important for biotechnology - derived parenteral products 
since these products are typically capable of supporting microbial growth. 
Special care should be taken to ensure not only that the fi nal packaged product 
does not contain microbial contamination but also that manufacturing equipment 
is also free from contamination. Monitoring bioburden and determining potential 
levels of microbial contamination on equipment surfaces are particularly important 
with respect to the material being evaluated. 
In general, bioburden counts in parenteral solutions are obtained by conducting 
the total aerobic counts and total yeast and mold counts as specifi ed in the USP 
microbial limits test (61) or an equivalent test [72] . In addition, membrane fi ltration 
of larger than specifi ed volumes may also be used to detect any microbial contamination 
when sample results are expected to contain a negligible number of microbial 
fl ora or in the presence of potential confounding factors, such as antimicrobial 

preservatives [68, 72] . It is important to note that the presence of a high bioburden 
count can present an endotoxin contamination problem, as whole microbial cells 
and spores can be removed by sterilizing grade fi ltration (0.2 . m), while endotoxins 
are not [68] . These issues also underscore the importance of cleaning methods and 
their respective validation as well as assessing relevant product contamination on 
manufacturing equipment. 
1.1.6.3 Scale - Up and Process Changes 
The FDA defi nes process validation as “ establishing documented evidence that 
provides a high degree of assurance that a specifi c process will consistently produce 
a product meeting its predetermined quality attributes ” [73] . While validation studies 
are typically performed at full scale, in most cases scale - down or laboratory - scale 
models were used to initially develop the manufacturing process. Consequently, 
scale - down process precharacterization and characterization studies are considered 
crucial to successful process validation for both API and drug product manufacturing 
schemes [74] . Although they do require qualifi cation work and a signifi cant 
commitment of time and resources, characterization studies provide signifi cant 
insight into the critical process and control parameters for each unit operation as 
well as improved success rates for process validation due to a better, more complete 
understanding of the process [74] . In engineering terms, characterization studies 
identify the critical parameters useful for dimensional analysis that enable successful 
process scale - up. 
While the above explanation attempts to simplify the scale - up process, it is not 
meant to trivialize it. In fact, scale - up is probably the most diffi cult manufacturing 
challenge for traditional small molecules, let alone biopharmaceuticals. Issues such 
as homogeneous mixing, bulk product holding and transfer, and sterile fi ltration 
could all be potentially compounded due to the increased scale and introduced 
stress. However, a QbD approach to rational drug design should enable simplifi ed 
process scale - up and validation. This is only true if experimental design approaches 
have been utilized to identify the design space for the processes involved in the 
production of the molecule. This is also where the greatest benefi t of developing 
empirical phase diagrams early in development could materialize. Essentially, the 
QbD approach identifi es the quality attributes of the product based on scientifi c 
rationale as opposed to attempting to fi t the proverbial square peg into a round 
hole through a trial - and - error approach. This rational design approach goes further 
to identify the limiting factors of each unit operation and provides the means of 
attempting to correlate how each unit operation affects the fi nal product quality 
attributes. 
In order to initiate a successful QbD program, the fi rst step is to identify those 
process parameters that are essential to product quality and develop well - validated 
analytical methodologies to monitor those parameters. In short, the process involves 
identifi cation of the potential design space for production of the molecule and con- 
fi rmation that design space through rational, deliberate experimentation. Ideally, 
process monitoring should be done in real time to minimize production time and if 
possible online; however, this may not always be the case or even necessary depending 
upon the relative duration of the process to the test. Recognizing potential 
quality metrics earlier in the development process could also potentially facilitate 
QUALITY BY DESIGN AND SCALE-UP 27

28 BIOTECHNOLOGY-DERIVED DRUG PRODUCT DEVELOPMENT 
greater fl exibility during product development and subsequent process characterization 
[74] . Certainly, manufacturing site - specifi c differences could also potentially 
introduce variability into processes. It is for this reason that site - specifi c personnel 
training, process/technology transfer and validation, and stability assessments are 
required to ensure product quality. 
By defi nition, a process designed under the auspices of QbD should enable a 
degree of process knowledge that allows for controlled process changes without 
affecting the fi nal product or requiring regulatory approval. For immediate - and 
controlled - release solid dosage products, SUPAC guidelines provide direction on 
the studies to conduct to determine the impact of a process change. Although there 
is some regulatory guidance available for biological products (e.g., “ Changes to an 
Approved Application for Specifi ed Biotechnology and Specifi ed Synthetic Biological 
Products ” or “ FDA Guidance Concerning Demonstration of Comparability of 
Human Biological Products, Including Therapeutic Biotechnology - Derived Products 
” ), process changes need to be evaluated on a case - by - case basis. The comparative 
analysis of process changes should also be evaluated with respect to defi ned 
product specifi cations. PAT will be invaluable in determining the potential impact 
of process changes. While stability is often the main metric for small - molecule 
drug product, bioactivity and immunogenicity will need to be added metrics for 
biopharmaceuticals. Therefore, any process change should be approached subjectively 
and care should be taken to validate the relative impact on the safety and 
effi cacy of the product. 
1.1.7 CONCLUDING REMARKS 
Although the goals are the same, developing biotechnology molecules presents 
challenges that are unique compared to the development of conventional small 
molecules. The innate complexity of the molecular and macromolecular structures 
requires three dimensionally viable stability assays and understanding. The complexity 
of possible physiological responses and interactions requires an enhanced 
understanding of the formulation and processing stresses to identify the minor but 
critical changes that result in product unacceptability. A key to addressing these 
challenges is the development of analytical techniques with the sensitivity and reliability 
to detect and monitor such changes and to provide data to another gap - 
closing activity — modeling unit operations. Also the need to develop meaningful 
kinetic models is obvious to everyone involved in the development of both large 
and small molecules. Linking this type of information to the major efforts in the 
discovery arena is a necessary step to bringing the products of the future to 
market. 
The use of biotechnology products is increasing exponentially and many 
opportunities exist to improve their development. The fi rst step may be defi ning 
rational biotechnology - derived drug “ developability ” standards that can be assessed 
during preclinical/early development testing. Such a tiered approach based upon 
the potential risk, the confi dence in methodology, and benefi t has of course been a 
proven strategy for small molecules, and a preliminary version applicable to biotechnology 
drug products is likely possible today given the topics discussed in this 
chapter. 

ACKNOWLEDGMENTS 
The authors would like to thank The School of Pharmacy and Pharmaceutical Sciences, 
the Department of Industrial and Physical Pharmaceutics of Purdue University 
and the National Institutes of General Medical Sciences (R01 - GM65448) for 
their fi nancial support. 
REFERENCES 
1. International Conference on Harmonisation of Technical Requirements for Registration 
of Pharmaceuticals for Human Use, Q5C: Quality of biotechnological products: Stability 
testing of biotechnological/biological products, Nov. 30, 1995 . 
2. Biotechnology Industry Organization . Available: http://www.bio.org/ , accessed Jan. 10, 
2007 . 
3. Nims , R. , Presene , E. , Sofer , G. , Phillips , C. , and Chang , A. ( 2005 ), Adventitious agents: 
Concerns and testing for biopharmaceuticals , in Rathore , A. S. , and Sofer , G. , Eds., Process 
Validation in Manufacturing of Biopharmaceuticals: Guidelines, Current Practices, and 
Industrial Case Studies , Taylor and Francis , Boca Raton, FL . 
4. Rathore , A. S. , and Sofer , G. ( 2005 ), Life span studies for chromatography and fi ltration 
media , in Rathore , A. S. , and Sofer , G. , Eds., Process Validation in Manufacturing of Biopharmaceuticals: 
Guidelines, Current Practices, and Industrial Case Studies , Taylor and 
Francis , Boca Raton, FL . 
5. Crommelin , D. J. A. , Storm , G. , Verrijk , R. , de Leede , L. , Jiskoot , W. , and Hennink , W. E. 
( 2003 ), Shifting paradigms: Biopharmaceuticals vs. low molecular weight drugs , Int. J. 
Pharm. , 266 , 3 – 16 . 
6. Tang , L. , Persky , A. M. , Hochhaus , G. , and Meibohm , B. ( 2004 ), Pharmacokinetic aspects 
of biotechnology products , J. Pharm. Sci. , 93 ( 9 ), 2184 – 2204 . 
7. Roberts , M. J. , Bentley , M. D. , and Harris , J. M. ( 2002 ), Chemistry for peptide and protein 
pegylation , Adv. Drug Deliv. Rev. , 54 , 459 – 476 . 
8. Frokjaer , S. , and Otzen , D. ( 2005 ), Protein drug stability: A formulation challenge , Nat. 
Rev. , 4 , 298 – 306 . 
9. Niu , C. , and Chiu , Y. ( 1998 ), FDA perspective on peptide formulation and stability issues , 
J. Pharm. Sci. , 87 , 1331 – 1334 . 
10. Washington , N. , Washington , C. , and Wilson , C. G. ( 2001 ), Physiological Pharmaceutics: 
Barriers to Drug Absorption , 2nd ed., Taylor and Francis , New York. 
11. Hussain , A. , Arnold , J. J. , Khan , M. A. , and Ashan , F. ( 2004 ), Absorption enhancers in 
pulmonary protein delivery , J. Controlled Release , 94 , 15 – 24 . 
12. Alpar , H. O. , Somavarapu , S. , Atuah , K. N. , and Bramwell , V. W. ( 2005 ), Biodegradable 
mucoadhesive particulates for nasal and pulmonary antigen and DNA delivery , Adv. 
Drug Deliv. Rev. , 57 , 411 – 430 . 
13. Thanou , M. , Verhoef , J. C. , and Junginger , H. E. ( 2001 ), Chitosan and its derivatives as 
intestinal absorption enhancers , Adv. Drug Deliv. Rev. , 50 , 91 – 101 . 
14. Schellekens , H. ( 2002 ), Immunogenicity of therapeutic proteins: Clinical implications and 
future prospects , Clin. Ther. , 24 , 1720 – 1740 . 
15. Bhadra , D. , Bhadra , S. , Jain , P. , and Jain, N. K. (2002), Pegnology: A review of PEG -ylated 
systems , Pharmazie , 57 , 5 – 29 . 
16. Matthews , S. J. , and McCoy , C. ( 2004 ), Peginteferon . 2a: A review of approved and investigational 
uses , Clin. Ther. , 26 , 991 – 1025 . 
REFERENCES 29

30 BIOTECHNOLOGY-DERIVED DRUG PRODUCT DEVELOPMENT 
17. Knudsen , L. B. , Nielsen , P. F. , Huusfeldt , P. O. , Johansen , N. L. , Madsen , K. , Pedersen , 
F. Z. , Thogersen , H. , Wilken , M. , and Agerso , H. ( 2000 ), Potent derivatives of glucagon - 
like peptide - 1 with pharmacokinetic properties suitable for once daily administration , J. 
Med. Chem. , 43 , 1664 – 1669 . 
18. Foldvari , M. , Attah - Poku , S. , Hu , J. , Li , Q. , Hughes , H. , Babiuk , L. A. , and Kruger , S. ( 1998 ), 
Palmitoyl derivatives of interferon . : Potential for cutaneous delivery , J. Pharm. Sci. , 87 , 
1203 – 1208 . 
19. Egrie , J.C. , and Browne , J. K. ( 2001 ), Development and characterization of novel erythropoiesis 
stimulating protein (NESP) , Nephrol. Dial. Transplant. , 16 (Suppl 3 ), 3 – 13 . 
20. Haselbeck, A. (2003), Epoetins: Differences and their relevance to immunogenicity , Curr. 
Med. Res. Opin. , 19 , 430 – 432 . 
21. Hochuli , E. ( 1997 ), Interferon immunogenicity: Technical evaluation of interferon -. 2A , 
J. Int. Cytokine Res. , 17 , S15 – S21 . 
22. Maack , T. , Johnson , V. , Kau , S. T. , Figueiredo , J. , and Sigulem , D. ( 1979 ), Renal fi ltration, 
transport, and metabolism of low - molecular - weight proteins: A review , Kidney Int. , 16 , 
251 – 270 . 
23. Takagi , A. , Masuda , H. , Takakura , Y. , and Hashida , M. ( 1995 ), Disposition characteristics 
of recombinant human interleukin - 11 after a bolus intravenous administration in mice , 
J. Pharmacol. Exp. Ther. , 275 , 537 – 543 . 
24. Anderson , P. M. , and Sorenson , M. A. ( 1994 ), Effects of route and formulation on clinical 
pharmacokinetics of interleukin - 2 , Clin. Pharmacokinet. , 27 , 19 – 31 . 
25. Rabkin , R. , Ryan , M. P. , and Duckworth , W. C. ( 1984 ), The renal metabolism of insulin , 
Diabetologia , 27 , 351 – 357 . 
26. Johnson , V. , and Maack , T. ( 1977 ), Renal extraction, fi ltration, absorption, and catabolism 
of growth hormone , Am. J. Phsiol. , 233 , F185 – F196 . 
27. Carone , F. A. , and Peterson , D. R. ( 1980 ), Hydrolysis and transport of small peptides by 
the proximal tubule , Am. J. Physiol. , 238 , F151 – F158 . 
28. Carone , F. A. , Peterson , D. R. , and Flouret , G. ( 1982 ), Renal tubular processing of small 
peptide hormones , J. Lab. Clin. Med. , 100 , 1 – 14 . 
29. Braeckman , R. ( 2000 ), Pharmacokinetics and pharmacodynamics of protein therapeutics , 
in Reid , R. , Ed., Peptide and Protein Drug Analysis , Marcel Dekker , New York . 
30. Meissner , H. C. , Groothuis , J. R. , Rodriguez , W. J. , Welliver , R. C. , Hogg , G. , Gray , P. H. , 
Loh , R. , Simoes , E. A. , Sly , P. , Miller , A. K. , Nichols , A. I. , Jorkasky , D. K. , Everitt , D. E. , 
and Thompson , K. A. ( 1999 ), Safety and pharmacokinetics of an intramuscular 
monoclonal antibody (SB 209763) against respiratory syncytial virus (RSV) in infants 
and young children at risk for severe RSV disease , Antimicrob. Agent Chemother. , 43 , 
1183 – 1188 . 
31. Prescribing information for Lupron Depot ® . Manufactured for TAP Pharmaceuticals, 
Inc., Lake Forest, IL 60045. 
32. Prescribing information for Viadur ® . Manufactured by Alza Corporation, Mountain 
View, CA 94043. 
33. Degim , I. T. , and Celebi , N. ( 2007 ), Controlled delivery of peptides and proteins , Curr. 
Pharm. Des. , 13 , 99 – 117 . 
34. Sylvester , R. K. ( 2002 ), Clinical applications of colony - stimulating factors: A historical 
perspective , Am. J. Health - Syst. Pharm. , 59 , s6 – s12 . 
35. Dorr , R. T. ( 1993 ), Clinical properties of yeast - derived versus Escherichia coli - derived 
granulocyte - macrophage colony - stimulating factor , Clin. Ther. , 15 , 19 – 29 . 
36. Jenkins , N. , Shah , P. M. , and Buckberry , L. D. ( 2000 ), Carbohydrate analysis of glycoproteins 
and glycopeptides , in Reid , R. , Ed., Protein and Peptide Drug Analysis , Marcel 
Dekker , New York . 

37. Berglund , M. , Bystroem , K. , and Persson , B. ( 1990 ), Screening chemical and physical stability 
of drug substances , J. Pharm. Biomed. Anal. , 8 , 639 – 643 . 
38. International Conference on Harmonisation of Technical Requirements for Registration 
of Pharmaceuticals for Human Use, Q2(R1): Validation of analytical procedures: Text 
and methodology, Nov. 2005 . 
39. U.S. Pharmacopeia (USP) , Chapter . 111 . Design and analysis of biological assays, USP 
26, 2003, Rockville, MD. 
40. Center for Drug Evaluation and Research, FDA ( 2001 , May). Guidance for industry, 
bioanalytical method validation, FDA, Washington, DC. 
41. International Conference on Harmonisation of Technical Requirements for Registration 
of Pharmaceuticals for Human Use, Q8: Pharmaceutical development, Nov. 10, 2005 . 
42. International Conference on Harmonisation of Technical Requirements for Registration 
of Pharmaceuticals for Human Use, Q8: Concept paper, available: http://www.ich.org/ 
LOB/media/MEDIA3096.pdf , accessed Sept. 19, 2003 . 
43. Avis , K. E. , and Levchuk , J. W. ( 2000 ), Remington 20th Edition: Parenteral Preparations , 
Lippincott, Williams & Wilkins , Philadelphia. 
44. Autian , J. ( 1968 ), Interrelationship of the properties and uses of plastics for parenterals , 
Bull. Parenteral Drug Assoc. , 22 , 276 – 288 . 
45. Avis , K. E. ( 1986 ), Sterile Products , in Lachman , L. , Lieberman , H. A. , and Kanig , J. L. , 
Eds., The Theory and Practice of Industrial Pharmacy, 3rd ed., Sterile Products , 3rd 
Ed. , Lippincott , Williams and Wilkins , reprint with permission by Stipes Publishing , 
Champaign, IL . 
46. Gombotz , W. , Pankey , S. , Bouchard , L. , Phan , D. , and MacKenzie , A. ( 2002 ), Stability, 
characterization, formulation, and delivery system development for transforming growth 
factor - beta1 , in Pearlman , R. , and Yang , Y. , Eds., Pharmaceutical Biotechnology , Vol. 9 , 
Formulation, Characterization, and Stability of Protein Drugs: Case Histories , Springer , 
New York . 
47. Bontempo , J. A. ( 1997 ), Considerations for elastomeric closures for parenteral biopharmaceutical 
drugs , in Bontempo , J. A. Ed., Development of Biopharmaceutical Parenteral 
Dosage Forms , Marcel Dekker , New York . 
48. Banga , A. K. ( 2006 ), Therapeutic Peptides and Proteins: Formulation, Processing and 
Delivery Systems , 2nd ed., Taylor and Francis , Boca Raton, FL. 
49. Yamamoto , O. ( 1992 ), Effect of radiation on protein stability , in Ahern , T. J. , and Manning , 
M. C. Eds., Pharmaceutical Biotechnology , Vol. 2 , Stability of Protein Pharmaceuticals. 
Part A: Chemical and Physical Pathways of Protein Degradation , Plenum , New York . 
50. Garfi nkle , B. D. , and Henley , M. W. ( 2000 ), Remington 20th Edition: Sterilization , Lippincott, 
Williams & Wilkins , Philadelphia. 
51. PDA Technical Report No. 26 ( 1998 ), Sterilizing fi ltration of liquids , PDA J. Pharm. Sci. 
Tech. , 52 ( 3 , Suppl). 
52. Defelippis , M. R. , and Akers , M. J. ( 2000 ), Pharmaceutical Formulation Development of 
Peptides and Proteins: Peptides and Proteins as Parenteral Suspensions: An Overview 
of Design, Development, and Manufacturing Considerations , Taylor and Francis , 
Philadelphia . 
53. Timasheff , S. N. ( 1998 ), Control of protein stability and reactions by weakly interacting 
cosolvents: The simplicity of the complicated , Adv. Protein Chem. , 51 , 355 – 432 . 
54. Shimizua , S. , and Smith , D. J. ( 2004 ), Preferential hydration and the exclusion of cosolvents 
from protein surfaces , J. Chem. Phys. , 121 , 1148 – 1154 . 
55. Wakankar , A. A. , and Borchardt , R. T. ( 2006 ), Formulation considerations for proteins 
susceptible to asparagine deamidation and aspartate isomerization , J. Pharm. Sci. , 95 , 
2321 – 2336 . 
REFERENCES 31

32 BIOTECHNOLOGY-DERIVED DRUG PRODUCT DEVELOPMENT 
56. Windheuser , J. J. ( 1963 ), The effect of buffers on parenteral solutions , Bull. Parenteral 
Drug Assoc. , 17 , 1 – 8 . 
57. Im - Emsap , W. , Paeratakul , O. , and Siepmann , J. ( 2002 ), Disperse Systems , in Banker , G. 
S. , and Rhodes , C. T. , Eds., Modern Pharmaceutics , 4th Ed. , Marcel Dekker , New York . 
58. U.S. Pharmacopeia (USP 26 2003) , Chapter . 51 . Antimicrobial effectiveness testing, 
Rockville, MD. 
59. International Conference on Harmonisation of Technical Requirements for Registration 
of Pharmaceuticals for Human Use, Q1A(R2): Stability testing of new drug substances 
and products, Feb. 6, 2003 . 
60. Fan , H. , Li , H. , Zhang , M. , and Middaugh , C. R. ( 2007 ), Effects of solutes on empirical 
phase diagrams of human fi broblast growth factor 1 , J. Pharm. Sci. , 96 , 1490 – 1503 . 
61. Harn , N. , Allan , C. , Oliver , C. , and Middaugh , C. R. ( 2006 ), Highly concentrated monoclonal 
antibody solutions: Direct analysis of physical structure and thermal stability , 
J. Pharm. Sci. , 96 , 532 – 546 . 
62. Peek , L. J. , Brey , R. N. , and Middaugh , C. R. ( 2007 ), A rapid, three - step process for the 
preformulation of a recombinant ricin toxin A - chain vaccine , J. Pharm. Sci. , 96 , 44 – 60 . 
63. Violand , B. N. , and Siegel , N. R. ( 2000 ), Protein and peptide chemical and physical stability 
, in Reid , R. , Ed., Protein and Peptide Drug Analysis , Marcel Dekker , New York . 
64. Manning , M. C. , Patel , K. , and Borchardt , R. T. ( 1989 ), Stability of protein pharmaceuticals 
, Pharm. Res. , 6 , 903 – 918 . 
65. International Conference on Harmonisation of Technical Requirements for Registration 
of Pharmaceuticals for Human Use, Q1B: Stability testing: Photostability testing of new 
drug substances and products, Nov. 6, 1996 . 
66. Fasani , E. , and Albini , A. ( 2005 ), Photostability stress testing , in Baertschi , S. W. , Ed., 
Pharmaceutical Stress Testing: Predicting Drug Degradation , Taylor and Francis , Boca 
Raton, FL . 
67. Ghosh , T. , Kalra , A. , and Garde , S. ( 2005 ), On the salt - induced stabilization of pair and 
many - body hydrophobic interactions , J. Phys. Chem. B. Condens. Matter Mater. Surf. 
Interfaces Biophys. , 109 , 642 – 651 . 
68. Birrer , G. A. , Murthy , S. S. , Liu , J. , and Estrada , J. ( 2001 ), Parenteral dosage forms , in 
Ahuja , S. , Ed., Handbook of Modern Pharmaceutical Analysis , Academic , New York . 
69. Birrer , G. A. , Liu , J. , Halas , J. M. , and Nucera , G. G. ( 2000 ), Evaluation of a container 
closure integrity test model using visual inspection with confi rmation by near infra - red 
spectroscopic analysis , PDA J. Pharm. Sci. Technol. , 54 , 373 – 382 . 
70. International Conference on Harmonisation of Technical Requirements for Registration 
of Pharmaceuticals for Human Use, Q1E: Evaluation for stability data, Feb. 6, 2003 . 
71. Godavarti , R. , Petrone , J. , Robinson , J. , Wright , R. , and Kelley , B. D. ( 2005 ), Scale - down 
models for purifi cation processes: Approaches and applications , in Rathore , A. S. , and 
Sofer , G. Eds., Process Validation in Manufacturing of Biopharmaceuticals: Guidelines, 
Current Practices, and Industrial Case Studies , Taylor and Francis , Boca Raton, FL . 
72. U.S. Pharmacopeia (USP 26 2003) , Chapter . 61 . Microbial limits test, USP, Rockville, 
MD. 
73. Center for Drug Evaluation and Research, FDA ( 1987 , May), Guideline on general 
principles of process validation. 
74. Seely , J. E. ( 2005 ), Process characterization , in Rathore , A. S. , and Sofer , G. Eds., Process 
Validation in Manufacturing of Biopharmaceuticals: Guidelines, Current Practices, and 
Industrial Case Studies , Taylor and Francis , Boca Raton, FL . 

33 
1.2 
REGULATORY CONSIDERATIONS IN 
APPROVAL OF FOLLOW - ON PROTEIN 
DRUG PRODUCTS 
Erin Oliver, 1 Stephen M. Carl, 2 Kenneth R. Morris, 2 
Gerald W. Becker, 3 and Gregory T. Knipp 1 
1 Rutgers, The State University of New Jersey, Piscataway, New Jersey 
2 Purdue University, West Lafayette, Indiana 
3 SSCI, West Lafayette, Indiana 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc. 
Contents 
1.2.1 Introduction 
1.2.1.1 Emergence of Biotechnology Industry 
1.2.1.2 Challenges Facing “ Biogenerics ” 
1.2.2 History of Biologics Regulation in United States 
1.2.2.1 Early Biologics Regulation (1800s – 1990s) 
1.2.2.2 Modern Biologics Regulation (1990s – Today) 
1.2.3 Regulatory Classifi cation of Proteins 
1.2.3.1 Defi nitions and Key Terminology 
1.2.3.2 Application of Defi nitions to Proteins: Is It a Drug or a Biologic? 
1.2.3.3 Regulatory Approval Path for Proteins 
1.2.4 Regulation of Generic Drugs 
1.2.4.1 History of Generic Drug Legislation in United States 
1.2.4.2 Approval Process for Generic Drugs 
1.2.4.3 Application of Generic Regulations to Biologics 
1.2.5 Legal Arguments Related to Follow - On Proteins 
1.2.5.1 Constitutionality of 505(b)(2) Process for Drugs 
1.2.5.2 Constitutionality of 505(b)(2) Process for Follow - On Proteins 
1.2.5.3 Applicability of 505(j)(1) or ANDA Process to Biogenerics 
1.2.5.4 Current Rules Relating to Bioequivalence of Generic Drugs 
1.2.5.5 Statutory Authority 
1.2.6 Scientifi c Issues Related to Follow - On Proteins (Data Requirements) 
1.2.6.1 “ Sameness ” as per Orphan Drug Regulations 
1.2.6.2 “ Sameness ” as per Postapproval Change Guidances 

34 REGULATORY CONSIDERATIONS IN APPROVAL 
1.2.7 Proposed Regulatory Paradigm: Case Studies 
1.2.7.1 Case Study 1: Fortical [Calcitonin - Salmon (rDNA Origin)] 
1.2.7.2 Case Study 2: Omnitrope [Somatropin (rDNA Origin)] 
1.2.7.3 Case Study 3: Generic Salmon Calcitonin 
1.2.8 Summary and Conclusions 
References 
1.2.1 INTRODUCTION 
The ongoing need to provide the U.S. population with cost - effective pharmacological 
therapies has led to an emergent public health initiative in this country, namely 
for generic versions of therapeutic proteins. Greater access to generic drugs was 
made possible by the passage of the 1984 Drug Price Competition & Patent Term 
Restoration Act, commonly referred to as Hatch – Waxman. Generics have historically 
afforded considerable savings to the American consumer in need of prescription 
medication. Ten years after the Hatch – Waxman amendments, the Congressional 
Budget Offi ce estimated that purchasers saved a total of $ 8 – $ 10 billion on prescriptions 
at retail pharmacies by substituting generic drugs for their brand - name counterparts 
in 1994 [1] . To put those numbers in the context of today ’ s pharmaceutical 
landscape, a recent report issued by the U.S. Department of Health and Human 
Services estimates that generic drugs constitute 63% of the total prescription medicines 
sold in the United States [2] . This same report suggests that generic drugs cost 
approximately 11% of the total cost of branded pharmaceuticals (on a per - dose 
basis). 
At the same time, the development and use of therapeutic proteins have increased 
dramatically, with more than 850 biotechnology drug products and vaccines currently 
in trials [3] . Further, it is estimated that by the year 2010 nearly one - half of 
all newly approved medicines will be of biological origin [4] . The industrial fi nancial 
incentives for the pursuit of follow - on biologics (heretofore termed biogenerics) are 
substantial with sales of biotechnology medicines in the United States rising 17% 
to approximately $ 30 billion in 2005 and growing at an annual rate of about 20% 
thereafter [3] . 
Not unexpectedly, the U.S. Food and Drug Administration (FDA) is experiencing 
mounting pressure to progress the cause of biogenerics. In a letter dated February 
10, 2006, Senators Henry Waxman and Orrin Hatch (authors of the original “ generic ” 
legislation) urged the FDA to develop and implement clear guidelines for the 
approval of follow - on biological products for certain well - characterized proteins 
like insulin and human growth hormone (HGH) [5] . Additionally, recent litigation 
has compelled the FDA to take action on a pending drug application for a follow - on 
protein (FOP) drug product [Omnitrope, somatropin (recombinant DNA, rDNA 
origin)] [6] . 
A signifi cant barrier to the emergence of “ biogenerics ” is the absence of a clear, 
effi cient abbreviated pathway for approval. This hurdle is linked to signifi cant scientifi 
c and legal issues in the United States in terms of how proteins are classifi ed 
(drug vs. biologic) and subsequently regulated as well as how “ generics ” are tradi

HISTORY OF BIOLOGICS REGULATION IN UNITED STATES 35 
tionally defi ned in terms of equivalence and substitutability. However, an examination 
of the vast array of biologicals on the market today reveals that not all proteins 
are created equal. This range of complexity may provide an opportunity for stepwise 
progress on the regulatory front. This chapter presents the background to this multifaceted 
issue and examines the key regulatory challenges facing biogenerics today. 
An appropriate regulatory paradigm for the approval of FOPs is proposed and 
supported though a discussion of recent case studies. 
1.2.1.1 Emergence of Biotechnology Industry 
The explosion of scientifi c advances over the last quarter century has spawned the 
biotechnology industry and whole new classes of therapeutic agents for the treatment 
and prevention of disease. In October of 1982, the FDA approved the fi rst 
protein - based therapeutic created by DNA technology in the form of Humulin 
(recombinant insulin). Developed by Eli Lilly & Co., with technical assistance from 
Genentech, Humulin is indicated for the treatment of diabetes. At the time, the use 
of recombinant technology was somewhat limited to the production of smaller, 
nonglycosylated proteins such as insulin (51 amino acids) and HGH (191 amino 
acids) using bacterial hosts. The seminal discovery by Columbia ’ s Richard Axel of 
the process of cotransformation enabled complex protein production and glycosylation 
and thus spurred the emergence of the modern biotechnology industry [7] . 
The phenomenal growth observed in the biotechnology sector is notable in terms 
of the extraordinary number and diversity of therapeutic peptides and proteins that 
have been developed within a period of only about 20 years. Examples of therapeutic 
proteins in current use include cytokines, clotting factors, vaccines, and monoclonal 
antibodies, as illustrated in Table 1 [8] . 
As presented in Table 2 , many of these “ early ” biotechnology products have 
reached the end of their period of patent exclusivity [4 – 9] . Thus, it is appropriate to 
now consider the next steps in the “ life cycle ” of these products as potential generic 
drugs. 
1.2.1.2 Challenges Facing “ Biogenerics ” 
The diversity and complexity of biologic molecules that drive their utility as therapeutic 
agents also contribute to the diffi culty in classifying them as pharmacological 
entities, namely, whether they are drugs or biologics. This diffi culty in classifi cation 
is of profound importance since there are fundamental differences in how the FDA 
regulates drugs and biologics. 
To appreciate the current challenges facing the pharmaceutical and biotechnology 
industry, it is informative to review the historical background associated with 
the classifi cation and regulation of biologics in the United States, particularly in the 
context of the nation ’ s evolving drug regulation system. 
1.2.2 HISTORY OF BIOLOGICS REGULATION IN UNITED STATES 
Due to the scientifi c limitations of the early to mid - 1900s, signifi cant differences 
existed between the approaches taken to manufacture and analyze biologics and 

36 REGULATORY CONSIDERATIONS IN APPROVAL 
TABLE 1 Examples of Therapeutic Peptide and Protein Molecules Currently Marketed 
in United States 
Peptides Antibiotics: bacitracin, bleomycin, gramicidine, capreomycin 
Hormones: corticotropin, glucagon, gonadrolein HCl, leuprolide 
acetate, histrelin acetate, oxytocin, secretin, goserelin acetate, 
vassopressin 
Others: polymixin B, eptifi batide, cyclosporine 
Nonglycosylated 
proteins 
Interleukins: andresleukin (IL - 1), denileukin diftitox (fusion, protein - 
IL - 2+ DT), anakinra (IL - 2) 
Interferons: interferon alpha - n1, interferon alpha - n3, interferon alpha - 
2a, peg interferon alfa - 2b, interferon alfacon - 1, Interferon alpha - 2b, 
interferon beta - 1b, interferon gamma - 1b, 
Enzymes/inhibitors: anistreplase, asparaginase, lactase, trypsin, alpha - 
1 proteinase inhibitor, urokinase, deoxyribonuclease, fi brinolysin, 
chymotrypsin, pancreatin, papain, urokinase 
Growth factors/hormones: Filigrastim pegfi lgrastim, somatropin, 
becaplermin, somatrem, menotropins 
Antithrombotic agents: thrombin, fi brinogen, hirudin, hirulog, fi brin 
Others: insulin, gelatin, prolactin, albumin (human), hemoglobin, 
collagen 
Glycosylated 
proteins 
Interferon beta - 1a 
Antithrombotic agents: alteplase, drotrecogin alfa, antithrombin III 
Antianemic: darbopoetin alfa, erythropoietin 
Growth hormones: follitropin alpha, follitropin beta, chorionic 
gonadotropin (Human) 
Immuno globulins (IG): pertusssis IG, rabies IG, tetanus IG, hepatitis 
B IG, varicella zoster IG, rho(D) IG, normal immune globulin, 
lymphocyte anti - thymocyte, IB (equine) 
Coagulation factors: factor VII antihemophilic factor, factor IX 
(human, recombinant) 
Factor VIII (others): etanercept (CSF), sargramostim (TNF) 
Monoclonal 
antiobodies 
avciximab, alemtuzamub, basiliximab, gentuzumab, satumomab, 
infl ixibam, palivizumab 
drugs. This reality led to the creation of separate and distinct regulatory pathways 
for drugs and biologics. As noted earlier, the developments in analytical chemistry 
and improvements in process technologies have, in recent times, blurred the lines 
between drug and biologic drug development. In the current era of pharmaceutical 
development and standards harmonization, one might question the continued need 
for two distinct pathways. Recognizing the shifting paradigm of drug development, 
the history of biologics regulation is discussed below in two parts: early history and 
present day. 
1.2.2.1 Early Biologics Regulation (1800s – 1990s) 
This country ’ s earliest experience with biologics dates back to the infectious scourges 
of the late 1800s and early 1900s when epidemics of typhoid, yellow fever, smallpox, 
diphtheria, and tuberculosis were being battled by new advances in immunology. 
The discovery and development of vaccines and antitoxins led to the creation of a 

HISTORY OF BIOLOGICS REGULATION IN UNITED STATES 37 
TABLE 2 Patent Expiration Dates for U.S. Marketed Biologics 
Brand Name Generic Name Indication Company 
Patent 
Expiry 
Humulin Recombinant insulin Diabetes Eli Lilly Expired 
Nutropin Somatropin Growth disorders Genentech Expired 
Abbokinase Eudurase urokinase Ischaemic events Abbott Expired 
Ceredase Alglucerase Gaucher disease Genzyme Expired 
Cerezyme Imiglucerase Gaucher disease Genzyme Expired 
Streptase Streptokinase Ischaemic events AstraZeneca Expired 
Intron A IFN - . - 2b Hepatitis B and C Biogen/Roche Expired 
Serostim Somatropin AIDS wasting Serono Expired 
Humatrope Somatropin Growth disorders Eli Lilly Expired 
Geref Sermorelin Growth hormone 
defi ciency 
Serono Expired 
(2004) 
Synagis Palivizumab Respiratory 
syncytial virus 
Abbott Expired 
(2004) 
Novolin Human insulin Diabetes Novo Nordisk 2005 
Protropin Somatrem Growth hormone 
defi ciency 
Genentech 2005 
TNKase Tenecteplase 
TNK - tPA 
Acute myocardial 
infarction 
Genentech 2005 
Actimmmune IFN - . - 1b Chronic 
granulomatous 
disease; 
malignant 
osteoporosis 
InterMune 2005, 2006, 
2012 
Activase, 
Alteplase 
tPA Acute myocardial 
infarction 
Genentech 2005, 2010 
Proleukin IL - 2 HIV Chiron 2006, 2012 
Epogen, 
Procrit, 
Eprex 
Erythropoietin Anemia Amgen 2013 
Neupogen Filgrastim (G - CSF) Anemia, 
leukemia, 
neutropenia 
Amgen 2015 
Note: Based on our search of available patent sites for only the reference product. 
IFN - Interferon; tPA - Tissue Plasminogen Activator, IL - interleukin; HIV - Human Immunodefi ciency 
Virus; G-CSF- Granulocyte-Colony Stimulating Factor; TNKase- Tenecteplase. 
whole new “ biopharmaceutical ” industry. As demand increased, the pharmaceutical 
manufacturers responded and in turn supplanted the government ’ s role in the public 
supply of vaccines (per Vaccine Act of 1813) [10] . Unfortunately, the commercialization 
of vaccines by smaller, less experienced, and likely less scrupulous manufacturers 
led to problems. Similar to the history of drug regulation, early advances in 
biologics regulation could be characterized as responsive rather than proactive. 
Change often occurred following tragedy and the result of government ’ s attempt to 
respond. Some of the key milestones of early biologics regulation are summarized 
in Table 3 . The following years saw many administrative changes in terms of the 
specifi c governmental agency responsible for regulating biologics, but with few 
substantive changes to the regulations themselves. 

38 REGULATORY CONSIDERATIONS IN APPROVAL 
TABLE 3 Key Milestones in Early Biologics Regulation 
1901 Ten children died in St. Louis from administration of tetanus - contaminated 
diphtheria antitoxin. In this case, no safety testing had been performed prior to 
use. 
1902 Biologics Control Act (BCA) signed into law: 
• Authorizing the regulation of commercial viruses, serums, toxins, and analogous 
products 
• Requiring the licensure of biologics manufacturers and establishments 
• Providing governmental inspectional authority 
• Making it illegal for the commercial distribution of product not manufactured 
and labeled in accordance with the act 
1906 Pure Food and Drug Act enacted (precursor of modern - day drug regulation). Lack 
of mention of biologics as a class effectively represents fi rst distinction between 
drug and biologic regulation. 
1919 BCA amended: 
• Required reporting of changes in equipment, manufacturing processes, personnel; 
establishment of formal quality control procedures; and submission of samples 
for regulatory inspection and approval for release 
• Recognized potential that slight changes to manufacturing conditions (raw 
materials, process, personnel, etc.) could have signifi cant and adverse effect on 
product quality 
• Required strict control of input (environment and manufacturing conditions) 
rather than end - stage testing of quality attributes due to limitations in analytical 
methodology to detect these effects 
1937 Elixir sulfanilamide, containing the poisonous solvent diethylene glycol, kills 107, 
many of whom are children. 
1938 Food, Drug and Cosmetic Act (FDCA) enacted: 
• Established concept of “ new drugs ” requiring proof of safety prior to marketing 
• Required submission of an investigational new Drug (IND) application prior to 
clinical use of an experimental drug in humans 
• Required approval of a new drug application (NDA) prior to commercial sale of 
drugs 
• Granted federal government power of seizure of misbranded or adulterated 
drugs 
• Defi ned “ drugs ” comprehensively; not excluding potential of “ biologics ” to 
function as drugs 
1941 • Approximately 300 deaths and injuries result from distribution of sulfathiazole 
tablets tainted with the sedative phenobarbital. 
• Insulin Amendment passed to require FDA testing/certifi cation of purity and 
potency. 
1944 Public Health Service (PHS) Act enacted to consolidate and codify previous 
biologics laws: 
• Outlined licensing requirements for biologics — for both product (product 
licensing application, or PLA) and establishment where the product was 
manufactured (establishment licensing application, or ELA) 
• Required submission of samples of each manufactured lot of all biologicals for 
government testing and certifi cation prior to commercial release 
• Required sponsors to own all of manufacturing facilities, effectively eliminating 
multiparty or contract manufacturing 

HISTORY OF BIOLOGICS REGULATION IN UNITED STATES 39 
1.2.2.2 Modern Biologics Regulation (1990s – Today) 
Whereas early biologics regulation was grounded by technical limitations, modern 
biologics regulation is driven by tremendous advances in scientifi c knowledge. 
Development of analytical tools and techniques has dramatically increased the 
ability to characterize proteins and substantiate the structure, composition, and 
function of the therapeutic molecule. These advances enable the detection of small 
differences in molecular weight; elucidation of primary, secondary, and tertiary 
protein structures; detection and quantifi cation of posttranslational modifi cations 
(i.e., patterns of glycosylation); and improved understanding of structure – function 
relationships and potential immunogenic responses. Simultaneously, developments 
in the fi elds of pharmaceutical and biotechnological manufacturing have greatly 
improved process effi ciency and control. This recent technological evolution has had 
a direct impact on biologics regulation as refl ected below in several key events: 
• In 1995, the FDA agreed to eliminate lot testing requirements for certain highly 
characterized products once the company ’ s ability to consistently manufacture 
product of acceptable quality was established. 
• In 1996, the FDA and Congress dismantled the dual - licensing process, requiring 
the submission of a single BLA (biologics license application), making the 
content and format of a biologics application similar to that required for new 
drug applications (NDAs). 
• In 1996, the Center for Biologics Evaluation and Research (CBER) liberalized 
its defi nition of “ legal manufacturer ” and eliminated many of the barriers to 
cooperative, multiparty manufacturing arrangements. 
• In 1997, Congress passed a noteworthy piece of legislation affecting modern 
pharmaceutical regulation in the Food and Drug Modernization Act (FDAMA). 
Among the many goals of the act was to harmonize the drug and biologic 
approval processes. 
In fact, current pharmaceutical/regulatory initiatives appear to extract the best 
practices from biologic and drug approaches which can apply equally to both classes 
of products: 
• The Quality Systems Approach and GMPs for the 21st Century, two initiatives 
being pursued by the FDA for drugs and devices, emphasize the utility of building 
quality into the process, consistent with the strict control of “ input factors ” 
seen in early biologic regulation. 
• Initiatives such as Process Analytical Technologies build on the concept of 
conventional drug product testing using increasingly sophisticated analytical 
techniques to provide continuous process monitoring and fi nished - product 
quality assurance of multiple pharmaceutical dosage forms. 
• The current global initiative to harmonize electronic submission format and 
content requirements effectively creates one standard data package for drugs 
or biologics. Thus, the eNDA (electronic new drug application) or eBLA (electronic 
biologics license application) will eventually be replaced by the eCTD 
(electronic common technical document). 

40 REGULATORY CONSIDERATIONS IN APPROVAL 
1.2.3 REGULATORY CLASSIFICATION OF PROTEINS 
Despite the blurring of lines between drugs and biologics, there remain two different 
mechanisms to bring protein drug products to the U.S. marketplace. The choice of 
approval framework is dependent on the protein ’ s classifi cation as a drug or biologic. 
The history of this regulatory distinction is rooted in the technical differences 
between small - molecule drugs and macromolecular biologics. Traditionally, drugs 
were characterized as having well - defi ned chemistry. Conversely, biologics were 
large, complex macromolecules whose active moiety defi ed characterization and 
quantitation. By necessity, different means of assuring the safety and effi cacy of 
these therapeutic products were required at the time. The modern - day consequence 
is a legal system that distinguishes between proteins as drugs and proteins as biologics. 
The distinction is based on statutory defi nitions as well as historical precedent 
and has implications in terms of the approval pathways for original and follow - on 
products. 
1.2.3.1 Defi nitions and Key Terminology 
Drugs are defi ned by the U.S. Food and Drug Act [FD & C Act, 21 U.S.C. 321(g)(1)] by 
function as any article Federal Food, Drug and Cosmetic Act (a) intended for use 
in the diagnosis, cure, mitigation, treatment, or prevention of disease in man or 
animals and (b) intended to affect the structure or function of the body [11] . 
Biologics as a class may be regulated as drugs but are defi ned within the Public 
Health Service Act [PHSA, 42 U.S.C. 262(a)] by category as “ a virus, therapeutic 
serum, toxin, antitoxin, vaccine, blood, blood component or derivative, allergenic 
product, or analogous product, or arsphenamine (or any other trivalent organic 
arsenic compound), applicable to the prevention, treatment, or cure of diseases or 
injuries of humans ” [12] . 
A cursory examination of these defi nitions reveals that they are not mutually 
exclusive, leading to confusion about how to appropriately and consistently 
apply them. This point is illustrated when one reviews the history of how the FDA 
has categorized and subsequently regulated these drugs and biologics as shown 
below. 
1.2.3.2 Application of Defi nitions to Proteins: Is It a Drug or a Biologic? 
The answer to this fundamental question is not straightforward and has evolved 
over time. Historically, some natural - source - derived proteins such as insulin, hyaluronidase, 
menotropins, and Human Growth Hormone (HGH) have been regulated 
as drugs. While other natural - source - proteins such as blood factors were regulated 
as biologics. When recombinant proteins and monoclonal antibodies began development 
in the 1970s – 1980s, these were regulated as follows: 
1. By the Center for Drug Evaluation and Research (CDER) under the Food, 
Drug and Cosmetic Act (FDCA) as drugs when they were hormones such as 
insulin, HGH, and parathyroid hormone (PTH) derivatives 
2. By the CBER under the PHSA as biologics when they were cytokines or blood 
factors such as factor VIII for hemophilia 

As other recombinant proteins and monoclonal antibodies came under development, 
the CBER held primary responsibility for this review, with the CDER retaining 
responsibility for hormones such as insulin and HGH. However, in 2003 all 
therapeutic proteins were transferred from the CBER to the CDER. This reassignment 
of review responsibility did not impact the legal classifi cation of these protein 
products, such that the Center for Drug Evaluation and Research assumed responsibility 
for the review and approval of biologics approved under Section 351 of the 
PHSA. 
The basic distribution of these therapeutic biologics to the respective FDA center 
is refl ected in Table 4 ; however, many of the current complex biotechnology - derived 
products do not fi t neatly into accepted defi nitions and require case - by - case classi- 
fi cation [13] . 
1.2.3.3 Regulatory Approval Path for Proteins 
The relevance of the preceding discussion becomes important with the understanding 
that therapeutic proteins classifi ed as drugs are governed under a different set 
of laws than those classifi ed as biologics. Drugs are approved via submission of 
NDAs under Section 505 of the FD & C Act, while biologics are supported by BLAs 
under the PHSA. These two approval paths are similar in terms of application 
content, that is, requirement of complete reports of clinical safety and effi cacy data 
to support approval. However, only the drug regulation, that is, Section 505 of the 
FD & C Act, has been amended to outline an abbreviated approval mechanism for 
generic products. 
1.2.4 REGULATION OF GENERIC DRUGS 
1.2.4.1 History of Generic Drug Legislation in United States 
In 1984, Congress responded to America ’ s need for safe, affordable medicines by 
passing a pivotal piece of legislation, The Drug Price Competition and Patent Term 
TABLE 4 FDA Center Regulatory Responsibility for Therapeutic Biological Products 
CDER CBER 
Monoclonal antibodies (in vivo use) 
Proteins intended for therapeutic use: 
Cytokines (e.g., interferons) 
Enzymes (e.g., thrombolytics) 
other novel proteins except those assigned to 
CBER 
Immunomodulators (nonvaccine, nonallergenic) 
Growth factors, cytokines, some hormones and 
monoclonal antibodies intended to mobilize, 
stimulate, decrease, or otherwise alter the 
production of hematopoietic cells in vivo 
Cellular product, including products 
composed of human, bacterial, or 
animal cells 
Vaccines 
Allergenic extracts 
Antitoxins, antivenoms, venoms 
Blood, blood components, plasma - 
derived products (e.g., albumin, 
immunoglobulins, clotting factors, 
fi brin sealants, proteinase inhibitors), 
recombinant and transgenic versions 
of plasma derivatives 
REGULATION OF GENERIC DRUGS 41

42 REGULATORY CONSIDERATIONS IN APPROVAL 
Restoration Act (Hatch – Waxman amendments). The intent of this act was to effectively 
balance the need to encourage pharmaceutical innovation with the desire to 
accelerate the availability of lower cost alternatives to approved drugs. The act also 
sought to eliminate unnecessary or redundant clinical testing to protect patients 
(reduce the number of patients in need receiving placebo in controlled clinical 
trials) and conserve industry and agency resources. To accomplish the goal of faster 
to market, cheaper alternatives, the amendments stipulated the following [14] : 
• For Innovator Companies The act encouraged continued innovation, research, 
and development activities by providing manufacturers with meaningful incentives 
in the form of patent protection/restoration and marketing exclusivity, 
thus allowing them to recoup some of their investments. 
• For Generic Companies The act provided access to certain innovator information 
without the threat of legal action via patent infringement suits (safe harbor 
provisions), allowing generics the opportunity to prepare for market introduction 
prior to the expiration of patent/exclusivity terms. This effectively limited 
the period of innovator exclusivity to the statutory timelines. 
1.2.4.2 Approval Process for Generic Drugs 
The act served as a boon to the generic industry by paving the path to abbreviated 
and accelerated drug approvals. From a legal perspective, the Hatch – Waxman 
amendments modifi ed Section 505 of the FD & C Act to create two new abbreviated 
approval pathways (see Table 5 ) [14] . 
In essence, the abbreviated NDA (ANDA) and 505(b)(2) processes allow generic 
manufacturers the ability to rely on what is already known about the drug and refer 
to the agency ’ s fi nding of safety and effi cacy for the innovator. For an ANDA, the 
generic product must meet certain criteria related to bioequivalence and product 
sameness. However, a 505(b)(2) application often describes a drug with substantial 
differences to the innovator (which would seem more closely related to FOPs). 
1.2.4.3 Application of Generic Regulations to Biologics 
A central question is “ Do biologics fall under the provisions of the Hatch – Waxman 
Act? ” Since the Hatch – Waxman Act specifi cally amended the FD & C Act, biologics 
TABLE 5 Description of NDA Approval Mechanisms 
Traditional path 1. 505(b)(1) — Application that contains full reports of investigations 
of safety and effectiveness to which sponsor has right of reference 
(stand - alone NDA) 
Abbreviated path 2. 505(b)(2) — Application that contains full reports of investigations 
of safety and effectiveness, where the sponsor relies on studies 
conducted by someone else to which the sponsor does not have 
right of reference 
Abbreviated path 3. 505(j)(1) — Abbreviated new drug application (ANDA) containing 
information to show the product is a duplicate of an already 
approved drug product 

approved via a BLA under the PHSA are not covered by this legislation nor does 
the PHSA have similar provisions for biogenerics. However, those few therapeutic 
proteins approved via Section 505 of the FDCA as NDAs are covered by the 
Hatch – Waxman amendments and thus are legally considered appropriate for fi ling 
a 505(b)(2) or 505(j)(1) application. For simple, well - characterized peptides and 
proteins regulated under Section 505 of the FD & C Act, mechanisms are already in 
place to bring FOPs to the market. In fact, several FOPs have already been approved 
by the FDA, including GlucaGen (glucagon recombinant for injection), Hylenex 
(hyaluronidase recombinant human), Hydase and Amphadase (hyaluronidase), Fortical 
(calcitonin salmon recombinant) Nasal Spray, and Omnitrope [somatropin 
(rDNA origin)] [15] . Further details related to the latter two are presented in the 
discussion of actual case studies. 
1.2.5 LEGAL ARGUMENTS RELATED TO FOLLOW - ON PROTEINS 
The legal arguments regarding the approval of biogenerics relate to several different 
aspects of drug/biologics law. 
1.2.5.1 Constitutionality of 505(b)(2) Process for Drugs 
The agency ’ s authority to grant approval of drugs via the 505(b)(2) process has 
previously been challenged by several companies. The nature of these challenges 
has questioned the FDA ’ s right to use proprietary information of the innovator in 
support of another company ’ s drug approval. Recall that the 505(b)(2) process 
allows a company to use data for which it does not have right of reference (i.e., 
another company ’ s safety and effi cacy data) in support of its own application. The 
FDA ’ s long - standing interpretation of the statute seems fi rm and well founded in 
precedent since over 80 applications for drugs have been approved via the 505(b)(2) 
route since its inception with indications ranging from cancer pain to Attention 
Defi cit Disorder (ADD) [16] . 
1.2.5.2 Constitutionality of 505(b)(2) Process for Follow - On Proteins 
The constitutionality issues related to FOPs are similar to those mentioned above 
for drugs, namely protection of proprietary information and intellectual property 
rights. Some critics opine that issues unique to FOPs create additional legal hurdles. 
For example, the rules pertaining to the disclosure of safety and effectiveness information 
are different for biologics licensed under the PHSA and drugs approved 
under the FDCA. When the rules were originally written (1974), it was thought that 
safety and effectiveness for one biologic would not support the licensure of another. 
So these data were deemed not to be protected trade secrets and could be publicly 
disclosed immediately after issuance of the biologic ’ s license [see 21 CFR 601.51(e), 
1974]. However, since this language applies strictly to the PHSA, it has no bearing 
on discussions related to the 505(b)(2) process. 
In other public challenges, opponents argue that the unique and complex nature 
of biologics and the close relationship between their method of preparation and 
clinical attributes require that the FDA use and disclose the manufacturing methods 
LEGAL ARGUMENTS RELATED TO FOLLOW-ON PROTEINS 43

44 REGULATORY CONSIDERATIONS IN APPROVAL 
and process information contained in an innovator ’ s application. Further, this use 
and disclosure would violate Trade Secret and Constitutional Law (Fifth Amendment 
“ taking clause ” ) [17, 18] . 
The concept of “ the product is the process ” may have been applicable to early 
biologics, but current capabilities allow the chemical, biologic, and functional comparison 
of well - characterized protein drugs. The follow - on manufacturer need not 
necessarily utilize the identical method of manufacture or proprietary technology 
to reproduce a follow - on biologic with similar clinical safety and effi cacy. Additionally, 
it is important to distinguish between the regulatory requirements for approval 
of an actual generic protein (duplicate of innovator; see discussion below) and those 
associated with a 505(b)(2), which requires a showing of similarity between two 
products. Any differences between the two would need to be adequately supported 
by bridging studies and appropriate clinical and/or nonclinical data. 
The FDA has confi rmed this interpretation in its response to petitions fi led 
regarding FOPs (both in general and targeted to specifi c applications). The FDA 
has clearly said, “ the use of the 505(b)(2) pathway does not entail disclosure of trade 
secret or confi dential commercial information, nor does it involve unauthorized 
reliance on such data ” [18] . 
1.2.5.3 Applicability of 505(j)(1) or ANDA Process to Biogenerics 
Biogenerics per se, that is, protein drug products approved via 505(j)(1), would need 
to demonstrate their bioequivalence to the innovator protein. However, due to their 
complexity and heterogeneity, the classical biopharmaceutical principles upon which 
the current ratings of therapeutic equivalence are based do not apply in their current 
language to complex macromolecules. For example, due to the nature and complexity 
of an immunogenic response, one concern would be if traditional bioequivalence 
appropriately addresses the complex safety issues associated with biologics. 
1.2.5.4 Current Rules Relating to Bioequivalence of Generic Drugs 
The list of approved drug products with therapeutic equivalence (Orange Book) 
was originally intended as an information source to states seeking formulary guidance 
[19] . The list provides the FDA ’ s recommendations as to which generic prescription 
drug products are acceptable substitutes for innovator drugs. The term 
innovator is used to describe the reference listed drug, or RLD [21 CFR 314.94(a)(3)], 
upon which an applicant (generic) relies in seeking approval of its ANDA. In layman 
’ s terms the RLD describes the original NDA - approved drug and is often 
referred to as the “ pioneer ” drug. 
Under the Drug Price Competition and Patent Term Restoration Act of 1984, 
manufacturers seeking approval to market a generic drug need to submit data to 
the FDA demonstrating that their proposed drug product is bioequivalent to the 
pioneer (innovator) drug product. A major premise underlying the 1984 law is that 
bioequivalent drug products are therapeutically equivalent, will produce the same 
clinical effect and safety profi le as the innovator product, and are therefore, interchangeable 
[19] . 
So how would FOPs be classifi ed using conventional defi nitions of bioequivalence? 
To answer this question, it is necessary to review current legal defi nitions of 
bioequivalence terms [19] : 

• Two products are bioequivalent in “ the absence of a signifi cant difference in the 
rate and extent to which the active ingredient or active moiety in pharmaceutical 
equivalents or pharmaceutical alternatives becomes available at the site of drug 
action when administered at the same molar dose under similar conditions in an 
appropriately designed study ” [21 CFR 320.1(e)]. An appropriately designed 
comparison could include (1) pharmacokinetic (PK) studies, (2) pharmacodynamic 
(PD) studies, (3) comparative clinical trials, and/or (4) in vitro studies. 
• Pharmaceutical equivalents are those drug products which are formulated to 
contain the same amount of active ingredient in the same dosage form to meet 
the same (compendial or other applicable) standards of quality. 
• Pharmaceutical alternatives are drug products that contain the same therapeutic 
moiety, or its precursor, but not necessarily in the same amount or dosage form. 
Drug products are considered to be therapeutic equivalents only if they are 
pharmaceutical equivalents and if they can be expected to have the same clinical 
effect and safety profi le when administered to patients under the conditions 
specifi ed in the labeling. Although pharmaceutical alternatives may ultimately 
be proven bioequivalent, given their differences they are not automatically 
presumed to be. 
Given these defi nitions, FOPs would likely be considered pharmaceutical alternatives 
if one presumes that pioneer and follow - on proteins are identical at a precursor 
stage, prior to potential post - translational modifi cation. This presumption may also 
be consistent with the similarity standard the agency applies to ascertain orphan 
drug status (see discussion in Section 1.2.6 ). Follow - on proteins cannot be considered 
to be therapeutic equivalents since they are not pharmaceutical equivalents 
and cannot be expected to have the same clinical effect and safety profi le in the 
absence of testing. This assertion is supported by the following: 
• The potential impact of how posttranslational modifi cations, such as glycosylation, 
can directly impact protein conformation and subsequently affect biological 
activity, including the overall safety and effi cacy of the drug product. 
• An underlying premise of bioequivalence assessments is a clearly defi ned pharmacokinetic/
pharmacodynamic relationship; however, the relation between 
blood levels and effect is less clearly established for proteins [20] . 
Consequently, within the current regulatory framework, FOPs are unique products 
that may be “ similar ” but are not the same as innovator proteins, consistent with 
their approval via a 505(b)(2) pathway. This interpretation is supported by the 
FDA ’ s designation of Omnitrope as having a BX rating in the Orange Book. The 
code BX in the Orange Book refers to drug products for which the data are insuf- 
fi cient to determine therapeutic equivalence as compared to a therapeutic rating of 
A indicative of interchangeability. This concept of similarity is also consistent with 
the defi nitions proposed by the European Agency for the Evaluation of Medicinal 
Products (EMEA) for generic versions of proteins [21] : 
Bio - similar products: second and subsequent versions of biologics that are independently 
developed and approved after a pioneer has developed an original version. 
Bio - similar products may or may not be intended to be molecular copies of the innovator 
’ s product; however, they rely on the same mechanism of action and therapeutic 
indication. 
LEGAL ARGUMENTS RELATED TO FOLLOW-ON PROTEINS 45

46 REGULATORY CONSIDERATIONS IN APPROVAL 
1.2.5.5 Statutory Authority 
Unlike the FDCA, which affords therapeutic protein drugs the legal pathway of 
abbreviated drug approval for a FOP, the PHSA currently has no similar provisions. 
Such a pathway for approval or licensure of FOP products under the PHSA would 
require new legislation and recent congressional developments suggest that work is 
underway to create this statutory pathway. 
Legislation proposed on September 29, 2006, by U.S. Representative Henry 
Waxman (D - CA) and Senator Charles Schumer (D - NY) seeks to amend the PHSA 
to authorize the FDA to approve abbreviated applications for biologic products that 
are “ comparable ” to previously approved (brand name) biologic products. Entitled 
The Access to Life - Saving Medicine Act, this bill outlines a process by which the FDA 
could determine, on a product - by - product basis, the studies necessary to demonstrate 
comparability of a FOP product to a brand name product and assure its safety and 
effectiveness. The act allows for an applicant to seek interchangeability with a brand 
name product, recognizing that the extent of data to support such a designation must 
be discussed with the FDA. To encourage the development of interchangeable products, 
the bill would authorize tax incentives and periods of marketing exclusivity. The 
bill would also seek to create an improved process to facilitate early resolution of 
patent disputes which might otherwise delay competition [22] . 
1.2.6 SCIENTIFIC ISSUES RELATED TO FOLLOW - ON PROTEINS 
(DATA REQUIREMENTS) 
The challenge of FOPs demonstrating similar quality, safety, and effi cacy to the 
innovator product relates to the poor predictability of physicochemical characteristics 
and biologic activity. For example, there are several different interferon - . and 
erythropoietin . and . products currently on the market. These variants are characterized 
by differences in sequence, glycosylation pattern, and in vitro measures of 
specifi c activity; however, their clinical safety and effi cacy profi les are considered 
similar [20] . 
In contrast, different formulations of insulin and growth hormone containing the 
same active ingredient exhibit signifi cant differences in bioavailability [20] . Additionally, 
the inability to adequately predict immunogenic responses from in vitro 
data or animal studies remains a concern. 
The answer to the challenge is that generic manufacturers must go through a 
similar process of in - depth characterization, including identifi cation of critical structural 
elements of the product (structure/function) when developing a FOP. Although 
the regulatory standards for demonstrating similarity are currently undefi ned, some 
insight can be gleaned from consideration of FDA expectations in terms of granting 
orphan drug status to similar proteins and assessing postapproval Chemistry, Manufacturing 
and Controls (CMC) changes for innovator proteins. 
1.2.6.1 “ Sameness ” as per Orphan Drugs Regulations 
The Orphan Drug Act of 1983 was implemented in response to the government ’ s 
concern that viable treatments for rare diseases were not being explored due to 
excessive costs of drug development in comparison to the relatively small popula

tion of potential users (and sales). Orphan drugs are (a) those used to treat rare 
diseases, defi ned by the act as affecting < 200,000 persons in the United States, or 
(b) those drugs whose development costs would not be recovered through sales of 
the drug. To encourage development, the government authorized incentives in the 
form of marketing exclusivity (seven years), tax credits, protocol assistance, and 
grants/contracts, with the fi rst being of primary importance to most drug sponsors. 
Since exclusivity is awarded only to the fi rst designated product to obtain approval 
for a given drug/indication, competition is fi erce. No approval would be given to a 
subsequent sponsor ’ s application for the same product/indication unless it was 
shown to be clinically superior (i.e., not the same). Thus, the agency needed to 
develop criteria upon which it would make these determinations. 
In 1992, the FDA ’ s orphan drug regulations fi rst established the conditions under 
which the agency could determine product “ sameness ” of protein drugs and therefore 
take action to block the approval of a second orphan drug product: “ two protein 
drugs would be considered the same if the only differences in structure between 
them were due to post - translational events, or infi delity of translation or transcription, 
or were minor differences in amino acid sequence; other potentially important 
differences, such as different glycosylation patterns or different tertiary structures, 
would not cause the drugs to be considered different unless the differences were 
shown to be clinically superior ” [23] . It should be noted that there may exist exceptions 
to this rule that depend on the interpretation of each individual case. For 
example, Eli Lilly & Co. successfully received orphan drug status in the late 1980s 
for the naturally occurring HGH to compete with the previously marketed Met - 
HGH, which only differed in the N - terminal methionine. 
The support for clinical superiority could be based on evidence of greater effectiveness 
and increased safety or represent a “ major contribution to patient care. ” In 
short, orphan drug regulations utilize clinical data to demonstrate product differences. 
Examples include [23] : 
• 1996: Biogen ’ s Avonex (interferon . ) was considered to be clinically superior to 
Berlex ’ s Betaseron based on improved safety (fewer site injection reactions). 
• 1999: In a law suit involving generic paclitaxel, Baker Norton, challenged the 
FDA ’ s sameness determinations based on active moiety alone, arguing that 
factors such as formulation and labeling should be considered. The challenge 
was unsuccessful. 
• 2002: Serono ’ s Rebif (interferon .1a ) was awarded exclusivity based on the 
clinical demonstration of improved effi cacy (reduced Multiple Sclerosis (MS) 
exacerbations). 
Therefore, it would appear that the orphan drug regulations provide some fl exibility 
to the sponsor (generic) in establishing product sameness but also reaffi rm the 
important role of clinical data in supporting product safety and effi cacy. 
1.2.6.2 “ Sameness ” as per Postapproval Change Guidances 
Guidelines for supporting postapproval changes to the chemistry, manufacturing, 
and controls of approved products (SUPAC guidances) take a somewhat different 
approach to establishing sameness. In essence, the SUPAC guidelines refl ect risk 
SCIENTIFIC ISSUES RELATED TO FOLLOW-ON PROTEINS 47

48 REGULATORY CONSIDERATIONS IN APPROVAL 
management practices in evaluating the potential of certain CMC changes to impact 
the identity, strength, quality, purity, and potency of the product as they may relate 
to overall safety and effi cacy. 
A long - held contention within the biologics industry is that the product is the 
process and, by extension, change is strongly discouraged. Without qualifi cation, this 
rather dated thinking is inconsistent with the fl exibility required in managing change 
throughout the life cycle of a product. Further, this thinking may serve to discourage 
the implementation of advanced technologies designed to improve not only effi - 
ciency but also product quality. Even current biologics regulations recognize 
the need to accommodate change; 21 CFR 601.12 (for biologics) states that for 
changes in the product, production process, quality controls, equipment, facilities, 
and so on, an applicant must assess the effects of the change and demonstrate 
through appropriate validation and/or other clinical and/or nonclinical laboratory 
studies the lack of adverse effect of the change on the identity, strength, quality, 
purity, or potency of the product as they may relate to the safety or effectiveness of 
the product. 
In fact, many of the challenges that generic manufacturers face in demonstrating 
sameness of FOPs to reference listed drugs are similar to those encountered by 
innovators in managing the dynamic CMC life cycle of a product. One of the tools 
available to assess the potential impact of product differences is a comparability 
protocol. The FDA described its expectations of the data requirements necessary 
to support postapproval CMC changes to protein drug product and biologic products 
in a Guidance to Industry on the use of comparability protocols for such products 
issued in 2003 [24] . Underpinning the successful application of a comparability 
protocol are extensive product development and characterization. 
Initial Product Development Prior to undertaking any comparative analysis, a 
manufacturer must perform two critical steps. First, the manufacturer needs to 
conduct thorough process development and optimization of the therapeutic protein 
product. Second, the sponsor (generic or innovator) needs to prospectively examine 
the impact of changes to all critical processing parameters during the development 
phase and determine the minimum data requirements necessary to assure the 
absence of adverse impact to product quality, safety, or effi cacy. The current state 
of technology provides us with better tools to more fully characterize the protein 
drug substance and drug product at all stages of production. 
Physicochemical Characterization and Process Development Some of the key steps 
to process development and product characterization include: 
• Production of a cell line/clone 
• Identifi cation and characterization of critical raw materials (media, resins, 
formulation excipients) 
• Development of internal standards, in - process controls, product 
specifi cations 
• Conduct of extensive pilot - scale manufacturing development: fermentation and 
downstream processing (separation and purifi cation) 
• Performance of process scale - up and optimization studies 

• Application of a comprehensive array of analytical techniques to fully characterize 
the drug product at each stage of development. Table 6 provides examples 
of methods to probe virtually every property of the protein and develop 
a fi ngerprint of the molecule. 
Other Testing Requirements The need for additional supportive studies beyond 
physicochemical characterization will increase proportionately with the complexity 
of the protein drug. The entire battery of tests may not be required for each FOP 
but may include the following data, bioassay, preclinical (pharmacology/toxicology/ 
pharmacokinetic/pharmacodynamic), clinical safety and effi cacy, and immunogenicity. 
The nature, number, and size of the trials should relate directly to the particular 
drug/indication/patient population. 
Bioassay A biological assay, or “ bioassay, ” is an analytical procedure capable of 
measuring the biologic activity of a substance based on a specifi c functional, biologic 
TABLE 6 Analytical Techniques for Physicochemical Characterization of Proteins 
Parameter Test 
Primary structure Amino acid sequencing, N - terminal Edman 
sequencing, peptide mapping 
Higher order structure CD, NMR, FTIR, Raman 
Mass LC - ESI - MS, MALDI - TOF - MS 
Size SDS - PAGE, DLS, SEC - MALLS 
Hydrophobicity RP - HPLC 
Binding Immunological binding 
Sulfhydryl groups/disulfi de bridges Peptide mapping (under reducing and nonreducing 
conditions) 
Glycan analysis: 
Monosaccharide analysis HPLC, MS 
Sialic acid content HPLC 
Molecular weight MALDI - MS, ESI - MS 
Impurity profi le 
Process - related impurities Immunoassay, HPLC, SDS - PAGE, MS, CD, capillary 
gel electrophoresis, size exclusion chromatography • Cell substrate derived 
• Cell culture derived 
• Downstream derived 
Product - related impurities 
• Truncated forms 
• Other modifi ed forms (i.e., 
deamidated, isomerized) 
• Aggregates 
Evaluation of stability HPLC 
CD, Circular Dichroism; NMR, Nuclear Magnetic Resonance; FTIR, Fourier transform infrared spectroscopy; 
LC - ESI - MS, Liquid chromatography electrospray ionisation mass spectrometry; MALDI - 
TOF - MS, Matrix - assisted laser desorption ionization - time of fl ight - mass spectrometry; SDS - PAGE, 
Sodium dodecyl sulfate (SDS) polyacrylamide gel electrophoresis; DLS. Dynamic light scattering; SEC - 
MALLS, Size exclusion chromatography - multi - angle laser light scattering; RP - HPLC, Reversed phase - 
high performance liquid chromatography; HPLC, High performance liquid chromatography; MALDI - MS, 
Matrix - assisted laser desorption ionization mass spectrometry; ESI - MS, Electrospray ionisation mass 
spectrometry; MS, Mass spectrometry. 
SCIENTIFIC ISSUES RELATED TO FOLLOW-ON PROTEINS 49

50 REGULATORY CONSIDERATIONS IN APPROVAL 
response of the test system. Bioassays should be predictive of clinical effect and are 
therefore used as a means of quantifying activity (in nonclinical manner) and ensuring 
effi cacy throughout development. They are informative in equivalence studies 
to the extent that a change affects a part of the molecule, which in turn impacts the 
molecule ’ s biologic activity. Bioassays may be based on animal models, in vitro cell 
lines, cell - based biochemical assays (i.e., kinase receptor activity), receptor binding 
assays, or enzyme assays. The selection of an appropriate bioassay is driven in part 
by the ability to demonstrate a correlation to clinical effect. An example of a predictive 
bioassay is the measurement of the antiviral activity of interferon as a function 
of its cytopathic effect on host cells [25] . 
Nonclinical (Pharmtox, PK , PD ) In the context of FOPs, the original sponsor will 
have demonstrated what the molecule per se does to the body; however, since the 
formulation is likely different, nonclinical studies are useful in demonstrating a lack 
of adverse impact due to dosage form, route of administration, excipient changes, 
manufacturing contaminants, and supporting sameness of the active moiety. 
Appropriate toxicology studies would include acute or subchronic testing in at 
least one relevant small animal species. Pharmacokinetic studies are highly useful 
in assessing the impact of changes in the manufacture of natural - source - and 
recombinant - derived proteins. Standard approaches used in bioequivalence studies 
[measurement of the area under the curve (AUC), Cmax, tmax ] can be used to make 
direct comparisons of innovator and follow - on profi les. Pharmacodynamic studies 
are similarly very informative. Direct comparison of innovator and follow - on products 
can be made by evaluating appropriate surrogate markers of effi cacy (i.e., 
platelet aggregation following anticoagulation therapy). 
Clinical ( PK , PD , Safety and Effi cacy) Human clinical studies can range in complexity 
from standard - design PK studies to complicated, long - term effi cacy trials 
evaluating one or more indications in multiple populations. Human PK studies are 
used as the benchmark for establishing bioequivalence of conventional dosage 
forms. For traditional pharmaceuticals for which reliance on systemic exposure may 
not be suitable, PD or clinical safety and effi cacy may be performed to show 
equivalence. 
The appropriate clinical program is infl uenced by many factors, including the 
degree of molecular complexity of the particular protein and the extent of physicochemical 
characterization; the mode of action, indication(s), and use population(s); 
the presence of established structure – activity relationships and validated bioassays; 
and the results of preclinical testing. 
As such, the nature and scope of each clinical support program need to be determined 
on a case - by - case basis in consultation between the sponsor and the regulatory 
agency. 
Immunogenicity The observation of serious adverse events with the use of some 
recombinant and natural source proteins [i.e., pure red cell aplasia (PRCA) detected 
with erythropoietin use] has highlighted immunogenicity as a major issue for consideration 
when assessing within and between manufacturer changes [8] . Although the 
exact immunological mechanism responsible for the increased number of PRCA 
cases is unknown, it appears to be linked to a formulation change associated with 
Eprex, a European epoetin - . product. Replacement of the stabilizer human serum 

albumin with polysorbate 80 and glycine correlated with a surge in PRCA reported 
cases [4] . An immunogenic effect may have no clinical impact or it could have serious 
clinical consequences as seen above. The immune response of the therapeutic protein 
should be fully characterized using both immunoassays which detect antibodies that 
bind to the drug as well as bioassays which detect neutralizing antibodies that might 
block the protein ’ s desired biological effect. Ultimately, this testing needs to be performed 
in humans, as animal testing is not truly predictive of human immune response. 
Antibody detection techniques include enzyme - linked immunosorbent assay radioimmunoassay, 
(ELISA), and surface plasmon resonance [8] . 
Comparability Testing to Demonstrate “Sameness” Following the developmental 
studies described above, comparative studies to directly evaluate pre - and postchange 
materials to one another and assess the impact of any process changes may 
be conducted. In a similar manner, comparative studies between pioneer drug and 
the follow - on can be used to systematically evaluate the impact of any differences 
between reference listed drug and proposed generic protein drug. When compiling 
information into an analytical characterization database, the data should be directly 
compared to the reference product and variation observed in multiple batches 
of test product (generic) should be similar to that of the reference innovator 
product. 
The FDA ’ s expectations in this regard are apparent in their description of the 
CMC data package supporting the comparability of Omnitrope to the innovator 
protein Genotropin. The FDA asserted [18] : 
Each biotechnology manufacturer, whether producing a new molecular entity or a 
follow - on product must independently develop its own cell expression, fermentation, 
isolation and purifi cation systems for the active ingredient in its product. Thus, the 
manufacturing process for each active ingredient is unique to each manufacturer. Nevertheless, 
as Sandoz has demonstrated in its Omnitrope application, for this relatively 
simple recombinant protein, it is possible to determine that the end products of different 
manufacturing processes are highly similar, without having to compare or otherwise 
refer to the [proprietary] processes. 
1.2.7 PROPOSED REGULATORY PARADIGM: CASE STUDIES 
Based on the nature and complexity of therapeutic protein products, an approval 
pathway for follow - ons may require moving away from the traditional generic paradigm 
in place for small molecules and creating a biosimilar paradigm for complex 
molecules. The proposed regulatory paradigm for the approval of FOP products 
could be similar for protein drugs approved under Section 505 of the FDCA or 
licensed as biologics under the PHSA and mirror the current 505(b)(2) process. This 
pathway permits the sponsor and agency to determine exactly what studies are 
necessary to support the proposed differences (see 21 CFR 314.54(a) [ “ a 505(b)(2) 
application need contain only that information needed to support the modifi cation(s) 
of the listed drug ” ]. Application of a 505(b)(2) paradigm removes the need to demonstrate 
bioequivalence per se and potentially reduces innovator intellectual property 
concerns that arise if a generic must “ duplicate ” the innovator. Guidance as to 
how similar a “ biosimilar ” needs to be exists in the form of current regulations 
related to orphan drugs and postapproval manufacturing changes. 
PROPOSED REGULATORY PARADIGM: CASE STUDIES 51

52 REGULATORY CONSIDERATIONS IN APPROVAL 
Several recent drug approvals illustrate how this regulatory framework may be 
applied and are described in the sections to follow. 
1.2.7.1 Case Study 1: Fortical [Calcitonin - Salmon ( r DNA origin)] 
On August 17, 1995, the FDA approved Novartis ’ s NDA for Miacalcin (calcitonin - 
salmon) Nasal Spray (Miacalcin NS) for the treatment of postmenopausal osteoporosis 
in females greater than fi ve years postmenopause with low bone mass relative 
to healthy premenopausal females. The active ingredient in Miacalcin NS is synthetic 
salmon calcitonin. On March 6, 2003, Unigene submitted a new drug application 
under Section 505(b)(2) for Fortical [calcitonin - salmon (rDNA origin)] Nasal 
Spray which relied in part on data submitted in the Miacalcin NS NDA. 
Comparability Program Fortical and Miacalcin NS differed in certain aspects, 
such as the use of recombinant versus synthetic salmon calcitonin and the use of 
different types and amounts of excipients. Given these differences, Unigene was 
required to submit data to establish that the fi ndings of safety and effi cacy for Miacalcin 
were relevant to Fortical (i.e., contain the same active ingredient and have 
comparable bioavailability) and that the formulation differences did not impact 
previous clinical profi le [26, 27] . 
Comparability Results 
Physicochemical Analysis Salmon calcitonin is a 32 - amino - acid, nonglycosylated 
peptide hormone. It is structurally simple, possessing limited secondary structure 
and a single disulfi de bond. The physicochemical characterization studies demonstrated 
that the primary and secondary structure of Fortical ’ s recombinant salmon 
calcitonin (sc) was identical to that of Miacalcin ’ s synthetic sc or naturally occurring 
sc. Further, the tertiary structures of the three were indistinguishable. 
Nonclinical PK / Tox The pharmacokinetic profi le of Fortical by different routes of 
administration was compared to Miacalcin, demonstrating similarity in PK profi les 
between the synthetic and recombinant peptides and toxicity results (28 - day rat 
intranasal toxicity study) were acceptable, particularly in light of clinical safety 
data. 
Clinical PK / PD Calcitonin has a well - established mechanism of action; published 
literature supports that salmon calcitonin, mediated through calcitonin receptors 
located on osteoclasts, inhibits bone resorption, thereby increasing bone mineral 
density. Since serum beta - CTx (C - telopeptides of type 1 collagen, corrected for 
creatinine) is a recognized marker of bone resorption, the effect of administered 
salmon calcitonin on serum beta - CTx is considered to be an adequate surrogate for 
pharmacodynamic comparisons. 
Fortical ’ s PD equivalence was shown in a double - blind, active - controlled, 24 - 
week study in 134 postmenopausal women randomized to Fortical (200 IU per day) 
or Miacalcin (200 IU per day). The primary outcome measure was change in serum 
beta - CTx from baseline. The results fell within prespecifi ed PD equivalence limits 
(. 0.08 to 0.06 ng/mL; equivalence margin of ± 0.2 ng.mL) and indicated Fortical was 
not inferior to Miacalcin. 

Fortical ’ s PK equivalence was assessed by comparing the relative bioavailability 
of Fortical to Miacalcin in a multidose, crossover study of 47 healthy female volunteers. 
Results indicated that Fortical was slightly more bioavailable than Miacalcin, 
but given the demonstration of similar PD activity, this difference were not considered 
to be clinically signifi cant. 
Immunogenicity Archived samples from the 24 - week PD study were used to 
compare the immunogenicity potential of both products. The results indicated there 
was no difference in terms of total immune response and the response of neutralizing 
antibodies between the two drugs. 
Conclusion to Case Study 1 On August 12, 2005, the FDA approved Unigene ’ s 
505(b)(2) application for Fortical for the same indication as Miacalcin NS [26, 27] . 
In the FDA ’ s analysis no statistically and/or clinically signifi cant differences were 
noted in any aspect of the comparability profi le, including clinical performance, and 
Fortical was approved. 
The basis of this comparison was strongly challenged in a citizen petition claiming 
that (1) recombinant salmon calcitonin is not the same as the synthetic version 
which could potentially cause differences in product effi cacy, safety, or both and 
(2) only a long - term clinical study (actual bone fracture data) would provide adequate 
support of sameness [28] . The FDA responded to this citizen petition by 
asserting its decision that the comparability data presented above collectively constituted 
suffi cient demonstration of sameness [27, 29] . 
1.2.7.2 Case Study 2: Omnitrope [Somatropin ( r DNA origin)] 
On August 24, 1995, the FDA approved NDA20 - 280 fi led by the Pharmacia & 
Upjohn Company for Genotropin (somatropin) (rDNA origin) for injection. Since 
that time, Genotropin has been marketed as a safe and effective therapy for growth 
hormone defi ciency (GHD) in children and adults. 
On July 30, 2003, Sandoz submitted a 505(b)(2) application for the approval of 
its recombinant HGH product (recombinant somatropin) indicated for long - term 
treatment of pediatric patients who have growth failure due to an inadequate secretion 
of endogenous growth hormone and for long - term replacement therapy in 
adults with GHD of either childhood or adult onset. This application relied in part 
on data submitted in the Genotropin NDA. 
Comparability Program As with the Fortical case study, Omnitrope and Genotropin 
differed in certain aspects. As such, Sandoz was required to submit substantial 
data to establish that Omnitrope was suffi ciently similar to Genotropin to warrant 
reliance on FDA ’ s fi nding of safety and effectiveness for Genotropin to support the 
approval of Omnitrope [18] . 
Comparability Results 
Physicochemical Analysis In terms of complexity, HGH is fairly simple and well - 
characterized. Human growth hormone is a single - chain, 191 - amino - acid, nonglycosylated 
protein with two intramolecular disulfi de bonds. Sandoz used a variety of 
physicochemical tests and analytical methods to confi rm the primary, secondary, and 
PROPOSED REGULATORY PARADIGM: CASE STUDIES 53

54 REGULATORY CONSIDERATIONS IN APPROVAL 
tertiary structures, molecular weight, and impurity profi le. Characterization studies 
performed to verify somatropin as the active ingredient in Omnitrope included 
reverse - phase liquid chromatography/mass spetrometry (RP - HPLC/MS), DNA 
sequencing, N - terminal and C - terminal sequencing, peptide mapping, circular 
dichroism (CD) analysis, UV spectroscopy, one - dimensional nuclear magnetic 
resonance spectroscopy (1D NMR), two - dimensional (2D) NMR, size exclusion 
chromatography (SEC), isoelectric focusing (IEF), sodium dodecyl sulfate polyacrylamide 
gel electrophoresis (SDS – PAGE), and capillary zone electrophoresis. 
Nonclinical PK / Tox Minimal toxicity data were needed on recombinant HGH 
(rHGH) itself, since the clinical effects of HGH excess are well established and 
understood and are extensively documented in published literature. Sandoz performed 
toxicity studies to appropriately qualify impurities specifi c to Omnitrope, 
that is, a subacute 14 - day rat study and a local (skin) tolerance study in rabbits. 
Further, the bioactivity of Omnitrope was assessed using a validated weight gain 
bioassay using a hypophysectomized (growth - hormone - defi cient) rats. 
Clinical PK / PD HGH has a well - established mechanism of action. Omnitrope was 
demonstrated to be pharmacokinetically and pharmacodynamically “ highly similar ” 
to Genotropin. The dataset comprised a total of three PK/PD studies, including a 
double - blind, randomized, two - way crossover study comparing Omnitrope and 
Genotropin. Additionally, Sandoz conducted three sequential, multicenter phase 3 
pivotal trials in 89 pediatric patients with GHD providing data in some patients for 
up to 30 months. A fourth phase 3 trial ( n = 51, 24 months) was submitted as part 
of its safety update. Collectively, these data in conjunction with the demonstrated 
comparability to the reference listed product provide substantial evidence of Omnitrope 
’ s safety and effectiveness. 
Immunogenicity A signifi cant number of patients who were administered an 
earlier version Omnitrope developed anti – growth hormone antibodies during the 
fi rst and second phase 3 clinical trials. In response, Sandoz implemented changes to 
the drug product to address this immunogenicity and evaluated the impact of these 
changes clinically. Data from the 24 - month clinical study demonstrated that Omnitrope 
has a low and acceptable level of immunogenicity (comparable to other 
rHGH products) as none of the patients developed anti – growth hormone antibodies 
during the duration of the study and only one patient developed anti – host cell 
protein antibodies, which were of no detectable clinical consequence. 
Conclusion to Case Study 2 This case provoked signifi cant challenges from interested 
parties voiced via several citizen petitions [18] . Furthermore, the FDA ’ s delay 
in approval prompted Sandoz to fi le suit to compel the FDA to rule on its application. 
On April 10, 2006, the Washington, D.C., District Court ruled that the FDA 
must meet its statutory obligations and take action on Sandoz ’ s outstanding NDA [6] . 
On May 30, 2006, the FDA approved Omnitrope [somatropin (rDNA origin)] as a 
“ follow - on protein product ” for use in the treatment of pediatric GHD. At the same 
time, the FDA responded to the related citizen petitions and defended its position 
that the data were adequate to demonstrate that Omnitorpe was suffi ciently similar 
to Genotropin to enable reliance on the agency ’ s previous fi ndings of safety and 

effi cacy for Genotropin. These data, in conjunction with the independent evidence 
of safety and effi cacy provided by Sandoz, supported Omnitrope ’ s approval. 
1.2.7.3 Case Study 3: Generic Salmon Calcitonin 
On February 17, 2004, Nastech Pharmaceutical Company announced its fi ling of an 
ANDA for a salmon calcitonin nasal spray drug product for the treatment of postmenopausal 
osteoporosis. As with Fortical, Novartis ’ s Miacalcin was cited as the 
reference listed drug; however, Nastech chose to submit an ANDA via the 505(j)(1) 
route, rather than a 505(b)(2) application. 
The distinction between the two regulatory routes has signifi cant implications for 
FOPs. Whereas 505(b)(2) allows products to be “ suffi ciently similar, ” an ANDA 
requires the applicant establish “ sameness ” of the active ingredients. The scope of 
data necessary to demonstrate that the actives are the same is unclear. Additionally, 
use of the ANDA route is appropriate for circumstances in which “ clinical studies 
are not necessary to show safety and effectiveness. ” If clinical data are required as 
proof of sameness, as in the previous example where clinical data were used to 
demonstrate comparable immunogenicity, then the ANDA route may not represent 
a viable regulatory path. 
On July 10, 2006, Nastech was notifi ed by the FDA that its ANDA for 
intranasal calcitonin salmon was not approvable at present based on concerns 
relating to the potential for immunogenicity that might result from a possible 
interaction between calcitonin salmon and chlorobutanol, the preservative in the 
formulation. 
Nastech has indicated it will continue to work with the agency to understand the 
data requirements and regulatory options, but the fi nal resolution remains presently 
unknown. This case study highlights the fact that demonstration of sameness of 
therapeutic proteins is more complex than for other drugs and that true “ biogenerics 
” may be hard to come by due to the complexity in establishing sameness versus 
similarity. 
1.2.8 SUMMARY AND CONCLUSIONS 
This chapter provides an overview of the complex scientifi c, legal, and policy issues 
facing the development of biogenerics today. Given the rising cost of health care 
and prescription medications in this country and the pivotal and expanding role of 
biologically derived products within the pharmaceutical landscape, these issues 
present a challenge to industry, regulators, and legislators alike. Substantial progress 
has already been made and the regulatory climate continues to evolve in response 
to advancing science and technology. Recent FDA approvals provide insight into 
the technical requirements for approval of well - characterized FOP products. They 
also demonstrate the appropriate use of an abbreviated approval pathway, that is, 
the 505(b)(2) pathway in place for drugs approved under the FDCA. Importantly, 
recent legislative proposals seek to amend the PHSA to eliminate the current legal 
barriers which prohibit abbreviated approval of protein biologics. This legislation 
reaffi rms the need for the FDA to determine on a case - by - case basis the nature and 
extent of supporting data required for a given product. 
SUMMARY AND CONCLUSIONS 55

56 REGULATORY CONSIDERATIONS IN APPROVAL 
REFERENCES 
1. Congressional Budget Offi ce ( 1998 ), How Increased Competition from Generic Drugs 
Has affected Prices and Returns in the Pharmaceutical Industry , Congressional Budge 
Offi ce , Washington, DC . 
2. Crawford , L. M. , Acting Commissioner of the Food and Drug Administration, in a Speech 
to the Generic Pharmaceutical Association on February 26, 2005, available: http://www. 
fda.gov/oc/speeches/2005/GPhA0301.html , accessed Apr. 23, 2005. 
3. Comments of the Generic Pharmaceutical Association (GPhA) (Sept. 29, 2006), available: 
http://www.gphaonline.org/AM/Template.cfm?Section=Media&Template=/CM/ 
HTMLDisplay.cfm&ContentID=2849 , accessed Jan. 23, 2007. 
4. Schellekens , H. ( 2005 ), Follow - on biologics: Challenges of the “ next generation ” , 
Nephrol. Dial. Transplant . 20 ( Suppl. 4 ), iv31 – iv36 . 
5. Congressional letter from Senators O. Hatch and H. Waxman to Andrew von Eschenbach, 
Acting Commissioner of the Food and Drug Administration (Feb. 10, 2006), available: 
http://www.henrywaxman.house.gov/news_letters_2006.htm , accessed Dec. 21, 2006. 
6. Messplay , G. C. , and Heisey , C. ( 2006 ), Follow - on biologics: The evolving regulatory landscape 
, Bioexec Int. , May, 42 – 45 . 
7. Biotechnol. Law Rept. , 2003 , 22(5), 485 – 508 . 
8. Comments from R. Williams, U.S. Pharmacopoeia (USP), to FDA Docket No. 2004N - 
0355, Mar. 15, 2005 . 
9. Herrera , S. ( 2004 ), Biogenerics standoff , Nat. Biotechnol. , 22 ( 11 ), 1343 – 1346 . 
10. Scott , S. R. ( 2004 ) What is a biologic ?, Chapter 1 in Mathieu , M. , Ed., Biologics Development: 
A Regulatory Overview , 3rd ed., Paraxel Intl. , Waltham, MA , pp. 1 – 16 . 
11. Federal Food Drug and Cosmetic Act , available: http://www.fda.gov/opacom/laws/fdcact/ 
fdctoc.htm , accessed Apr. 21, 2005. 
12. Public Health Service Act , available: http://www.fda.gov/opacom/laws/phsvcact/phsvcact. 
htm , accessed Apr. 21, 2005. 
13. U.S. Department of Health and Human Services, Food and Drug Administration Transfer 
of Therapeutic Products to the Center for Drug Evaluation and Research , available: 
http://www.fda.gov/cber/transfer/transfer.htm , accessed Apr. 23, 2005. 
14. U.S. Department of Health and Human Services (DHHS) ( 1999 , Oct.), Food and Drug 
Administration, Center for Drug Evaluation and Research , Guidance for Industry: 
Applications covered by Section 505(b)(2), DHHS, Washington, DC. 
15. U.S. Department of Health and Human Services, Food and Drug Administration, Omnitrope 
(somatropin [rDNA origin]) questions and answers, available: http://www.fda.gov/ 
cder/drug/infopage/somatropin/qa.htm , accessed Dec. 21, 2006. 
16. Letter of J. Woodcock. (CDER, FDA) to Docket Nos. 2001P - 0323/CP1, 2002P - 0447/CP1, 
and 2003P - 0408/CP1 (Oct. 14, 2003 ). 
17. Glidden , S. ( 2001 ), The generic industry going biologic , Biotechnol. Law Rept , 20 ( 2 ), 
172 – 181 . 
18. Letter from S. Galson (CDER, FDA) in response to Docket Nos. 2004P - 023 11CP1 and 
SUP 1,2003P - 0 1 76lCP 1 and EMC 1, 2004P - 0171lCP1 and 2004N - 0355 (May 30, 2006 ). 
19. U.S. Department of Health and Human Services, Food and Drug Administration, Center 
for Drug Evaluation and Research, Offi ce of Pharmaceutical Science, Offi ce of Generic 
Drugs , Electronic orange book: Approved drug products with therapeutic equivalence 
evaluations, available: http://www.fda.gov/cder/ob/default.htm , accessed Apr. 21, 2005. 
20. Schellekens , H. ( 2004 ), How similar do “ biosimilars ” need to be ? Nat. Biotechnol . 22 ( 11 ), 
1357 – 1359 . 

21. Webber , K. ( 2005 ), Relevant terminology. A presentation conducted at the Public Workshop 
on the Development of Follow - On Protein Products, Sept. 14, 2004, available: 
http://www.fda.gov/cder/meeting/followOn/followOnPresentations.htm , accessed Feb. 2, 
2005. 
22. Waxman , H. , Schumer , C. E. , and Clinton , H. R. (2006), Congress of the United States, 
H.R. 6257, Access to Life - Saving Medicine Act, ” available: http://www.waxman.house. 
gov/pdfs/bill_generic_biologics_9.29.06.pdf , accessed Sept. 29, 2006. 
23. Mathieu , M. , and Evans , A. G. ( 2005 ), The FDA ’ s Orphan Drug Development Program , 
in Ed., New Drug Development: A Regulatory Overview , 7th ed., Paraxel Intl. , Waltham, 
MA , pp. 307 – 317 . 
24. U.S. Department of Health and Human Services (DHHS) ( 2003 , Sept.), Food and Drug 
Administration, Center for Drug Evaluation and Research, Guidance for industry: Comparability 
protocols — Protein drug products and biological products — Chemistry, manufacturing 
and controls information , DHHS , Washington, DC . 
25. Beatrice , M. ( 2002 ), Regulatory considerations in the development of protein Pharmaceuticals 
, in Nail , S. , and Akers , M. , Eds., Development and Manufacture of Protein Pharmaceuticals, 
Pharmaceutical Biotechnology , Vol. 14, Kluwer Academic/Plenum ,New York , 
pp. 405 – 457 . 
26. Letter from R. Levy (Unigene) to FDA Docket No. 2004P - 0015 (Apr. 11, 2005 ). 
27. Letter from S. Galson (CDER, FDA) in response to Docket No. 2004P - 0015/CP1 (Aug. 
12, 2005 ). 
28. Letter from N. Buc to FDA Docket No. 2004P - 0115/CP1 (Jan. 9, 2004 ). 
29. FDA Week , 11(34), Aug. 26, 2005. 
REFERENCES 57


59 
1.3 
RADIOPHARMACEUTICAL 
MANUFACTURING 
Brit S. Farstad 1 and Iv a n Pe n uelas 2 
1 Institute for Energy Technology, Isotope Laboratories, Kjeller, Norway 
2 University of Navarra, Pamplona, Spain 
Contents 
1.3.1 Introduction 
1.3.1.1 Radiopharmacy 
1.3.1.2 Characteristics of Radiopharmaceuticals 
1.3.1.3 Ideal Characteristics of Radiopharmaceuticals 
1.3.1.4 Radioactive Decay 
1.3.1.5 Principles of Radiation Protection 
1.3.1.6 Detection Devices for Clinical Nuclear Imaging 
1.3.2 Product Development 
1.3.2.1 Radionuclides 
1.3.2.2 Carrier Molecules/Active Ingredients 
1.3.2.3 Radiolabeling Techniques 
1.3.2.4 Manufacturing Scale - Up 
1.3.2.5 Automation 
1.3.3 Manufacturing Aspects 
1.3.3.1 Design of Manufacturing Sites 
1.3.3.2 Design of Production Processes 
1.3.3.3 Design of Production Equipment 
1.3.3.4 Cleaning and Sanitation of Production Equipment 
1.3.3.5 Environmental Control 
1.3.3.6 Sterilization of Radiopharmaceuticals 
1.3.3.7 Starting Materials 
1.3.3.8 Labeling and Packaging 
1.3.4 Product Manufacturing 
1.3.4.1 Production of Radionuclides 
1.3.4.2 Production of Radiopharmaceuticals 
1.3.5 Quality Considerations 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

60 RADIOPHARMACEUTICAL MANUFACTURING 
1.3.5.1 Documentation 
1.3.5.2 Qualifi cation of Personnel 
1.3.5.3 Quality Control 
1.3.5.4 Validation and Control of Equipment and Procedures 
1.3.5.5 Stability Aspects of Radiopharmaceuticals 
1.3.6 Extemporaneous Preparation of Radiopharmaceuticals 
References 
Further Readings 
1.3.1 INTRODUCTION 
1.3.1.1 Radiopharmacy 
Radiopharmacy is a patient - oriented science that includes the scientifi c knowledge 
and professional judgment required to improve and promote health through assurance 
of the safe and effi cacious use of radiopharmaceuticals. Radiopharmacy encompasses 
studies related to the pharmaceutical, chemical, physical, biochemical, and 
biological aspects of radiopharmaceuticals. 
Radiopharmacy comprises a rational understanding of the design, preparation, 
and quality control of radiopharmaceuticals, the relationship between 
the physicochemical and biological properties of radiopharmaceuticals and 
their clinical applications, as well as radiopharmaceutical chemistry and issues 
related to the management, selection, storage, dispensing, and proper use of 
radiopharmaceuticals. 
1.3.1.2 Characteristics of Radiopharmaceuticals 
A radiopharmaceutical is any medicinal product which, when ready for use, contains 
one or more radionuclides (radioactive isotopes) included for a medicinal purpose. 
This generic defi nition of radiopharmaceutical thus includes both diagnostic and 
therapeutic radiopharmaceuticals. 
A radiopharmaceutical can be as simple as a radioactive element such 
as 133 Xe, a simple salt such as 131 INa, a small labeled molecule such as 
l - ( S - [ 11 C]methyl)methionine, or a protein labeled with a radionuclide such as 
99m Tc - labeled albumin or 90 Y - labeled monoclonal antibodies. 
In clinical nuclear medicine, roughly 95% of radiopharmaceuticals are used with 
diagnostic purposes. Radiopharmaceuticals are administered to the patients only 
once, or a few times at most, in their lifetime. They contain minute amounts of active 
ingredients, with a radionuclide somehow linked to or being the active ingredient 
itself, with the main purpose of obtaining an image or a measure of their biodistribution. 
Radiopharmaceuticals do not usually show any measurable pharmacodynamic 
activity, as they are used in tracer quantities. Hence, there is no dose – response 
relationship in this case and thus differs signifi cantly from conventional drugs. 
Radiation is of course an inherent characteristic of all radiopharmaceuticals. 
Hence, patients always receive an unavoidable radiation dose. In the case of therapeutic 
radiopharmaceuticals, radiation is what produces the therapeutic effect. 

The terms tracer, radiotracer , and radiodiagnostic agent , although long used as 
equivalent to radiopharmaceutical, should be avoided. The preferred and correct 
term is radiopharmaceutical , as the other names can be confusing or do not clearly 
show the nature of these compounds as pharmaceuticals. 
The composition of radiopharmaceuticals is not constant as it varies with time 
as the radionuclide disintegrates. Very often, the half - life of the labeled molecule is 
so short that it must be readily prepared just before its administration to the patient. 
This implies in many cases the use of “ semimanufactured ” , such as radionuclide 
generators, precursors, and cold kits that are also considered a medicinal product 
according to directive 2001/83/EC. 
1.3.1.3 Ideal Characteristics of Radiopharmaceuticals 
Radiopharmaceuticals should have several specifi c characteristics that are a combination 
of the properties of the radionuclide used as the label and of the fi nal radiopharmaceutical 
molecule itself. The radiopharmaceutical should ideally be easily 
produced (both the radionuclide and the unlabeled molecule) and readily available. 
The half - life of the radionuclide should be adequate to the diagnostic or therapeutic 
purpose for which it is designed. It has to be considered that radiopharmaceuticals 
disappear from the organism by a combination of two different processes. The biological 
half - life (showing the disappearance of a radiopharmaceutical from the body 
due to biological processes such as metabolization, excretion, etc.) and the physical 
half - life (due to the radioactive decay of the radionuclide). The combination of both 
parameters gives the effective half - life : 
T 
TT 
T T e 
p b 
p b 
= 
+ 
where T e is the effective half - life, T p the physical half - life, and T b the biological half - 
life. Radiopharmaceuticals should have an effective half - life adequate to the use for 
which they are intended. It should be short (hours) for diagnostic radiopharmaceuticals 
(not longer than the time necessary to complete the study in question) and 
longer for therapeutic radiopharmaceuticals (most often days) as the intended 
effect should have a suffi cient duration. 
The type of decay of the radiopharmaceutical should also be adequate for its 
intended use. Diagnostic radiopharmaceuticals should decay by . emission, electron 
capture, or positron emission, and never emit . or even . particles. On the contrary, 
therapeutic radiopharmaceuticals should decay by . or . emission because the 
intended effect is in fact radiation damage to specifi c cells. 
Regarding the energy emission of diagnostic radiopharmaceuticals, the fi nally 
produced . rays should be powerful enough to be detected from outside of the body 
of the patient. The ideal energy for nuclear medicine equipment is around 150 keV. 
. rays should be monochromatic and photon abundance should be high to decrease 
the imaging time. 
1.3.1.4 Radioactive Decay 
Radionuclides are unstable nuclei that are stabilized upon radioactive decay. More 
than 2000 unstable nuclides have been described so far, most of them radioactive. 
INTRODUCTION 61

62 RADIOPHARMACEUTICAL MANUFACTURING 
The stabilization process can proceed by several different processes, such as spontaneous 
fi ssion, . - particle emission, . - particle emission, positron emission, . - ray 
emission, or electron capture. In all decay processes the mass, energy, and charge of 
radionuclides must be conserved, and many nuclides can decay by a combination 
of any of the above - mentioned processes. 
Fission is the process in which a nucleus breaks down into two fragments (thus 
leading to two different new nuclides) with an emission of two or three neutrons 
and a lot of energy. Spontaneous fi ssion is a rare process that can only occur in heavy 
nuclei. Fission can also be produced by bombardment of certain nuclides with high - 
energy particles (such as neutrons) and is in fact the nuclear process used for the 
production of energy in nuclear energy plants by bombardment of highly enriched 
uranium with neutrons. 
The usual decay process of heavy nuclei is . - particle emission. An . particle is 
a helium ion containing two protons and two neutrons. Alpha particles are heavy 
particles that have a very short range in matter due to their mass, and radiopharmaceuticals 
labeled with . emitters are used only with therapeutic purposes. Their 
clinical use is very limited, and they are mainly used for research purposes or in 
early phase clinical studies. 
Radioactive nuclides that are neutron rich disintegrate by . decay. A . . particle 
is originated by the conversion of a neutron into a proton, along with the emission 
of an antineutrino to conserve energy in the decay process. Beta - emitting radionuclides 
are also used in radiopharmaceuticals for therapeutic purposes. 
Positron decay occurs in proton - rich nuclei. In this case, the positron (or .+ particle) 
is originated by conversion of a proton into a neutron, along with the emission 
of a neutrino to conserve the energy. Positrons are the antiparticle of electrons. In 
a very fast process (10 . 12 s), emitted positrons collide with an electron of a nearby 
atom and both particles disappear in a process called annihilation. The necessary 
conservation of mass and energy accounts for the transformation of the mass of 
both particles into energy, which is characteristically emitted in the form of two 
511 - keV photons almost in opposite directions. Consequently, positron emitters are 
used to label radiopharmaceuticals produced with diagnostic purposes by imaging. 
Proton - rich nuclei can also decay by electron capture. In this process, an electron 
from the innermost electron shell orbitals is captured into the nucleus and transforms 
a proton into a neutron (and a neutrino is emitted for conservation of energy). 
The vacancy created by the lost electron is fi lled by the transition of an electron 
from a higher level orbital, and the energy difference between the intervening orbitals 
is emitted as energy in the form of an X ray. 
For any particular nucleus, several different energy states can be defi ned by 
quantum mechanics. All the excited energy states above the ground state are referred 
to as isomeric states and decay to the ground state by the so - called isomeric transition. 
In . , positron, or electron - capture decay processes, the parent nucleus may 
reach any of these isomeric states of the daughter nucleus. The energy difference 
between the nuclear energy states can be emitted as . rays. A particular situation 
for isomeric transition is that in which the excited state is long lived and is then 
called the metastable state. 
Radioactive Decay Equations, Magnitudes, and Units Radioactive decay is a 
random process, being impossible to tell which particular atom from a group of atoms 
will decay at a specifi c moment. It is then only possible to talk about the average 

number of atoms that disintegrate during a certain period of time, giving the disintegration 
rate ( . dN/dt ) of a particular radionuclide that is proportional to the total 
number of radioactive atoms present at that time. This magnitude is usually called the 
radioactivity (or mainly simply the activity) of a radionuclide and denoted by 
A 
dN 
dt 
N = . = . 
where . is the decay constant and N the number of radioactive atoms. The previous 
differential equation is mathematically solved leading to the exponential equation 
N Ne t 
t = . 
0 
. 
where N t and N are the number of radioactive atoms present at time t = 0 and 
t = t , respectively. 
Radioactivity is expressed in becquerels (Bq), the Internationale System (SI) unit 
for the magnitude A . One Becquerel is defi ned as one disintegration per second 
(dps). Usual activities used in radiopharmacy are in the range of megabecquerels 
or gigabequerels. There is (as usual) a non - SI unit called the curie (Ci). It was 
initially defi ned in a trivial way as the disintegration rate of one gram of radium, 
which was considered to be 3.7 . 10 10 dps. Thus the equivalence between the becquerel 
and the curie is as follows: 
1 27 10 1 37 11 Bq Ci Ci GBq = . = . . 
The decay constant . is a specifi c characteristic of any single radionuclide, but being 
related to probability, it is diffi cult to understand its meaning. Thus, a new magnitude 
is defi ned: the half - life ( t 1/2 ), which is the time required to reduce the initial activity 
of a radionuclide to one - half. In consequence, after one half - life the activity of a 
radionuclide would be A /2, after two half - lives A /4, after three half lives A /8, and 
so on. 
The relationship between the decay constant . and the half - life t 1/2 can be derived 
from the general radioactive decay equation 
t1 2 
2 
/ = ln
. 
An additional (and commonly misunderstood concept) is the mean life . ,which is 
the average life of a certain group of radioactive atoms that is mathematically also 
derived from the decay constant . as 
. 
. 
= = = 1 
2 
1 44 1 2 
1 2 
t 
t / 
/ ln 
. 
1.3.1.5 Principles of Radiation Protection 
Production, transportation, and use of radiopharmaceuticals, as radioactive products, 
is governed by regulatory agencies dealing with radiation protection and 
nuclear safety. 
INTRODUCTION 63

64 RADIOPHARMACEUTICAL MANUFACTURING 
In any case, and albeit the different regulation in different countries, as a general 
principle only licensed personnel working in an authorized facility are authorized 
to handle and use radiopharmaceuticals. Facilities and procedures are subject to 
periodic inspection by offi cial radiation safety offi cers that control production and 
handling of radioactive material, its transportation, proper use, as well as personnel 
dosimetry and radioactive waste disposal. 
The general principles of radiation protection are very simple: 
Justifi cation. All procedures involving radioactive material must be justifi ed. 
Optimization. The radiation exposure to any individual should be as low as reasonable 
achievable. This principle is the widely known ALARA concept, an 
acronym derived from as low as reasonable achievable. 
Limitation. The radiation dose received by the personnel handling radioactive 
material will never exceed the legally established dose limits. It has to be taken 
into account that such limitations do not apply to patients receiving radiopharmaceuticals 
as either diagnostic or therapeutic agents. But nuclear medicine 
physicians, nuclear physicists, and radiopharmacists must ensure that the 
amount of radiopharmaceutical administered to a patient is adapted to his or 
her disease and optimized to obtained the intended result. 
Operational Radiation Protection The fundamentals of operational radiation 
protection (i.e., how to proceed when working with radioactive products) are based 
on three factors: distance, time, and shielding. In any case, it is obvious that the 
radiation hazard is increased with the activity of the radiation source, as can be 
derived from the mathematical equation to calculate the exposure rate X given 
by 
X 
A
d 
= .
2 
where A is the activity of the radiation source, . a constant that is characteristic of 
every radionuclide, and d the distance to the source. 
Distance should be increased as much as possible to decrease exposition and 
exposure time should be reduced to a minimum. Adequate shielding (depending on 
the radionuclide and its emission characteristics) should be used whenever possible 
and handling of high activities should only be carried out by either automated 
systems or proper manipulators. 
1.3.1.6 Detection Devices for Clinical Nuclear Imaging 
Diagnostic radiopharmaceuticals are mostly used for in vivo imaging of the biodistribution 
of the radiopharmaceutical. Depending on whether . or positron emitters 
are used, different devices are employed for clinical imaging. In any case, imaging 
devices are based on detection of the high - energy photons coming from the body 
of the patient upon administration and specifi c uptake of a radiopharmaceutical. 
Advances in nuclear medicine imaging devices now permit in vivo noninvasive 
imaging of such biodistribution and to obtain tomographic (i.e., three - dimensional) 

images that can also give quantitative or semiquantitative information about the 
amount of radiopharmaceutical and even its kinetics. 
1.3.2 PRODUCT DEVELOPMENT 
1.3.2.1 Radionuclides 
When designing a radiopharmaceutical one should have in mind the potential 
hazard the product may have to the patient. The goal must be to have maximum 
amounts of photons with a minimum radiation exposure of the patient. For use in 
therapy, . emitters and . emitters are particularly useful. For diagnostic purposes, 
. emitters are most widely used. In general, those . emitters with a short physical 
half - life and with a . energy between 100 and 300 keV are most widely used in 
medical application, since these can easily be detected by standard . cameras. 
However, positron emission tomography (PET) radiopharmaceuticals involve 
short - lived radionuclides (positron emitters) giving a double set of photons at 
511 keV each. 
1.3.2.2 Carrier Molecules/Active Ingredients 
The function of the carrier molecule is to carry the radioactivity to the target organ 
and to make sure the radioactivity stays there. The uptake of radioactivity should 
be as specifi c as possible in order to minimize irradiation of other organs and parts 
of the body. This is particularly important when using radiopharmaceuticals for 
therapy. But, also, for use in diagnostics, it is desirable that the radiopharmaceutical 
is localized preferentially in the organ under study since the activity from nontarget 
areas can obscure the structural details of the pictures of the target organ. It is 
therefore important to know the specifi c uptake in an organ for a potential chemical 
carrier and also the rate of leaking out of the organ/organ system. Thus, the target - 
to - background activity ratio should be large. There are several approaches to develop 
targeting radiopharmaceuticals. Radioimmunotargeting is one approach frequently 
used for radiopharmaceuticals, where monoclonal antibodies (MAbs) or fractions 
of MAbs are the carrier molecules for the radioactivity. These are binding specifi - 
cally to receptors on cell surfaces in the target organs. 
The target - binding surface of the cell has been well explored with a range of 
tumor - associated and other antigens, identifi ed, and used for pathological tissue 
characterizations. 
The active analog approach in general, whereby a set of compounds is synthesized 
so as to mimic features of a chosen natural compound, has been successful 
[1] . The active analog approach includes the pharmacophore. The concept of the 
pharmacophore is to look at features common to a set of drugs or compounds 
binding to and acting on the same receptors. 
1.3.2.3 Radiolabeling Techniques 
When a labeled compound is to be prepared, the fi rst criterion to consider is whether 
the label can be incorporated into the molecule to be labeled [2] . This may be 
PRODUCT DEVELOPMENT 65

66 RADIOPHARMACEUTICAL MANUFACTURING 
assessed from knowledge of the chemical properties of the two partners. Furthermore, 
one needs to know the amount of each component to be added. This is particularly 
important in tracer level chemistry and in 99m Tc chemistry. 
In a radiolabeled compound, atoms, or groups of atoms of a molecule, are substituted 
by similar or different radioactive atoms or groups of atoms. Saha [2] lists 
six major methods employed in the preparation of labeled compounds for clinical 
use: isotope exchange reactions, introduction of a foreign label, labeling with bifunctional 
chelating agents, biosynthesis, recoil labeling, and excitation labeling. Among 
these, three frequently used methods in radiopharmaceutical synthesis are briefl y 
described below. 
Isotope Exchange Reactions In isotope exchange reactions, isotopes of the same 
elements having different mass numbers replace one or more atoms in a molecule. 
Examples are labelling of iodide - containing material with iodine radioisotopes. 
Since the radiolabeled and parent molecules are identical except for the isotope 
effect, they are expected to have the same biological and chemical properties. 
Introduction of a Foreign Label In this type of labelling, a radionuclide is incorporated 
into a molecule primary by the formation of covalent or coordinated covalent 
bonds. The tagging radionuclide is foreign to the molecule and does not label it 
by exchange of one of its isotopes. Examples are 99m Tc – DTPA (Diethylenetriaminepentacetic 
acid), 51 Cr - labeled red blood cells, and many iodinated proteins and 
enzymes. In many compounds of this category, the chemical bond is formed by chelation. 
In chelation, one atom donates a pair of electrons to the foreign acceptor atom, 
which is usually a transition metal. Most of the 99m Tc - labeled compounds used in 
nuclear medicine are formed by chelation. 
Labeling by Bifunctional Chelating Agents In this approach, a bifunctional chelating 
agent is conjugated to a macromolecule (e.g., protein) on one side and to a metal 
ion by chelation on the other side. Examples of bifunctional chelating agents are 
DTPA, metallothionein, diamide dimercaptide (N 2 S 2 ), and dithiosemi carbazone [2] . 
There are two methods: the preformed radiometal – chelate method and the indirect 
chelator — antibody method. Various antibodies are labelled by the latter, where 
the bifunctional chelating agent is initially conjugated to a macromolecule, which is 
then allowed to react with a metal ion, to form a metal – chelate – macromolecule 
complex. Due to the presence of the chelating agent, the biological properties of 
the labeled protein may be altered and must be assessed before clinical use. 
1.3.2.4 Manufacturing Scale - Up 
As the radiolabeled substances emerge from the laboratory to the clinics, there will 
be a need for scaling up the batch size of the product. This can be done by increasing 
either the total volume of the produced batches or the specifi c activity of the 
product or both. When doing this, the following aspects should be considered: 
The infl uence on the stability of the product itself due to possible radiolysis 
The need for additional operator protection due to handling of increased amounts 
of radioactivity 

Product Stability The stability of a labeled compound is one of the major problems 
in labeling chemistry. It must be stable both in vitro and in vivo. Many labeled 
compounds are decomposed by radiation emitted by the radionuclides in them. This 
kind of decomposition is called radiolysis. Radiation may also decompose the 
solvent, producing free radicals that can break down the chemical bonds of the 
labeled compounds (indirect radiolysis). In general, the risk of radiolysis increases 
with higher specifi c activity of the product. In addition, the more energetic the radiation, 
the greater is the radiolysis. Alpha emitters, leaving most of its energy close by 
the molecules, and thus a high potential risk of radiolysis, give rise to major challenges 
when scaling up is necessary. 
Operator Radiation Protection Even for the largest commercial manufacturer of 
radiopharmaceuticals, the batch volumes are small compared to nonradioactive 
pharmaceuticals. So even a scaled - up production batch can be contained within a 
limited space. When scaling up a radiopharmaceutical production, one always has 
to assure that the radiation outside the contained work unit is acceptable for the 
operator. 
The production of a radiopharmaceutical will normally take place within a contained 
box unit. Depending on the kind of radionuclides used and the amount of 
radioactivity handled in the production process, the box units are shielded by lead 
walls, typically 5 – 15 cm in thickness. When the box is used for production of radiopharmaceuticals 
incorporating . - or . - emitting radionuclides, closed box units 
without any lead coating are suffi cient. Working with these types of radionuclides 
or with smaller amounts of . - emitting radioactivity, as in research scale, suitable 
glove boxes can be used. When working with larger quantities of . - emitting radionuclides, 
the material must be handled by either remote control equipment or 
manipulator tongs incorporated in the wall. 
1.3.2.5 Automation 
Because of the unique operational and safety requirements of radiopharmaceutical 
synthesis, the motivation for the development of automated systems is clear. 
These unique constraints include short synthesis times and control from behind 
bulky shielding structures that make both access to and visibility of radiochemical 
processes and equipment diffi cult. The need for automated systems is particularly 
expressed for PET radiopharmaceutical synthesis, with the short - lived 
radionuclides emitting high - energy . photons at 511 keV. Automated synthesis 
systems require no direct human participation. The short half - lives of the 
PET radionuclides may require repeated synthesis during the day, thus being 
a potential radiation burden for the operator when not using automated 
systems. 
Furthermore, radiopharmaceutical synthesis must be reliable and effi cient and 
result in pharmaceutical - quality products. In addition, the processes must be well 
documented and controlled. Automated systems may support all these challenges 
and requirements. 
One must keep in mind, though, that success in synthesis automation requires 
fi rst and foremost innovative chemistry. PET radiosynthesis draws from a broad 
chemistry knowledgebase rooted in synthetic organic chemistry [3] . 
PRODUCT DEVELOPMENT 67

68 RADIOPHARMACEUTICAL MANUFACTURING 
1.3.3 MANUFACTURING ASPECTS 
1.3.3.1 Design of Manufacturing Sites 
The manufacturing of radiopharmaceuticals is potentially hazardous. Both small - 
and large - scale production must take place on premises designed, constructed, and 
maintained to suit the operations to be carried out. Radiation protection regulations 
stipulate that radionuclides must only be used in specially designed and approved 
“ radioisotope laboratories. ” National regulations with regard to the design and classifi 
cation of radioisotope laboratories must be fulfi lled. Such laboratories are normally 
classifi ed according to the amount of the various radionuclides to be handled 
at any time and the radiotoxicity grading given to each radionuclide. When planning 
the layout of the laboratory, it is recommended to allocate separate working areas 
or contained units for the various procedures to avoid possible cross - contamination 
of radionuclides [4, 5] . Premises must be designed with two important aspects in 
mind: 
The product should not be contaminated by the operator. 
The operator and the environment should be protected from contamination by 
the radioactive product. 
This is the basic principle of good radiopharmaceutical practice (GRPP). 
One of the most important factors in planning a radioisotope laboratory is the 
design of the ventilation system. Laboratories with medium and high grading must 
be designed with the purpose of protecting the personnel from inhaling radioactive 
gases or particles. The system should be designed to provide lower pressure at the 
actual working area compared to the surrounding environment. Furthermore, the 
system should have an appropriate number of air changes per hour and the replacement 
air should be fi ltered. Air extracted from the area where radioactive products 
are handled, though, should not be recirculated. Exhaust air to the environment 
should be monitored for radioactivity, and it may be necessary to install active 
charcoal fi lters to absorb radioactive gases and small particles [4] . 
Aseptic production of radiopharmaceuticals, that is, when the products cannot 
be terminally sterilized, will increase the requirements for the design and construction 
of the premises. Contained workstations and clean - room technology will be 
applied to a much higher degree. The general requirements for the design of such 
premises are the same as for nonradioactive pharmaceuticals, including entry of staff 
and the introduction of materials through air locks. The main difference is found in 
the planning and design of the ventilation system. Laboratories for aseptic work 
normally have a positive pressure relative to the surrounding areas. On the other 
hand, in laboratories for work with radioactivity, it is good practice to have a negative 
pressure to avoid the spread of radioactive material. In order to meet both 
pharmaceutical and radiation protection requirements, it is necessary to balance 
carefully the air pressures in the clean rooms, the air locks, and the surrounding 
areas. From a pharmaceutical point of view a negative pressure in the area designated 
for aseptic work can only be accepted in special cases. There are various ways 
to meet the required balance between these apparently contradictory principles. 
A frequently chosen solution is to use sealed production units or contained work

stations supplied with unidirectional airfl ow (UDAF) and with a lower pressure 
compared to the aseptic laboratory. The laboratory itself may then have positive 
pressure in relation to the surrounding premises. 
Waste management is an important aspect when planning a radiopharmaceutical 
manufacturing site. The key factor is to reduce the amount of radioactive waste to 
a minimum. There should be a system for dividing the waste according to physical 
half - life and radiotoxicity, both for solid and liquid waste. As an example, waste 
containing . emitters is normally kept separately, when possible. National legislation 
will vary considerably and infl uence the requirement that must be set for handling 
of radioactive waste material. 
1.3.3.2 Design of Production Processes 
The design of a radiopharmaceutical production process depends very much upon 
the kind of radiopharmaceutical to be made. Although most radiopharmaceuticals 
are intended for parenteral use, also oral radiopharmaceuticals in different forms 
are widely used. One must emphasize different factors when planning for production 
of parenteral radiopharmaceuticals compared to oral radiopharmaceuticals. 
Still, a common factor is the involvement of radioactive materials, and the radiation 
protection of the personnel must always be an integral part of the design. 
The production of a radiopharmaceutical will normally take place within a contained 
box unit, consisting of either plastic walls or a combination of plastic and 
stainless steel. The latter is more optimal for clean - room work. The box units may 
be shielded by lead, either as large lead panels or as lead brick walls (see Figure 1 ). 
Depending on the kind of radionuclides used and the amount of radioactivity 
handled in the box, the walls are typically 5 – 15 cm in thickness. Shielded production 
units like these are often called “ hotcells. ” 
When the box is used for productions of radiopharmaceuticals incorporating . - 
or . - emitting radionuclides, closed box units without any lead coating may be suffi - 
cient. When handling radionuclides with mixed emitting properties, a possibility is 
to concentrate the shielding to critical parts of the process. This can be done by use 
of local shielding inside the production unit. However, for aseptic production, one 
must keep in mind a potential disturbance of the airfl ow inside the box. 
FIGURE 1 Shielding of box units (hot cells) with lead bricks. ( Photo courtesy of Institute 
for Energy Technology .) 
MANUFACTURING ASPECTS 69

70 RADIOPHARMACEUTICAL MANUFACTURING 
Working with . and . radionuclides and also limited amounts of . - emitting 
radioactivity, the boxes may be mounted with special protection gloves. When 
working with larger quantities of . - emitting radionuclides, the material must be 
handled by either remote control equipment or manipulator tongs incorporated in 
the wall (see Figure 2 ). 
The design of the elements and their assembly on the production unit should be 
such that there are no radiation leaks at the interface. 
When using lead bricks to construct the wall of the production unit, they should 
have a special design. When they are stacked on top of each other, they should 
interlock (see Figure 3 ). This is important to avoid cracks in the wall through which 
radiation can escape. 
Manipulator tongs are fi tted into the wall as part of a large tungsten sphere which 
acts as a ball bearing and thereby allows more fl exibility for the movement of the 
tong inside the box (see Figure 2 ). Lead glass windows, with good shielding properties, 
are fi tted in the lead bricks to allow the operator to overlook the process. 
When large lead panels are used, they should be reinforced with suitable steel 
structures [International Organization for Standardization (ISO) 10648 - 1: 1997]. 
The surfaces of the lead shielding must be smooth and easy to clean. This can be 
achieved by painting the surface of the wall. 
FIGURE 2 Hot cells for manufacturing of larger quantities of . - emitting radionuclides. 
( Photo courtesy of Institute for Energy Technology .) 
FIGURE 3 Lead bricks are interlocked when they are stacked on top of each other. ( Photo 
courtesy of Institute for Energy Technology .) 

In general, the manufacturing of most radiopharmaceuticals consists of the 
following: 
Nuclear synthesis, synthesis of the radionuclide 
Synthesis of the radiolabeled compound 
Pharmaceutical formulation of the radiopharmaceutical 
Nuclear Synthesis Except for radionuclides with ultrashort half - lives, like most 
PET radionuclides, the production of these is normally performed well in advance 
(see Section 1.3.4.1 ). Thus, the radionuclide is considered as a starting material and 
must undergo controls as a starting material. 
Synthesis of Radiolabeled Compound The complexity of a radiopharmaceutical 
may differ greatly, with the radioactive element itself or simple salts as the less 
complex. Very often, though, the radiolabel is part of a larger molecule, and thus a 
radiolabeling procedure is required. This is part of the synthesis of the radiopharmaceuticals, 
which also may involve chemical alteration of a precursor of the active 
ingredient. Both labeling methods and synthesis may involve steps at elevated temperatures 
or even cooling steps. Thus, equipment for heating or cooling must be part 
of the production line. Furthermore, an important part of a synthesis is often the 
purifi cation step, and equipment for this must be available. Typically, this is simple 
chromatographic or ion exchange columns. 
Planning of the process very much depends on the complexity of the process. In 
general, keeping in mind the limited possibilities of direct handling of the materials, 
it is important to keep the processes as simple as possible. For more complex processes, 
automation may be the best solution, if available. 
Pharmaceutical Formulation Even when the radiochemical part of a product is 
simple, the radiopharmaceutical may be a complex solution. A pharmaceutical formulation 
often contains additives in the form of buffers and preservatives: buffers 
to keep the solution at a pH suitable for injection and preservatives to preserve the 
integrity and effi ciency of the radiopharmaceutical. 
Ideally, a solution for injection should be an isotonic solution with a neutral 
(physiological) pH. However, the pH of a radiopharmaceutical is very important for 
its stability, and for labelled compounds, the pH for optimal stability is not always 
equivalent to physiological pH. For iodide solutions, the pH should be alkaline to 
prevent loss of radioiodine. Reducing agents, such as thiosulfate, are often added to 
radioiodide solutions to help this situation. A preservative can act as a stabilizer, an 
antioxidant, or a bactericidal agent. 
Some additives, like benzyl alcohol, are added for a double action. Benzyl alcohol 
0.9% is widely used as a bactericide. In addition, benzyl alcohol reduces radiolysis 
in radiopharmaceuticals and thus acts as a stabilizer. 
1.3.3.3 Design of Production Equipment 
The equipment used for manufacturing operation should be reserved exclusively 
for radiopharmaceuticals [6] . Furthermore, two principles are of utmost importance 
in the design of production equipment [4] : 
MANUFACTURING ASPECTS 71

72 RADIOPHARMACEUTICAL MANUFACTURING 
The equipment must be easy to repair after it has been installed in the production 
unit. 
The equipment must have a simple construction and be easy to assemble, so a 
substitution can be done quickly when total renovation of the equipment is 
necessary. 
Glass is an important material in the construction of production equipment for 
radiopharmaceuticals. This material will become discolored and brittle when affected 
by radiation, and thus repair and/or change of parts of the equipment may be necessary. 
Due to radioactive contamination of the equipment, repair and maintenance 
can often be complicated, and time for decay must be included in the maintenance 
period. To secure the continuous supply of products, it may be necessary to construct 
two production lines in separate production units, where one is kept as a backup 
facility. 
Sometimes it will be necessary to substitute not only parts of a production line 
but also the assembly of equipment as a whole. To facilitate this operation and 
thereby reduce time and radiation exposure, it can be advantageous to build the 
whole production line on a stainless steel support frame fi tted with simple connections 
to electricity, water, and air supplies [4] . The complete withdrawal of a production 
line from a box and the introduction of a new one can then be performed in a 
very short time. 
It is also important to keep in mind, when designing production equipment, that 
all sense of touch is lost when fi ngers are replaced by remote handling tongs. 
The design of the equipment must therefore be as simple as possible. On the 
other hand, when using hot - cell units mounted with handling tongs, it may be favorable 
to use more automated systems in the production line. Systems like these can 
be run and controlled from steering panels outside the box unit. 
Finally, equipment should be constructed so that surfaces that come in contact 
with the product are not reactive, additive, or absorptive so as to alter the quality 
of the radiopharmaceutical. 
1.3.3.4 Cleaning and Sanitation of Production Equipment 
Preparation equipment should be designed so it can be easily and thoroughly 
cleaned. Procedures for cleaning, sanitation, and storage of production equipment 
used in radiopharmaceutical production must be established. Special training is 
necessary for personnel involved in this kind of work with regard to both clean - 
room aspects and radiation protection aspects. 
Before any equipment or materials used during production are removed from 
the production unit, a check for radioactive contamination must be performed. After 
removal, the equipment should be allowed to decay further in a special storage area 
before it is cleaned and made ready for assembly again. 
Glass equipment will normally be sterilized by dry - heat sterilization. Smaller 
equipment, like plastic tubes and rubber stoppers, can be sterilized by autoclaving. 
If available locally, also . irradiation may be a suitable method for sterilization 
of equipment. One must keep in mind, though, that sterilization by irradiation may 
change the composition of plastic and rubber materials. In addition, glass materials 
may be discolored by . irradiation. 

Production equipment that cannot be sterilized must be sanitized and disinfected 
by an appropriate method. This can be done by use of biocides like alcohols (70%), 
hydrogen peroxide, or formaldehyde - based chemicals or a combination of these. 
These can either be used for surface disinfections by wiping or spraying or even 
better by use of gas or dry fog systems for application of the disinfectants. The effect 
of cleaning and sanitation should be monitored. Microbiological media contact 
plates can be used to test critical surfaces, as inside the hot cells or glove boxes. The 
test samples must then be handled and monitored as radioactive contaminated 
units. 
A system must be established for sanitation of all equipment before these are 
transferred into clean areas. 
1.3.3.5 Environmental Control 
Workstations and their environment should be monitored with respect to radioactivity, 
particulate, and microbiological quality. Active air sampling from production 
units for radioactive products (hot cells or glove boxes) is subject to a safety consideration. 
There is always a risk of bringing radioactive contaminated air outside 
the workstation. To avoid the spread of radioactivity during the test, all possible 
exhaust from the test equipment must be sampled and/or controlled. 
A possible approach for testing of particulate and microbiological quality of air 
inside the hot cells or glove boxes is to gain information about airborne particles 
during simulated operations (without radioactivity). 
The use of settle plates is common practice for monitoring of the microbiological 
quality of air inside production units. These must then be placed as close a possible 
to critical parts of the production process in order to show the real microbiological 
burden to the product. 
Warning systems must be installed to indicate failure in the fi ltered air supply to 
the laboratory. Recording instruments should monitor the pressure difference 
between areas where this difference is of importance. 
1.3.3.6 Sterilization of Radiopharmaceuticals 
Sterile radiopharmaceuticals may be divided into those which are manufactured 
aseptically and those which are terminally sterilized. In general, it is advisable to 
use a terminal sterilization whenever this is possible. Terminal sterilization is defi ned 
as a process that subjects the combined product/container/closure system to a sterilization 
process that results in a specifi ed assurance of sterility [7] . Since sterilization 
of solutions normally means autoclaving (steam sterilization), one must assure that 
the radiopharmaceutical product does not decompose when it is heated to temperatures 
above 120 ° C. Many radiolabeled compounds are susceptible to decomposition 
at higher temperatures. Proteins, such as albumin, are good examples of this. Others, 
such as 18 F - fl uodeoxyglucose (FDG), can be autoclaved in some formulation but 
not in others. 
Furthermore, these processes take time, typically 20 – 30 min in total when heating 
up to 121 ° C. For very short - lived product with a half - life of only a few minutes, this 
is not an adequate method. On the other hand, these short - lived products are not 
subject to any storage, and thus the risk of microbiological growth is more limited. 
MANUFACTURING ASPECTS 73

74 RADIOPHARMACEUTICAL MANUFACTURING 
Alternatively, a shorter cycle at a higher temperature might be used, assuming that 
the temperature does not decompose the radiopharmaceutical. 
If terminal sterilization is not possible, aseptic processing must be performed. 
Aseptic processing is a process that combines presterilized materials and presterilized 
equipments in a clean area. 
Heating of radioactive solutions, particularly under elevated pressure (e.g., steam 
sterilization), is also a matter of safety. In order to avoid any contaminated air to 
escape if a container or a seal is broken, autoclaves used for radioactive solutions 
should be placed inside negative - pressure sealed units. Autoclaves used for sterilizing 
high - energy . - emitting radiopharmaceuticals should in addition be supplied with 
proper lead shielding. 
1.3.3.7 Starting Materials 
As for manufacturing of other pharmaceuticals, a system should be established to 
verify the quality of the starting materials used in manufacturing radiopharmaceuticals. 
This system must assure that no material is used for production until it has 
been released by a competent person [qualifi ed person (QP) or others given this 
responsibility]. 
The starting materials as well as the packaging materials should be purchased 
from qualifi ed vendors. It is recommended to use materials described in a pharmacopoeia, 
whenever this is available. Supplier approval should include an evaluation 
that provides adequate assurance that the material consistently meets 
specifi cations. 
Radionuclides involved in manufacturing radiopharmaceuticals must be considered 
as starting materials. For very short - lived radionuclides, where batch analysis 
is not possible, the validation of the production process of the radionuclide is of 
utmost importance. 
1.3.3.8 Labeling and Packaging 
Packaging material should be purchased from qualifi ed vendors. Primary containers 
and closures must be tested to verify that there are no interactions between the 
radiopharmaceutical and packaging material during storage of the product. 
Due to the risk of radiation exposure, it is accepted that most of the labeling of 
the primary (direct) container is done prior to manufacturing. The empty vial can 
be prelabeled with partial information prior to fi ltration and fi lling [6] . This procedure 
should be designed so as to not compromise sterility or prevent visual inspection 
of the fi lled vial. After fi lling of radioactive products, the primary containers 
(vials) must be placed within a shielded container. These containers, which can be 
made of lead or tungsten, vary in size and thickness depending on the amount of 
radioactivity in the vial as well as the radiation properties of the radionuclide. 
Radiopharmaceuticals containing . or . emitters may be placed in thin lead pots, 
typically 2 – 4 mm in wall thickness. On the other hand, for vials containing regular 
doses of high - energy . emitters, such as PET radionuclides, shielding with 3 – 5 cm 
lead/tungsten may be needed. 
Necessary information about the product must be given on the label of the lead 
or tungsten container. Hence, there is no need to study the label on the direct con

PRODUCT MANUFACTURING 75 
tainer. The name of the radiopharmaceutical, including the radionuclide, together 
with the amount of radioactivity in the vial at a stated calibration time is part of the 
necessary information. So is the expiry date of the product. Furthermore, the symbol 
for radioactivity, designed as a black propeller, is obligatory on labels for radioactive 
solutions. 
When the products are intended for distribution and transport, the packaging 
and labeling of the outer packages must be done according to the national regulation 
of the country from which the shipments will depart, transfer, and arrive. The 
outer packaging material must be properly tested in accordance with the type of 
shipment, most frequently type A packages for radiopharmaceuticals. Furthermore, 
the packages must be labeled with radionuclide data, such as type and amount of 
radioactivity, along with the transport index (TI), which indicates the radiation from 
the package at 1 m distance. While the information on the product itself (outside 
the lead pot) is intended for the physicians, the information outside the package is 
intended for the transport personnel. 
1.3.4 PRODUCT MANUFACTURING 
1.3.4.1 Production of Radionuclides 
Radiopharmaceuticals are labeled with artifi cial radionuclides that are obtained by 
bombardment of stable nuclei with subatomic particles or photons. Nuclear reactions 
produced in such a way convert stable in unstable (radioactive nuclei). Several 
kind of devices are used for such purposes, including nuclear reactors, particle accelerators, 
and generators. 
Various types of targets have been designed and used for both reactor and cyclotron 
irradiation. In the design of targets, primary consideration is given to heat 
deposition in the target by irradiation with neutrons in the reactors or charged 
particles in the cyclotrons [2] . As the temperature can rise to 1000 ° C during irradiation 
in both reactors and cyclotrons, the target needs proper cooling to avoid 
burning. Most often, the targets are designed in the form of a foil to maximize the 
heat dissipation. The target element should ideally be monoisotopic or an enriched 
isotope to avoid extraneous nuclear reactions. 
Nuclear Reactors Nuclear reactors are highly complex systems in which two kinds 
of nuclear reactions are useful for the production of clinically useful radionuclides: 
Neutrons produced by the fi ssion of heavy nuclides (such as 235 U or 239 Pu) are used 
in a neutron capture (n, . ) reaction to produce an isotope of the same element 
that is bombarded by the neutrons. Such reactions can be produced almost in all 
elements with different probability. Examples of useful nuclear reactions are 
130 Te(n, . ) 131 Te (which produces 131 I after emission of . particles with a half - life of 
25 min), 50 Cr(n, . ) 51 Cr, 58 Fe(n, . ) 59 Fe, and 98 Mo(n, . ) 99 Mo. The second possibility for 
the use of nuclear reactors is to use fi ssion reactions (n,f) in which a heavy nuclide 
is broken down into two fragments. Many clinically relevant radionuclides can be 
produced from thermal fi ssion of 235 U, such as 131 I, 117 Pd, 133 Xe, and 137 Cs. The isotopes 
produced by this kind of fi ssion reaction must be separated and purifi ed by appropriate 
chemical procedures, but since the chemical behavior of many different heavy 

76 RADIOPHARMACEUTICAL MANUFACTURING 
elements is similar, contamination can often become a problem in the isolation of 
the radionuclide of interest. 
As an example, and due to the particular interest of 99 Mo in radiopharmacy (as 
it is the parent nuclide of 99m Tc in the 99 Mo – 99 mTc generator), the complex process 
used to produce and purify 99 Mo is described below. 
Molybdenum - 99 is produced by fi ssion of 236 U as follows: 
235 1 236 99 135 1 2 U n U Mo Sn n + > > + + 
After irradiation of the uranium target, it is dissolved in nitric acid and the fi nal 
solution adsorbed on an alumina column that is washed with nitric acid to remove 
uranium (and other fi ssion products). Molybdenum is fi nally eluted with ammonium 
hydroxide and further purifi ed by absorption on an anion exchange column from 
which ammonium molibdate is eluted with dilute hydrochloric acid after washing 
the resin with concentrated HCl. The 99 Mo is obtained in no - carrier - added conditions, 
and the most common contaminants can be 131 I and 103 Ru. 
Particle Accelerators: Cyclotrons Both linear and circular particle accelerators 
(cyclotrons) can be used, but the latter have many advantages and are mainly used 
for the production of clinically relevant radionuclides. 
A cyclotron is basically a cylinder - shaped high - vacuum chamber in which by 
means of a magnetic fi eld and a radio - frequency system used to generate an alternating 
electric fi eld, elemental particles can be accelerated to very high energies and 
used as projectiles. The bombardment of stable elements loaded in a properly 
designed target (either solid or fi lled with a liquid or a gas) induces different types 
of nuclear reactions that fi nally lead to the production of radioactive elements. 
Most cyclotrons accelerate negative particles (such as 2 H, 1 H, or even heavier 
particles such as helium cations) that are stripped off the electrons in the stripping 
foils that are used also to focus the beam on the target. As the energy of the incident 
particle is increased, a much greater variety of nuclides can be produced. 
When the nuclides produced have atomic numbers different from those of the 
target elements, such preparations have no stable isotope of the intended element 
and can be considered to be produced in no - carrier - added conditions. 
The target material should ideally be monoisotopic to avoid the production of 
extraneous radionuclides. However, in many cases this is not possible and only isotopically 
enriched targets can be used, thus leading to the production of different 
radionuclides. In this case appropriate methods must be used to separate the different 
elements produced in the target. 
An interesting concept that must always be taken into account in cyclotron - 
produced radionuclides is the saturation activity characteristic of each target and 
each nuclear reaction. The saturation activity is the activity of the radionuclide in 
which the secular equilibrium is obtained between the activity produced in the 
target and the disintegration of the radioisotope. The activity produced at a target 
can be calculated by the equation 
A A e t T = .. 
S 
/ A ( )( ) (ln ) 1 2 . 
where A is the activity obtained for a radionuclide with a half - life of T after irradiation 
of the target during a time of t at a current of . A microamperes. From the 

PRODUCT MANUFACTURING 77 
practical point of view, almost 97% of the saturation activity value is reached after 
irradiation of the target for fi ve half - lives of the radionuclide. Longer irradiation 
times do not produce signifi cant increases in the activity obtained. Methods to 
obtain several cyclotron - produced radionuclides are described below. 
Iodine - 123 can be produced either directly or indirectly in a cyclotron. Direct 
reactions usually lead to 123 I contaminated with other iodine radioisotopes, such as 
124 I or 125 I, due to side nuclear reactions. Using nuclear reactions such as 123 Te(p,n) 123 I, 
122 Te(d,n) 123 I, or 124 (p,2n) 123 I produces 123 I that is obtained after dissolving the target 
in hydrochloric acid by distillation into dilute NaOH. 
In the indirect methods the radionuclide produced after bombardment of the 
target is not 123 I, but a radionuclide that decays to 123 I with a short half - life. The most 
widely used nuclear reactions produce 133 Xe (which decays to 123 I with a half - life of 
2.1 h) by bombardment with high - energy 3 He or 4 He particles or 123 Cs (which decays 
to 123 Xe with a half - life of 5.9 min, and then 123 Xe decays to 123 I) after irradiation of 
124 Xe with high - energy protons. Complex processing and purifi cation processes must 
be used to obtain 123 I in any of these cases, and adequate design and composition 
of the target are critical to facilitate the process. 
Thallium - 201 is obtained using an indirect reaction such as 203 Tl(p,3n) 201 Pb in 
which 201 Pb decays to 201 Tl with a half - life of 9.4 h. Thallium - 201 can in this way be 
obtained pure and free from other contaminants after several purifi cation steps and 
letting the target product decay for 35 h. 
Indium - 111 is produced by a direct nuclear reaction by irradiation of an 111 Cd 
target with 15 - MeV protons. After irradiation the target is dissolved in HCl and 
purifi ed in an anion exchange column. 
Positron emission tomography has become a widely used diagnostic technique in 
nuclear medicine. Ultrashort half - live radionuclides are used in these cases, and such 
radionuclides are mostly obtained in small cyclotrons with high yields and short 
irradiation times. The overall process will be described further in this chapter when 
PET radiopharmaceuticals are described. 
Generators A generator is constructed on the principle of the decay – growth relationship 
between a parent radionuclide with longer half - life that produces by disintegration 
a daughter radionuclide with shorter half - life. The parent and the 
daughter radionuclide must have suffi ciently different chemical properties in order 
to be separated. The daughter radionuclide is then used either directly or to label 
different molecules to produce radiopharmaceutical molecules. 
A typical radionuclide generator consists of a column fi lled with adsorbent material 
in which the parent radionuclide is fi xed. The daughter radionuclide is eluted 
from the column once it has grown as a result of the decay of the parent radionuclide. 
The elution process consists of passing through the column a solvent that 
specifi cally dissolves the daughter radionuclide leaving the parent radionuclide 
adsorbed to the column matrix. 
The main advantage of the generators is that they can serve as top - of - the - bench 
sources of short - lived radionuclides in places located far from the site of a cyclotron 
or nuclear reactor facilities. 
A generator should ideally be simple to build, the parent radionuclide should 
have a relatively long half - life, and the daughter radionuclide should be obtained 
by a simple elution process with high yield and chemical and radiochemical purity. 
The generator must be properly shielded to allow its transport and manipulation. 

78 RADIOPHARMACEUTICAL MANUFACTURING 
Several different generators are used in radiopharmaceutical procedures, but the 
99 Mo/ 99m Tc is with great difference the most important generator of all of them and 
will be described in detail later on in this chapter. 
1.3.4.2 Production of Radiopharmaceuticals 
More than 90% of the radiopharmaceuticals used in nuclear medicine are for diagnostic 
use. PET radiopharmaceuticals, with their ultrashort half - lives, have become 
a signifi cant part of this group of products. Hence PET investigation has been the 
fastest growing imaging modality worldwide the last few years [8] . 
Also for conventional radiopharmaceuticals used in diagnostic, it is favorable to 
use products with short half - lives. Radionuclide generator systems are widely used 
for supply of short - lived radionuclides/radiopharmaceuticals. Several generator 
systems are available and routinely in use within nuclear medicine. Some of these 
are listed in Table 1 . 
Because of the short half - life, the coupling of the radionuclide to the carrier 
molecule must be done immediately before the administration. Hence, there is a 
need to have a constant supply of carrier molecules that can be labeled effi ciently 
on site. For this purpose, several preparation kits have been developed. 
Ready - for - use diagnostic radiopharmaceuticals which are intended for transport 
over some distance typically include radionuclides with half - lives from 13 h and up. 
Among these, products involving the radionuclide 131 I are used for both diagnostic 
and therapeutic indications. This is based upon the mixed emitting properties of 
the radionuclide, giving both . and . emission. The availability, price, and half - life 
(8 days) of this radionuclide, together with the physical properties, have probably 
made it the most commonly used radionuclide in radiotherapy. Although 131 I also is 
frequently used for diagnostic purposes, the radiation characteristics of this radionuclide 
are not really ideal for use in conventional scintigraphy (SPECT) due to 
the high . energies. In addition, the . emission from this radionuclide gives the 
patients an unnecessary radiation burden. Hence, other radionuclides are preferred 
for use in diagnostic nuclear medicine. 
The radioiodine 123 I, on the other hand, is very useful in nuclear medicine because 
it has good radiation characteristics for scintigraphy, such as decay by electron 
capture, a half - life of 13 h, and . emmision of 159 keV. However, the much shorter 
half - life, together with the more complex radionuclide production, makes this radionuclide 
less available and more expensive compared to 131 I. 
There are several 131 I and 123 I radiopharmaceuticals on the market, for both oral 
and parenteral administration. Ready - for - use radiopharmaceuticals that contain 
TABLE 1 Several Radionuclide Generator Systems Useful in Nuclear Medicine 
Parent Nuclide t1/2 Daughter Nuclide t1/2 
68 Ge 280 days > 68 Ga 68 min 
81 Rb 4.7 h > 81m Kr 13 s 
99 Mo 66 h > 99m Tc 6 h 
113 Sn 117 days > 113m In 100 min 
188 W 69.4 days > 188 Re 17 h 

PRODUCT MANUFACTURING 79 
these radionuclides will normally be manufactured by radiopharmaceutical 
companies and distributed to the marked according to a marketing authorization 
(MA). 
Although therapeutic application represents less than 10% of the nuclear medicine 
investigations, therapeutic radiopharmaceuticals are a very important group 
of radiopharmaceuticals. Hence, a brief description is outlined for production of 
therapeutic radiopharmaceuticals following some other selected groups of 
radiopharmaceuticals. 
99 M o / 99m T c Generators The essential part of the most commonly available generator 
system is a simple chromatography column to which the mother radionuclide is 
absorbed on a suitable support material. The daughter radionuclide is a decay 
product of the mother nuclide. Since it is the daughter nuclide that is used to label 
the carrier molecules, it must be possible to separate this from the parent nuclide 
by a chemical separation. 
In a 99 Mo/ 99m Tc generator, the 99 Mo (molybdenum) is fi xed as molybdate to aluminum 
oxide in the column. The daughter nuclide, 99m Tc (technetium), is eluted from 
the column as pertechnetate when using saline solution. Molybdenum - 99 has a half - 
life of 66 h, while 99m Tc has a half - life of 6 h. This is an ideal combination of half - lives, 
giving a system where the daily supply of 99m Tc can easily be calculated from the 
known amount of 99 Mo on the column. The half - life of 99m Tc, along with the radiation 
characteristics of the nuclide, makes it excellent for use in nuclear medicine 
imaging. After reconstitution of kits and formation of various radiopharmaceuticals, 
this radionuclide is used in a major part of all nuclear medicine procedures. 
Although the principle for the generators is similar, the design of 99 Mo/ 99m Tc 
generators from different manufacturers can differ a lot. A drawing of a 99 Mo/ 99m Tc 
generator is shown in Figure 4 . In general, the generator consists of a column with 
adsorbent material where the radionuclide 99 Mo is applied. The column is combined 
with a needle system necessary for the elution process. A sterile fi lter is fi tted on 
the air inlet side of the needles to keep an aseptic system during elution. The saline 
solution for elution may be supplied as a bulk solution suffi cient for several elutions 
FIGURE 4 Typical radionuclide generator system ( ISOTEC, GE Healthcare, AS ). 
1. Saline solution, volume: 5,10, or 15mL 
2. Evacuated vial 
3. Lead shield for eluate 
4. Air filter (0.22 .m) 
5. Special designed stainless steel needles 
6. Glass column with Al2O3 
7. Plastic container 
8. Lead shield (min. 45-mm lead) 
9. Laboratory shield (min. 50-mm lead)

80 RADIOPHARMACEUTICAL MANUFACTURING 
or dispensed volumes suffi cient for a single elution. For both, vacuum is normally 
used to run the elution of the column using sterile evacuated vials. 
Finally, due to the relatively high radiation from 99 Mo, the system must be properly 
shielded by either lead or a combination of lead and tungsten. 
Whether the column is designed to contain liquid after and between elutions, 
determine if this is a wet - column generator or a dry - column generator. When 
liquid is retained at the column (wet generator), radiolysis of water on the column 
may occur as a result . irradiation from 99 Mo. This may change the chemistry on 
the column and thus reduce the yield when eluting the generator. Most commonly, 
when manufacturing wet - column generators, oxidizing agents are added either to 
the saline or to the column itself to avoid reduction of pertechnetate on the 
column. 
A radionuclide generator must be sterile and pyrogen free. Most commonly, the 
generator is sterilized by autoclaving the entire column after the molybdate has 
been bound to the aluminum oxide. Other critical procedures during the production 
and the assembly of the generator must be performed under aseptic conditions. 
Elution of the generator must also be carried out under aseptic conditions while 
using only sterile accessories. 
Other Generators Of the generators listed in Table 1 , two systems are of particular 
interest in nuclear medicine today along with the 99 Mo/ 99m Tc generator, namely the 
68 Ge/ 68 Ga generator and the 81 Rb/ 81m Kr generator. 
68 G e / 68 G a Generator Germanium - 68 has a half - life of 271 days, and 68 Ga (gallium) 
a half - life of 68 min. Gallium - 68 is a PET emitter, and this generator system is a 
valuable source of a short - lived radionuclide in a radiopharmacy or nuclear medicine 
department. However, the system is not as easy or effi cient as the 99 Mo/ 99m Tc 
generator. On the other hand, the longer half - life of the mother nuclide allows use 
of the system for several months. 
This generator can be made up of aluminum loaded on a plastic or glass column. 
Carrier - free 68 Ge in concentrated HCl is neutralized in ethylenediaminetetraacetic 
acid (EDTA) solution and adsorbed to the column. Then 68 Ga is eluted from the 
column with 0.005 M EDTA solution. Alternatively, 68 Ge is adsorbed on a stannous 
dioxide column and 68 Ga is eluted with 1 N HCl [2] . 
81 R b / 81m K r Generator Rubidium - 81 has a half - life of 4.6 h and decays to 81m Kr 
(krypton) by electron capture. Krypton - 81m has a half - life of 13 s and decays by 
isomeric transition emitting . rays of 190 keV. Being an inert gas 81m Kr is used for 
lung ventilation study. 
The parent 81 Rb is adsorbed on an ion exchange resin, and the daughter 81m Kr is 
eluted with air. Because of the very short half - life of 13 s, the studies can be repeated 
every few minutes, and no radiation safety precaution for trapping 81m Kr is needed 
[2] . 
Radiopharmaceutical Kits Radiopharmaceutical kits are nonradioactive ( “ cold ” ) 
products containing the sterile ingredients needed to prepare the fi nal radiopharmaceutical. 
Immediately before administration to the patient, the radionuclide is 
added. From the point of licensing, these semimanufactured products are defi ned 
as radiopharmaceuticals, as they have no other application in medicine [2] . 

PRODUCT MANUFACTURING 81 
Most of these preparation kits have been developed for labeling of various substances 
with 99m Tc. Labeling is normally a single - or two - step procedure consisting 
of adding a solution of 99m Tc - pertechnetate to the preparation kit. The preparation 
kit contains the ingredient necessary for labeling, such as the substance or ligand to 
be labeled, a reducing agent, buffers for pH adjustments, and various stabilizers. The 
reducing agent, very often a stannous salt, is added to bring the radionuclide into a 
valence state with high reactivity. 
Most preparation kits are lyophilized, and the reason for this is to extend the 
shelf life of the products. Some preparation kits can in fact be stored for more than 
one year. Since these products are not radioactive, conventional clean rooms and 
clean - room technology can be applied for production of preparation kits. Most of 
these products have to be produced aseptically, as they cannot be sterilized with 
other methods. During lyophilization of the preparation kits used for 99m Tc labeling, 
it is very important to remove all the oxygen from the kit vial. This is to ensure the 
right valence of the tin salt. Normally, the vials are fi lled with an inert gas, such as 
nitrogen, before the vials are closed completely. It is important, though, that the gas 
is dried. Some manufacturer chose to not completely replace the removed oxygen, 
giving a slightly negative pressure inside the kit vial. This may be favorable for the 
kit - labeling procedure. 
Therapeutic Radiopharmaceuticals Radiopharmaceuticals used for therapy 
(radiotherapy) are designed such that, after administration, they act locally at a 
target by either damaging or killing cells by irradiation. One of the attractions of 
radionuclide therapy is the existence of radiation with quite different dimensions 
of effectiveness, ranging from subcellular (Auger electrons) to hundreds of cell 
diameters ( . particles). In between, . emitters have a tissue range equivalent to 
only a few cell diameters [9] . Alpha emitters have a very high linear energy transfer 
(LET), being very potent at short distances. 
Table 2 lists a selection of radionuclides and radiopharmaceuticals used in 
radiotheraphy. 
TABLE 2 Selected Radionuclides and Radiopharmaceuticals Used for Radiotherapy in 
Routine Use or as Part of Clinical Investigations 
Radionuclide Mode of decay t1/2 Radiopharmaceuticals 
131 I . . / . 8.04 days 131 I - NaI, 131 I - MIBG, 131 I - mAbs 
90 Y . . 2.7 days 90 Y - colloid, 90 Y - DOTATOC, 90 Y - mAbs 
186 Re . . / . 3.8 days 186 Re - sulfi de, 186 Re - HEDP 
188 Re . . / . 17 h 188 Re - HEDP 
177 Lu . . / . 6.6 days 177 Lu - DOTA - Tyr3 - octreotide 
153 Sm . . / . 1.9 days 153 Sm - EDTMP 
89 Sr . . 50.6 days 89 Sr - chloride 
223 Ra . / . 11.4 days 223 Ra - chloride 
211 At . 7.2 h 211 At - mAbs 
213 Bi . 46 min 213 Bi - mAbs 
166 Ho . . / . 26.8 days 166 Ho - colloid 
169 Er . . 9.4 days 169 Er - citrate colloid 
165 Dy . . / . 2.3 h 165 Dy - ferric hydroxide macroaggregate 
32 P .. 14.3 days 32 P - ortho - phosphate 

82 RADIOPHARMACEUTICAL MANUFACTURING 
Pure . and . emitters are easy to shield, and thus production involving these can 
be performed in sealed production units with no lead protection. One must keep in 
mind, though, the potential hazard when inhaling some of these materials. Moreover, 
many radionuclides used for radiotherapy have an additional . component. 
Hence, local lead shielding may be necessary. If the . component is larger or represents 
very high energy emission, a total lead shielded unit may be necessary. The 
latter will be the case when manufacturing 131 I radiopharmaceuticals for therapy, 
since 131 I is a radionuclide consisting of a high - energy . photon together with the . 
component. 
Radiopharmaceuticals for therapeutic use must have a high target - to - background 
ratio. Targeted radiotherapy involves the use of molecular carrier such as a 
receptor - avid compound or an antibody to deliver a radionuclide to cell 
populations. 
A challenge when performing radiolabeling of carrier molecules for targeted 
radiotherapy is the potential risk of radiolysis due to the radiation characteristics 
of the radionuclides involved. When increasing the specifi c activity, as part of the 
scaling up, the risk of radiolytic decomposition of the labeled compound also 
increases. This is particularly pronounced when using . emitters. The addition of 
stabilizers in the form of scavengers can reduce this risk. Benzyl alcohol is an 
example of a compound that acts as a scavenger by catching up with free radicals 
in the solution. 
Another approach is to use kit formulations also for this kind of product. Therapeutic 
radiopharmaceuticals have been developed where the carrier molecule is 
formulated in a lyophilized kit and supplied together with the radionuclide. An 
example of this is the MAb ibritumomab tiuxetan formulated for labeling with the 
. - emitting radionuclide 90 Y. Yttrium - 90 ibritumomab tiuxetan (Zevalin) is used in 
the treatment of non - Hodgkin ’ s lymphoma (NHL). 
The labeling is performed in a centralized radiopharmacy, hospital radiopharmacy, 
or nuclear medicine department immediately before use. 
Radioactive Sanitary Products Radioactive sanitary products could be considered 
as radiopharmaceuticals according to the defi nition given in directive 2001/83/ 
EC, although there are signifi cant differences between radioactive sanitary products 
and classical radiopharmaceuticals. The former can in fact be considered as 
encapsulated radioactive sources, although with the use of microencapsulated sanitary 
products (such as micrometer - sized glass or polymer beads loaded with a 
radionuclide), the difference between both types is becoming more diffi cult to 
establish. 
In any case, radioactive sanitary products are delivered locally (and not systemically 
or orally) for the local treatment of a disease. The idea is to give a high dose 
of radiation to a specifi c part of the body by the implantation of the corresponding 
sanitary product in the desired zone. The sanitary product must not be metabolized, 
destroyed, or removed from the place it has been located during a suffi ciently long 
time as to give the desired high radiation dose. 
The most commonly used radioactive sanitary products are millimeter - sized 
seeds or needles loaded with 103 P, 192 Ir, 90 Sr, or 125 I. Currently micrometer - sized or 
even nanometer - sized beads loaded with 90 Y are being used for the treatment of 
specifi c diseases. 

PRODUCT MANUFACTURING 83 
PET Radiopharmaceuticals PET radiopharmaceuticals are labeled with short - 
lived positron - emitting radionuclides. Such radionuclides can either be produced in 
a cyclotron or obtained from an appropriate radionuclide generator. 
General Considerations The synthesis of PET radiopharmaceuticals has several 
peculiarities substantially different from the procedures followed to prepare conventional 
. - emitting radiopharmaceuticals. A very important issue that must be 
considered is the specifi c activity. For all radiopharmaceuticals it is usually very high 
and can be calculated from the formula 
A 
k 
AT e 
/ 
= 
1 2 
where A e is the specifi c activity, A the mass number of the radionuclide, and T 1/2 its 
half - life. It is then clear that the achievable specifi c activity is higher for radionuclides 
with shorter half - lives, as is the case for the most relevant PET radionuclides 
( 18 F and 11 C). As an example, 18 F produced in no - carrier - added conditions can be 
obtained with specifi c activities of almost 10 10 Ci/mmol, resulting in PET radiopharmaceuticals 
with extremely high specifi c activities. 
For PET radiopharmaceuticals we must always consider that synthesis processes 
must be extremely fast. Consequently, synthesis schemes with as few steps as possible 
must be used, and each of the steps must proceed with high effi ciency. The 
incorporation of the radionuclide to the molecule should ideally be done in the fi nal 
steps of the synthesis. In this way two objectives can be achieved: reduce the overall 
synthesis time (thus increasing the yield) and reduce the number of side reactions 
and secondary undesired products obtained during the synthesis. 
The synthesis of PET radiopharmaceuticals is always carried out at very small 
scale (only a few dozen micrograms of the radiopharmaceutical are obtained) and 
each batch can sometimes only be used for a single patient or a few patients at most. 
Consequently, there is always a big excess of the precursor in the reaction medium, 
and proper purifi cation systems must be used to get rid of all the possible contaminants. 
Such systems must also be very effi cient and fast, and the most usual is to 
apply either semipreparative high - performance liquid chromatography (HPLC) or 
solid - phase extraction - based procedures. 
The position of the radionuclide in the molecule of interest is also critical as it 
will affect the biological behavior of the radiopharmaceutical. Chemical reactions 
must be designed to be stereospecifi c in many cases, as the production of a mixture 
of different stereoisomers complicates the purifi cation of the fi nal radiopharmaceutical. 
Synthesis procedures must also be easy to automate, as very elevated activities 
are used for the synthesis of PET radiopharmaceuticals (several curies usually) and 
appropriate radiation protection systems must be used. 
PET Generators Table 3 summarizes the characteristics of some PET generators. 
So far, the most widely used system has been the 82 Sr/ 82 Rb generator, although due 
to the specifi c physical and chemical characteristics of the daughter radionuclide 
and the half - life of the parent radionuclide, the 68 Ge/ 68 Ga generator is probably one 
of the most interesting systems. Recent advances in gallium chemistry have permitted 
the development of 68 Ga radiopharmaceuticals of clinical interest making 

84 RADIOPHARMACEUTICAL MANUFACTURING 
available PET studies at stand - alone PET centers without a cyclotron with other 
compounds different from the classical 18 FDG. 
PET Cyclotrons Cyclotrons used to produce positron emitters of clinical interest 
(see Tables 4 and 5 ), mainly 18 F and 11 C, do not need to be very big. In fact, small 
devices installed in hospital or academic institutions have long been used for such 
purposes (see Figure 5 ). These devices are easy to operate and maintain, and even 
with single - particle low - energy cyclotrons, it is possible to produce multicurie 
amounts of 18 F and 11 C. 
Some Positron Emitters of Clinical Interest Fluorine - 18 is undoubtedly the most 
widely used positron - emitting radionuclide. This is mainly due to the wide use of 
18 FDG, the PET radiopharmaceutical that has permitted PET to become an everyday 
clinical tool. With the exception of 18 FDG and probably 18 FDOPA, the use of 
other 18 F - labeled radiopharmaceuticals is very limited. However, the chemical and 
physical characteristics of 18 F are excellent: 
TABLE 4 Physical Characteristics of Some Positron 
Emitters of Clinical Use 
Isotope T1/2 (min) % E. + (keV) 
11 C 20.4 99.7 960 
13 N 9.9 99.8 1198 
15 O 2.0 99.9 1732 
18 F 109.6 96.7 634 
TABLE 5 Nuclear Reactions for Production of Most 
Widely Used Positron Emitters 
11 C 13 N 15 O 18 F 
14N(p,a)11C 16O(p,a)13 N 14 N(d,n) 15 O 18 O(p,n) 18 F 
10 B(d,n) 11 C 13 C(p,n) 13 N 15 N(p,n) 15 O 20 Ne(d, . ) 18 F 
11 B(d,2n) 11 C 12 C(d,n) 13 N 16 O( . ,pn) 18 F 
11 B(p,n) 11 C 19 F(p,pn) 18 F 
12 C(p,pn) 11 C 
Note: Most common reactions used in small cyclotrons are bolded. 
Different energies of the incident particle are needed for the different 
nuclear reactions 
TABLE 3 Selected of PET Generators 
Generator Parent T1/2 Daughter T1/2 
Fe/Mn 52 Fe 8.27 h 52m Mn 21.1 min 
Zn/Cu 62 Zn 9.13 h 62 Cu 9.73 min 
Ge/Ga 68 Ge 270.8 days 68 Ga 68.3 min 
Sr/Rb 82 Sr 25.6 days 82 Rb 76.4 s 

PRODUCT MANUFACTURING 85 
It can easily be produced in very high quantities (up to 7 – 9 Ci per batch) even 
in small cyclotrons with just a few hours irradiation time. 
The mean positron emission energy of 18 F is just 0.64 MeV (the lowest of all 
positron emitters with clinical use) and this has several important consequences: 
The dose of radiation received by the patient will be lower and the 
distance between disintegration of the radionuclide and the annihilation site 
(after collision of the positron with an electron) is reduced, thus making PET 
images with higher resolution possible. 
The half - life of 18 F (109 min) is suffi ciently long to carry out complex synthesis 
procedures, apply long PET imaging protocols, and carry out metabolite analysis. 
Furthermore, it is possible to produce the radiopharmaceutical in a laboratory 
and transport it to a distant site only equipped with an imaging device. 
These kinds of “ satellite PET centers ” have boomed all around the world and 
permitted the fast expansion of PET as an everyday clinical tool in certain 
pathologies (mainly in oncological diseases). 
Fluorine is not common in biological molecules, but many drugs contain this 
atom. Fluorine and hydrogen have quite similar radii, and changing a hydrogen to 
a fl uorine atom in a molecule does not usually generate substantial steric differences 
between both molecules. Nonetheless, the electronegativity of fl uorine is usually 
FIGURE 5 Small (less than 2 m in diameter) dual - beam negative ion cyclotron capable of 
easily producing multicurie amounts of 18 F and 11 C. ( Photo courtesy of PET - CUN Center, 
University of Navarra .)

86 RADIOPHARMACEUTICAL MANUFACTURING 
going to change substantially the physicochemical properties of the molecule (reactivity, 
hydrogen bonding, interactions with cognate receptors, metabolization, etc.). 
It is not possible to assume that the biological behavior of a molecule and its fl uorinated 
analog is going to be similar. On the contrary, it is advisable to fi nd substantial 
differences in lipophilicity, biodistribution, protein binding, affi nity for receptors, 
and so on. However, such modifi cations are in many cases very useful to permit the 
use of a 18 F - fl uorinated analog as a PET radiopharmaceutical. In fact, that is the 
case for the most widely use one: FDG. This compound, which accounts for probably 
more than 90% of the PET studies performed in the world every day, is a glucose 
analog that is taken up by the cells by GLUT transporters and metabolized just as 
glucose at the very fi rst steps of glicolysis. But as a consequence of the change of 
the C 2 OH group in natural glucose by a 18 F atom in FDG, the latter cannot be 
isomerized (once phosphorilated) and suffers metabolic trapping being specifi cally 
accumulated in tumoral cells. 
Carbon - 11 has a very short half - life (just 20.4 min) but the chance to substitute 
a carbon atom in any biological molecule by a positron - emitting 11 C is a very interesting 
possibility. This has led to a substantial development of 11 C - labeled tracers. 
The short half - life conditions everything and only PET centers equipped with a 
cyclotron can have a clinical program with 11 C tracers. The production of the radiopharmaceutical 
must in these cases be performed just before the imaging study and 
is usually not started until the patient is already on the PET scanner. 
The 12 C – 11 C substitution will produce chemically identical molecules and give the 
chance to study many biological processes by this noninvasive methodology and can 
also be used in new - drug research and development (R & D). 
Synthesis of PET Radiopharmaceuticals Albeit the requirements for the synthesis 
of PET radiopharmaceuticals previously described, the synthesis process could conceptually 
be reduced to a very simple scheme, as shown in Figure 6 . 
The concept is really simple, but there are considerable diffi culties in each of the 
steps. In many cases it is diffi cult to synthesize a properly designed cold precursor 
that will permit a simple direct reaction with few secondary products. No modifi ca- 
FIGURE 6 General reaction scheme for synthesis of PET radiopharmaceuticals. The precursor 
molecule (A) is designed with the adequate protecting groups (  ) and a reactive 
leaving group ( . ). A reactive form of the radionuclide (  ) is covalently joined to the precursor 
at the reaction site, while the leaving group is eliminated. An intermediate radioactive 
product (B) is obtained that is hence deprotected (2) to produce the fi nal radiopharmaceutical 
(C). A fast and effi cient purifi cation process of C is needed to get read of unreacted cold 
precursor, radionuclide, and intermediate products. 

PRODUCT MANUFACTURING 87 
tions in the confi guration of the chiral centers should be produced during the overall 
process and a simple purifi cation system able to purify the fi nal product in a very 
short time should be found. Additionally, all the reactions should be very fast (just 
several minutes at most) and be easy to automate to be performed in a computer - 
controlled device placed in a shielded hot cell. 
Production Process and Quality Control The production process includes the 
following: 
• Production of the radionuclide in the cyclotron and sending it to the PET 
radiopharmaceutical laboratory 
• Reaction of the radionuclide with an appropriate cold precursor, either in solution 
or in solid phase 
• Purifi cation of the radiopharmaceutical, usually by semipreparative radio 
HPLC or solid - phase extraction 
• Formulation of the fi nal product as an injectable solution (frequently including 
phase change in a rotary evaporator) and the adjustment of tonicity and pH 
• Sterile fi ltration or autoclaving 
The quality control of the fi nal product must be carried out before release of the 
batch (except for the sterility and the endotoxin tests for extremely short - lived 
radionuclides). Consequently, all procedures must not only be very fast but also very 
accurate, and in all cases it is very important to have a properly established quality 
assurance system that might permit parametric release of the produced batches. The 
quality control assays that must be carried out in the radiopharmaceutical includ 
the following: 
• Radionuclidic purity 
• Radionuclidic identity 
• Chemical purity 
• Radiochemical purity 
• Specifi c activity 
• Residual solvents 
• Visual inspection 
• Tonicity 
• pH 
• Sterility 
• Endotoxin 
A PET radiopharmaceutical laboratory must include the cyclotron bunker (where 
positron - emitting radionuclides are produced), the production laboratory, the 
quality control laboratory, and several different ancillary areas. 
In the production laboratory all synthesis and purifi cation processes are carried 
out in remote - operated fully automated computer - controlled systems (synthesis 
modules, see Figure 7 ) located in heavily shielded hot cells (see Figure 8 ). Dispensing 
of individual doses is in many cases also carried out by automated systems. 

88 RADIOPHARMACEUTICAL MANUFACTURING 
1.3.5 QUALITY CONSIDERATIONS 
1.3.5.1 Documentation 
Good documentation constitutes an essential part of the quality assurance system. 
As claimed in the European Community (EC) Guide to Good Manufacturing Practice 
(GMP), Chapter 4: “ Clearly written documentation prevents errors from spoken 
communications and permits tracing of batch history. ” In general, the requirements 
for documentation related to manufacturing of pharmaceuticals, as set in the GMP 
FIGURE 7 Automated synthesis module for PET radiopharmaceutical synthesis located in 
a shielded hot cell. ( Photo courtesy of PET - CUN Center, University of Navarra .) 
FIGURE 8 Production laboratory for PET radiopharmaceuticals. The 10 - cm lead shielded 
hot cells contain computer - controlled automated synthesis modules. ( Photo courtesy of PET - 
CUN Center, University of Navarra .)

regulations, are also valid for manufacturing of radiopharmaceuticals. A recent draft 
proposal of EC GMP Annex 3, “ Manufacture of Radiopharmaceuticals, ” outlines 
the following regarding this issue: 
All documents related to the manufacture of radiopharmaceuticals should 
be prepared, reviewed, approved, and distributed according to written 
procedures. 
Specifi cations should be established and documented for raw materials, labeling 
and packaging materials, critical intermediates, the fi nished radiopharmaceutical, 
and any other critical material. 
Acceptance criteria should be established for the radiopharmaceutical, including 
criteria for release and shelf life specifi cations. 
Records of major equipment use, cleaning, sanitization or sterilization, and maintenance 
should show the product, batch number, date and time, and signatures 
of the persons involved. 
Records should be retained for at least three years unless another time frame is 
specifi ed in national requirements. 
It is of utmost importance to have a system for implementing such documents. 
Any new master document or a new version of such a document must be followed 
by a training process for relevant operators. This training must be recorded as 
well. 
The recording of production data will make it necessary to bring batch documentation 
into the radioisotope laboratory. Hence, it is important to have routines that 
minimize the risk for radioactive contamination of the documents and to ensure 
that any contaminated documents will not leave the controlled area. Today, the use 
of computers instead of paper documents in the laboratory leaves most of the 
paperwork outside the controlled area. 
1.3.5.2 Qualifi cation of Personnel 
As a general principle in GMP, there should be suffi cient qualifi ed personnel to 
carry out all the tasks that are the responsibility of the manufacturer. Furthermore, 
individual responsibilities should be clearly understood by the individuals and 
recorded. 
For personnel working with radiopharmaceuticals, training and qualifi cation 
should cover general principles of GMP and radiation protection. This includes also 
personnel in charge of cleaning premises and equipment used for this type of production. 
All manufacturing operations should be carried out under the responsibility 
of a QP with additional competence in radiation protection. 
1.3.5.3 Quality Control 
All quality control procedures that are applied to nonradioactive pharmaceuticals 
are in principle applicable to radiopharmaceuticals. In addition, tests for radionuclidic 
and radiochemical purity must be carried out. Furthermore, since radiopharmaceuticals 
are short - lived products, methods used for quality control should 
QUALITY CONSIDERATIONS 89

90 RADIOPHARMACEUTICAL MANUFACTURING 
be fast and effective. Still, some radiopharmaceuticals with very short half - lives may 
have to be distributed and used after assessment of batch documentation even 
though all quality control tests have not been completed. It is acceptable, though, 
for these products to be released in a two - stage process, before and after full analytical 
testing. In this case there should be a written procedure detailing all production 
and quality control data that should be considered before the batch is dispatched. 
A procedure should also describe the measures to be taken by the QP if unsatisfactory 
test results are obtained after dispatch (GMP, Annex 3). 
The quality control tests fall in two categories: biological tests and physiochemical 
tests. The biological tests establish the sterility and apyrogenicity, while the 
physiochemical tests include radionuclidic, chemical, and radiochemical purity tests 
along with determination of pH, osmotic pressure, and physical state of the sample 
(for colloids). 
For lyophilized preparation kits containing reducing agents, such as 99m Tc kits, a 
test for moisture content can be necessary. Residual water in the freeze - dried pellet 
may lead to oxidation of the reducing agent. 
Radionuclidic Purity Radionuclidic purity is defi ned as the fraction of the total 
radioactivity in the form of the desired radionuclide present in a radiopharmaceutical. 
Radionuclide impurities may arise from impurities in the target material or from 
fi ssion of heavy elements in the reactor [2] . In radionuclide generator systems, the 
appearance of the parent nuclide in the daughter nuclide product is a radionuclidic 
impurity. In a 99 Mo/ 99m Tc generator, 99 Mo may be found in the 99m Tc eluate due to 
breakthrough of 99 Mo on the aluminum column. The presence of these extraneous 
radionuclides increases the radiation dose to the patient and may also obscure the 
scintigraphic image. 
Radionuclidic purity is determined by measuring the characteristic radiations 
emitted by individual radionuclides. Gamma emitters are distinguished from another 
by identifi cation of their . energies on the spectra obtained from a NaI crystal or a 
Ge (germanium) detector. This method is called . spectroscopy. 
Pure . emitters are not as easy to check as the . emitters. However, they may be 
checked for purity with a . spectrometer or a liquid scintillation counter. 
Radiochemical Purity The radiochemical purity (RCP) of a radiopharmaceutical 
is the fraction of the total radioactivity in the desired chemical form in the radiopharmaceutical. 
Radiochemical impurities arise from decomposition due to the 
action of solvent, change in temperature or pH, light, presence of oxidizing or reducing 
agents, and radiolysis [2] . Examples of radiochemical purity are free 99m Tc - 
pertechenetate and hydrolyzed 99m Tc in labeled 99m Tc radiopharmaceuticals. The 
presence of radiochemical impurities in a radiopharmaceutical results in poor - 
quality images due to the high background from the surrounding tissues and blood. 
It also gives the patient unnecessary radiation doses. 
A number of analytical methods are used to detect and determine the radiochemical 
impurities in a given radiopharmaceutical. Most commonly used are 
methods like paper (PC), thin - layer (TLC), and gel chromatography, paper and gel 
electrophoresis, HPLC, and precipitation. A common principle for the different 
methods is that they can chemically separate the different radiolabeled components 
in the radiopharmaceutical. It may sometimes be necessary to perform more than 

one test method, for instance, TLC and HPLC, to get a complete picture of the different 
radiochemical impurities. Alternatively, one can use one chromatographic 
method consisting of a constant stationary phase but varying the mobile phase 
(solvent). An example is the radiochemical purity test of 99m Tc - methylenediphosphate 
(MDP), a radiolabeled phosphate used in bone scintigraphy. When using two 
TLC systems, one with sodium acetate as a solvent and one with methyl ethyl ketone 
(MEK) as a solvent, the different 99m Tc compunds in the product can be determined. 
A small aliquot of the radiopharmaceutical preparation is spotted on an instant 
thin - layer chromatography (ITLC) strip. The strip is dipped into the chromatography 
fl ask while keeping the spot above the solvent. During the chromatography 
process, the different components of the sample distribute differently in the ITLC 
strip, depending on the solubility and polarity of the components. In systems like 
this, each component is characterized by an R f value, defi ned as the ratio of the distance 
traveled by the component to the distance the solvent front has advanced 
from the original point of application of the test material. The distribution of the 
radioactive components on the strips can be monitored by use of an appropriate 
device for measuring radioactivity and printed in a chromatogram. Figure 9 shows 
typical chromatograms for 99m Tc - MDP in the TLC systems described above. 
Chemical Purity The chemical purity of a radiopharmaceutical is the fraction of 
the material in the desired chemical form. Chemical impurities may arise from the 
breakdown of the material either before or after labeling. Chemical impurities may 
also arise from the manufacturing process, such as aluminum in a 99m Tc eluate, 
coming from the aluminum column on the generator. Residuals of solvent from the 
radiopharmaceutical synthesis are also considered as chemical impurities. If the 
chemical impurity is present before labeling, the result may be undesirable labeled 
molecules. Furthermore, chemical impurities may cause a toxic effect. High - 
performance liquid chromatography and gas chromatography (GC) are important 
methods for determination of chemical impurities in a radiopharmaceutical. 
FIGURE 9 Typical chromatograms for 99m Tc - MDP. The left strip and chromatogram are 
obtained with ITLC - SG in sodium acetate. The right strip and chromatogram are obtained 
in methyl ethyl ketone (MEK). When combining these, any free pertechnetate ( 99m TcO 4 . ) 
and/or hydrolyzed 99m Tc can be detected. Thus the fraction representing 99m Tc - MDP (RCP) 
can be calculated. 
L1 L2 
TC-MDP + Hydr. Tc 
TC-MDP + Tc04– 
==> RCP =100% 
3954 
2966 
1978 
991
3
0.0
O F F 
51.5 103.0 154.5 206.0 
26054 1 1 
19540 
13027 
6514
0
0.0
O 
51.5 103.0 154.5 206.0 
Counts 
Counts 
Distance (mm) Distance (mm) 
QUALITY CONSIDERATIONS 91

92 RADIOPHARMACEUTICAL MANUFACTURING 
Sterility and Pyrogen Testing Sterility indicates the absence of any viable bacteria 
or microorganisms in a radiopharmaceutical preparation. Hence, sterility testing is 
performed to prove that radiopharmaceuticals are essentially free of viable microorganism. 
The test for microbial contamination of these products is best carried out 
with fi lter methods. It is a great advantage to incubate only the fi lters instead of the 
radioactive solutions. 
The test is performed according to the Ph.Eur/USP monograph on Sterility tests 
[13, 14] , but with an important modifi cation. Small batch sizes, typical for radiopharmaceuticals, 
make it necessary to use smaller test volumes than required in the 
monographs. Also the risk for radiation exposure supports this modifi cation. 
All radiopharmaceuticals for human administration are required to be pyrogen 
free. Also the tests for apyrogenicity must be modifi ed when applied for these products. 
The classical rabbit test for pyrogens was never a convenient test for parenteral 
radiopharmaceuticals. Practical problems due to radioactive rabbits and the need 
for larger test volumes made this a diffi cult task. Today, the Limulus amebocyte test 
(LAL) is the method of choice and has been accepted by the Ph. monographs for 
many years. This test is normally done within an hour, compared to several days for 
the rabbit test. 
However, even the LAL test may be too time consuming for the very short lived 
PET radiopharmaceuticals. Hence, less time consuming methods are in progress and 
will probably improve this situation. Meanwhile, it is accepted that the test for apyrogenicity, 
like the sterility test is for most radiopharmaceuticals, is fi nished after 
release of the most short lived radiopharmaceuticals. 
Bubble Point Testing of Filters Parenteral radiopharmaceuticals that are not terminally 
sterilized must undergo a sterile fi ltration process as part of the aseptic 
production procedure. Although the supplier certifi es the fi lters used, they must be 
checked for integrity after use to assure that there has been no leakage during the 
fi ltration. The integrity of the fi lter may be demonstrated by bubble point testing . In 
this test, the fi lter is placed and monitored under controlled pressure. When the test 
is done on wet fi lters, the pressure needed to push gas through the fi lter is defi ned 
as the bubble point. A fi lter with given pore width has a corresponding bubble point 
value. Most frequently, sterile fi ltration is performed by 0.22 - . m fi lters; hence the 
bubble point is about 3 – 4 bars. However, the fi lter supplier should specify the bubble 
point valid for a specifi c fi lter. 
Since this is an in - process test, special caution must be given to radiation protection. 
The test equipment should be placed within a closed and shielded unit and a 
system should be in place to collect any radioactive spill from the test. 
When the fi lter integrity test fails, the sterile fi ltration process must be rejected. 
Visual Inspection of Finished Product As part of the quality control, all parenterals 
will be subject to an inspection for the possible content of particles. Visual 
inspection of radiopharmaceuticals is more complicated than for other pharmaceuticals, 
as radiation protection guidelines strongly discourage any direct eye contact 
with radioactive sources. Normally, the visual inspection of a radiopharmaceutical 
is performed by placing the vial on a rotating station connected to a camera. The 
station is properly shielded, and the operators can study the solution on a distant 
screen. 

1.3.5.4 Validation and Control of Equipment and Procedures 
Preventive maintenance, calibration, and qualifi cation programs should be operated 
to ensure that all facilities and equipment used in the manufacture of radiopharmaceuticals 
are suitable and qualifi ed (GMP, Annex 3). Special emphasis should be put 
on critical equipment for handling of radiopharmaceuticals, such as dose calibrators 
that are used to check the accuracy of the dispensing of patient doses. Particular 
programs are outlined for checking the dose calibrator, including constancy, accuracy, 
linearity, and geometry. The general principles of validation outlined in the 
GMP regulations are valid for radiopharmaceuticals as well as for other pharmaceuticals. 
All validation activities should be planned and clearly defi ned and documented 
in a validation master plan (VMP). Special emphasis should be given on 
the validation of aseptic processes in the production of radiopharmaceuticals. 
Studies, including media fi ll tests, must be performed and recorded to demonstrate 
maintenance of sterility throughout the production process. This is particularly 
important since most radiopharmaceuticals are dispatched and used before the 
sterility test is fi nished. 
1.3.5.5 Stability Aspects of Radiopharmaceuticals 
As discussed already, radiopharmaceuticals are exposed to stability problems, particularly 
when radiolabeled compounds are involved. Decomposition of labeled 
compounds by radiolysis depends on the specifi c activity of the radioactive material, 
the energy of the emitted radiation, and the half - life of the radionuclide. Particles, 
such as . and . radiation, are more damaging than . rays, due to their short range 
and local absorption in matter. The stability of a compound is time dependent on 
exposure to light, change in temperature, and radiolysis. The longer a compound is 
exposed to these conditions, the more it will tend to break down. 
Stabilizers such as ascorbic acid and benzyl alcohol may be added to inhibit or 
delay the decompostion. Many preparations are stored in the dark under refrigeration 
to slow down the degradation of the material [2] . The expiry date of a radiopharmaceutical 
is based upon data from stability studies designed to demonstrate 
the described effects on the product after storage. 
Hence, for most stability studies on radiolabeled compounds, the radiochemical 
purity and pH are the most important physiochemical parameters to study. Moreover, 
for parenteral radiopharmaceuticals, a stability study also has to demonstrate 
the maintenance of sterility and apyrogenicity after storage. 
1.3.6 EXTEMPORANEOUS PREPARATION OF 
RADIOPHARMACEUTICALS 
An extemporaneous preparation is defi ned as a product which is dispensed immediately 
after preparation and not kept in stock [10] . Hence, many radiopharmaceuticals 
could fall into this category due to their limited shelf life. 
The use of extemporaneous preparation should be limited to situations where 
there is no product with marketing authorization (MA) available. This could be 
prepared based upon a prescription for a named patient (magistral preparation) or 
a production based upon a formula and prepared on a regular basis. The latter is a 
EXTEMPORANEOUS PREPARATION OF RADIOPHARMACEUTICALS 93

94 RADIOPHARMACEUTICAL MANUFACTURING 
common situation for many radiopharmaceuticals. For radiopharmaceuticals with 
short half - lives or rare indications, no sizable commercial market exists. Consequently, 
no pharmaceutical company will be prepared to obtain a MA for a product 
that will not yield a profi t due to these limitations. Still, there is a need from a 
medical point of view to have such products available. For radiopharmaceuticals 
incorporating radionuclides with a physical half - life of only a few minutes, only 
local production is feasible. They are therefore prepared in hospital pharmacies or 
laboratories and supplied for individual or small numbers of patients on a daily 
basis. 
The extemporaneous preparation of radiopharmaceuticals is regulated on a 
national level, and hence this regulation may differ from country to country. The 
Pharmaceutical Inspection Convention (PIC/S) has drafted a guide to good practices 
for preparations of medicinal products in pharmacies [10] , valid for medicinal 
products that do not have a MA, prepared extemporaneously or for stock. For 
medicinal products prepared to a larger extent or for use in clinical trials, industrial 
GMPs are applicable. Although the suggested guide outlines a general principle 
according to GMP, different requirements are particularly evident when it comes to 
documentation and quality control testing. There is also a discussion about the 
grades of background environment needed for production, with a differentiation 
between products with shelf lives less than or longer than 24 h [10] . While aseptic 
manufacturing according to industrial GMP has to be performed in grade A with a 
grade B background, this proposed guide opens for a relaxation to this. For an 
aseptic preparation of a product with a shelf life of less than 24 h, using a biohazard 
safety cabinet (BSC), the background environment may be grade D. Even for products 
with a shelf life longer than 24 h, an extensively documented procedure may 
allow grade C in background, as long as grade B clothing is worn. In general, the 
referred draft guide is based much upon a risk related approach and is graduated, 
depending on the size and type of prepared medicinal products. 
As to documentation for extemporaneous prepared products, the proposed guide 
set as a minimum requirement to specify the name, strength, and expiry date of the 
product. If a product is prepared for a single patient (magistral production), it is 
assumed that no end product testing will be required. For radiopharmaceuticals, 
though, the activity in each dose must be measured before administration. Chemical 
and microbiological quality control is not required for products that have a shelf 
life of 24 h or less, provided that frequent process validation is performed. In addition, 
chemical and microbiological information must be available to justify the shelf 
life for the product. 
For products that are prepared extemporaneously at a regular basis or even for 
a limited stock, a product specifi c documentation (product fi le) is needed. This will 
include specifi cations, instructions, and records but also a pharmaceutical assessment 
of safety data, toxicity, biopharmaceutical aspects, stability, and product design. 
The product fi le should also include a product review as soon as a product is used 
repeatedly or over longer periods. 
Furthermore, the drafted guide suggests that the level of end - product testing 
for those products will depend on the associated risk connected to the scale of 
operation, shelf life of the product, frequency of preparation, as well as type of 
product (parenterals, orals) and type of facility where the product has been 
prepared. 

Independent of which regulation applies at a national level to extemporaneous 
or magistral preparation of radiopharmaceuticals, the patients should be entitled to 
expect that these products are prepared accurately, are suitable for use, and will 
meet the expected standards for quality assurance. Pharmacists involved in this kind 
of production must ensure that they and any other staff involved are competent to 
undertake the tasks to be performed and that the requisite facilities and equipment 
are available [11] . As for other radiopharmaceutical production, systems must be in 
place to ensure the operator safety due to handling of radioactive materials. All 
involved staff must have suffi cient training in radiation safety issues, in addition to 
training in GMP. 
REFERENCES 
1. Britton , K. ( 1996 ), Radiopharmaceuticals for the future , Curr. Dir. Radiopharma. Res. 
Dev . (Ed. by Stephen Mather ), viii. Developments in Nuclear medicine, Vol XXX. London, 
UK. 
2. Saha , G. B. ( 1998 ), Fundamentals of Nuclear Pharmacy , 4th ed., Springer , Heidelberg, 
Germany . 
3. Alexoff , D. L. Automation for the synthesis and application of PET radiopharmaceuticals, 
BNL - 68614 Offi cinal File Copy. 
4. Bremer , P. O. ( 1995 ), Aseptic production of radiopharmaceuticals , in Aseptic Pharmaceutical 
Manufacturing , Vol. II, Application for the 1990s , Interpharm , Michael J. Groves and 
Ram Murty , pp. 153 – 180 . 
5. Nordic Council on Medicines . ( 1989 ), Radiopharmacy: Preparation and Control of Radiopharmaceuticals 
in Hospitals , NLN Publications No. 26 , Uppsala, Sweden . 
6. European Commision ( 2003 ), EU Guide to Good Manufacturing Practice , Annex 1 and 
3, Brussels, Belgium, October 8. 
7. Dabbah , R. ( 1995 ), Controlled environments in the pharmaceutical and medical products 
industry: A global review from regulatory, compendial, and industrial perspectives , in 
Aseptic Pharmaceutical Manufacturing, Vol. II, Application for the 1990s , Interpharm , 
Michael J. Groves and Ram Murty , pp. 11 – 40 . 
8. Lee , M. C. , PET and PET/CT are the fastest growing imaging modalities worldwide, paper 
presented at the 5th International Conference on Isotopes (5ICI), Brussels, Belgium, Apr. 
25 – 29 , 2005 . 
9. Zalutsky , M. R. , Pozzi , O. , and Vaidyanatha , G. , Targeted radiotherapy with alpha particle 
emitting radionuclides, paper presented at the International Symposium on Trends in 
Radiopharmaceuticals (ISTR -2005), Vienna, Austria, Nov. 14–18, 2005. 
10. Pharmaceutical Inspection Convention (2006, Aug.), PIC/S guide to good practices 
for preparation of medicinal products in pharmacies, PE 010 - 1 (Draft 2), Geneva , 
Switzerland . 
11. Standards for good professional practice ( 2000 ), Pharm. J . 265 ( 7109 ), 233 . 
12. Kowalsky , R. J. , and Falen , S. W. ( 2004 ), Radiopharmaceuticals in Nuclear Pharmacy and 
Nuclear Medicine , American Pharmacists Association , Forrester Center, WV . 
FURTHER READINGS 
European Commision . ( 2006 ), EU Guide to Good Manufacturing Practice , Annex 3; draft 
proposal, Brussels, Belgium, Apr. 12. 
FURTHER READINGS 95

96 RADIOPHARMACEUTICAL MANUFACTURING 
Rootwelt , K. ( 2005 ), Nukle . rmedisin , 2nd ed. Gyldendal Norsk Forlag AS , Oslo, Norway . 
Schwochau , K. ( 2000 ), Technetium: Chemistry and Radiopharmaceutical Applications , VCH 
Verlagsgesellschaft Mbh , Weinheim, Germany . 
Welch , M. J. , and Redvanly , C. S. , Eds. ( 2002 ), Handbook of Radiopharmaceuticals , Wiley , 
Hoboken, NJ . 

ASEPTIC PROCESSING 
SECTION 2


99 
2.1 
STERILE PRODUCT 
MANUFACTURING 
James Agalloco 1 and James Akers 2 
1 Agalloco & Associates, Belle Mead, New Jersey 
2 Akers Kennedy & Associates, Kansas City, Missouri 
Contents 
2.1.1 Introduction 
2.1.2 Process Selection and Control 
2.1.2.1 Formulation and Compounding 
2.1.2.2 Primary Packaging 
2.1.2.3 Process Objectives 
2.1.3 Facility Design 
2.1.3.1 Warehousing 
2.1.3.2 Preparation Area 
2.1.3.3 Compounding Area 
2.1.3.4 Aseptic Compound Area (If Present) 
2.1.3.5 Aseptic Filling Rooms and Aseptic Processing Area 
2.1.3.6 Capping and Crimping Sealing Areas 
2.1.3.7 Sterilizer Unload (Cooldown) Rooms 
2.1.3.8 Corridors 
2.1.3.9 Aseptic Storage Rooms 
2.1.3.10 Lyophilizer Loading and Unloading Rooms 
2.1.3.11 Air Locks and Pass - Throughs 
2.1.3.12 Gowning Rooms 
2.1.3.13 Terminal Sterilization Area 
2.1.3.14 Inspection, Labeling, and Packaging 
2.1.4 Aseptic Processing Facility Alternatives 
2.1.4.1 Expandability 
2.1.5 Utility Requirements 
2.1.5.1 Water for Injection 
2.1.5.2 Clean (Pure) Steam 
2.1.5.3 Process Gases 
2.1.5.4 Other Utilities 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

100 STERILE PRODUCT MANUFACTURING 
2.1.6 Sterilization and Depyrogenation 
2.1.6.1 Steam Sterilization 
2.1.6.2 Dry - Heat Sterilization and Depyrogenation 
2.1.6.3 Gas and Vapor Sterilization 
2.1.6.4 Radiation Sterilization 
2.1.6.5 Sterilization by Filtration 
2.1.7 Facility and System: Qualifi cation and Validation 
2.1.8 Environmental Control and Monitoring 
2.1.8.1 Sanitization and Disinfection 
2.1.8.2 Monitoring 
2.1.9 Production Activities 
2.1.9.1 Material and Component Entry 
2.1.9.2 Cleaning and Preparation 
2.1.9.3 Compounding 
2.1.9.4 Filling 
2.1.9.5 Stoppering and Crimping 
2.1.9.6 Lyophilization 
2.1.10 Personnel 
2.1.11 Aseptic Processing Control and Evaluation 
2.1.11.1 In - Process Testing 
2.1.11.2 End - Product Testing 
2.1.11.3 Process Simulations 
2.1.12 Terminal Sterilization 
2.1.13 Conclusion 
Appendix 
References 
Additional Readings 
2.1.1 INTRODUCTION 
The manufacture of sterile products is universally acknowledged to be the most 
diffi cult of all pharmaceutical production activities to execute. When these products 
are manufactured using aseptic processing, poorly controlled processes can expose 
the patient to an unacceptable level of contamination. In rare instances contaminated 
products can lead to microbial infection resulting from products intended to 
hasten the patient ’ s recovery. The production of sterile products requires fastidious 
design, operation, and maintenance of facilities and equipment. It also requires 
attention to detail in process development and validation to ensure success. This 
chapter will review the salient elements of sterile manufacturing necessary to 
provide acceptable levels of risk regarding sterility assurance. 
Commensurate with the criticality associated with sterile products, the global 
regulatory community has established a substantial number of the basic requirements 
that fi rms are expected to adhere to in the manufacture of sterile products. 
The most extensive of these are those defi ned by the Food and Drug Administration 

(FDA) in its 2004 Guideline on Sterile Drug Products Produced by Aseptic Processing 
and the European Agency for the Evaluation of Medicinal Products (EMEA) 
Annex 1 on Sterile Medicinal Products [1, 2] . Substantial additional information is 
available from the International Organization for Standardization (ISO), the Parenteral 
Drug Association (PDA), and the International Society for Pharmaceutical 
Engineering (ISPE) (see Appendix ) [3] . The organizations have provided a level of 
practical, experience - based detail not found in the regulatory documents, thereby 
better defi ning practices that are both compliant with regulatory expectations and 
based upon rational, evidence - based science and engineering. 
Consideration of patient risk associated with pharmaceutical production emerged 
largely from regulatory impetus, by which the regulatory community stated its 
intended goal to structure its inspectional process using patient safety as a major 
focus in determining where to allocate their inspectional and review resources. 
Emanating from the International Conference on Harmonization (ICH) efforts to 
produce a harmonized approach to pharmaceutical regulation, risk - based compliance 
has been adopted in Europe, Japan, and the United States [4, 5] . Sterile products, 
especially those made by aseptic processing, have been properly identifi ed as 
a high priority by the global regulatory community. Several risk analysis approaches 
have been developed that can help the practitioner review practices with the goal 
of minimizing risk to the patient [6 – 8] . 
2.1.2 PROCESS SELECTION AND DESIGN 
The production of sterile products is profoundly impacted both by formulation and 
the selection of primary packaging components. Design parameters for a facility and 
selection of appropriate manufacturing technologies for the product require that 
the formulation process and packaging components be chosen and evaluated in 
advance. 
2.1.2.1 Formulation and Compounding 
The vast majority of parenteral formulations are solutions requiring a variety of 
tankage, piping, and ancillary equipment for liquid mixing (or powder blending), 
fi ltration, transfer, and related activities. Suspensions, ointments, and other similar 
products, including the preparation of the solutions for lyophilized products, can be 
manufactured in the same or very similar equipment. The scale of manufacturing 
can vary substantially, with the largest batches being well in excess of 5000 L (typically 
for large - volume parenteral production), down to less than 50 mL for radiopharmaceuticals 
or biologicals customized for a particular patient. 
The majority of this equipment is composed of 300 series austenitic stainless steel, 
with tantalum or glass - lined vessels employed for preparation of formulations sensitive 
to iron and other metal ions. The vessels can be equipped with external jackets 
for heating and/or cooling and various types of agitators, depending upon the mixing 
requirements of the individual formulation. In many facilities, a variety of tank sizes 
are available for use. Larger facilities may have the high - capacity tanks permanently 
installed and permanently connected to process utilities. Smaller vessels are generally 
mobile and positioned in individual processing booths or rooms as needed. 
PROCESS SELECTION AND DESIGN 101

102 STERILE PRODUCT MANUFACTURING 
After sterilizing fi ltration (or sterilization by heat or other means), comparably sized 
vessels are sometimes utilized to contain the product prior to and during the fi lling 
process. These holding vessels are often steam sterilized along with the connecting 
piping prior to use. There are a number of fi rms that fi ll directly from the compounding 
vessel using in - line fi ltration eliminating the intermediate vessel. When this 
approach is used, a small moist - heat - sterilized surge tank or reservoir tank may be 
required, particularly with modern time – pressure fi lling systems. This practice may 
reduce initial facility and equipment cost but places additional constraints 
on operational fl exibility. The use of disposable equipment for compounding and 
holding of sterile formulations is coming into greater use. This eliminates the cleaning 
of vessels prior to reuse, but confi rmation of material compatibility is required. 
Disposable equipment is often used with products manufactured in small to moderate 
volumes, and while reducing initial equipment expenses disposable equipment 
also results in contaminated waste, which cannot be recycled or reused and must be 
treated appropriately. 
Aseptic compounding as required for suspensions and other formulations in 
which open - vessel processes are required mandate an ISO 5 environment providing 
ideally > 400 air changes/hour in which these steps can be performed with minimal 
opportunity for adventitious contamination. This could be accomplished using a 
protective curtain and a unidirectional fl ow hood (UFH) or other more evolved 
designs such as a restricted access barrier (RABs) system or an isolator (technologies 
that provide a higher level of employee separation from the area in which 
materials are handled can get by with lower air exchange rates). All activities requiring 
opening of processing lines such as sampling or fi lter integrity testing should be 
performed using similar protective measures. The preparation of sterile suspensions 
requires a facility/equipment design capable of safe addition of sterile solids to a 
liquid vehicle and is conventionally performed using a specifi cally designed processing 
area to minimize contamination potential. Comparable and greater complexity 
is generally required for creams, ointments, emulsions, and the increasingly common 
liposome formulations. 
Some sterile powder formulations (these are predominantly, but not exclusively, 
antibiotics) may require sampling, mixing, milling, and subdivision activities similar 
to those found in oral powder manufacturing. The facilities and equipment utilized 
for these products is substantially different from that used for liquids, and the production 
area bears little resemblance to that utilized for liquids. These materials are 
received sterile and must be processed through sterilized equipment specifi cally 
intended for powder handling in a fully aseptic environment with ISO 5 protection 
over all open container activities. 
2.1.2.2 Primary Packaging 
The primary package for parenteral formulations provides protection to the sterile 
materials throughout the shelf life. The components of the primary package are 
every bit as important to contamination control and hence safety of the fi nished 
product as the formulation itself, and their preparation must be given a comparable 
level of consideration. The most commonly used container is glass; vials are still the 
most common, although increasingly prefi lled syringes are chosen. Glass ampoules 
are still seen. However, although convenient from a manufacturing perspective, the 

diffi culty involved in opening ampoules while at the same time avoiding problems 
with glass particulate or microbial contamination has reduced their popularity. The 
use of plastic containers (as vials, ampoules, or syringes) is increasingly common 
given their reduced weight and resistance to breakage. Blow - fi ll seal (BFS) and 
form - fi ll seal (FFS) are utilized for the fi lling of numerous ophthalmic and other 
noninjectable formulations in predominantly low - density polyethylene (LDPE) 
containers. With the exception of ampoules and BFS/FFS, an elastomeric closure 
system is also necessary to seal the containers. Some delivery systems (i.e., prefi lled 
syringes, multichamber vials, and others may require more than one elastomeric 
component to operate properly. In the case of vials, an aluminum crimp is applied 
to secure the closure to the vial. Prefi lled syringes may require the preparation and 
assembly of additional components such as needles, needle guards, stoppers, diaphragms, 
or plungers, depending on the specifi cs of the design. Lyophilization is 
required to ensure the stability of some formulations and requires the use of closures 
that allow venting of the container during the freeze - drying process. Full 
seating of the closure is accomplished within the lyophilizer using moving shelves 
to seat the closure. 
Glass is ordinarily washed prior to sterilization/depyrogenation to reduce contamination 
with foreign material prior to fi lling. In aseptic fi ll processes, the glass is 
then depyrogenated using dry heat. This can be accomplished using either a continuous 
tunnel (common for larger volumes and high - speed lines) or a dry heat oven 
(predominantly for small batches). The depyrogenation process serves to sterilize 
the glass at the same time, and thus the glass components must be protected postprocessing. 
This is generally accomplished by short - term storage in an ISO 5 environment 
often accompanied by covering within a lidded tray. There are suppliers 
that offer depyrogenated glass vials and partially assembled syringes in sealed packages 
for fi lling at a customer ’ s site. In this instance, the supplier assumes responsibility 
for the preparation, depyrogenation, and aseptic packaging. Glass ampoules are 
available presealed and depyrogenated; the end user has merely to open, fi ll, and 
reseal the syringe under appropriate conditions. 
Plastic components (whether container or closure) can be sterilized using steam, 
ethylene oxide, hydrogen peroxide, or ionizing radiation. The . irradiation is accomplished 
off - site by a subcontractor with appropriate expertise as these methods are 
considered the province of specialists because of the extreme health hazards directly 
related to the sterilization method. Electron beam sterilization may also be done 
by a contractor, although compact lower energy electron beam systems have been 
introduced that allow sterilization in - house. Steam sterilization is ordinarily performed 
in house, though many common components are becoming available presterilized 
by the supplier. Preparation steps prior to sterilization vary with the 
component and the methods used to produce the component. Rubber components 
are washed to reduce particles, while this is less common with plastic materials. 
Syringes vary substantially in design details and can be aseptically assembled 
from individual components. However, increasingly, these are supplied as presterilized 
partial assemblies in sealed containers. 
The BFS and FFS are unique systems in that the fi nal container is formed as a 
sterile container just prior to the aseptic fi lling step. The BFS requires careful control 
over the endotoxin content of the LDPE (and other polymeric materials) beads 
used to create the containers as well as the melting conditions utilized to form them. 
PROCESS SELECTION AND DESIGN 103

104 STERILE PRODUCT MANUFACTURING 
The FFS utilizes in - line sterilization/drying of the fi lm prior to shaping of the 
containers. 
2.1.2.3 Process Objectives 
The production of parenteral products requires near absolute control over microorganisms. 
Endotoxin contamination is a serious health concern, particularly among 
neonates and infants and also requires a high level of control and validation. Additionally, 
the control of foreign matter, including particles and fi bers of various types, 
is also vitally important to end - user safety. Assuring appropriate control over these 
potential contaminants requires careful attention to several factors: facility design, 
equipment selection, sterilization procedures, cleaning regimens, management of 
personnel, and the process details associated with compounding, fi lling, and sealing 
of product containers. Each of these will be discussed in detail. 
2.1.3 FACILITY DESIGN 
To provide control of microbial, pyrogen, and particles controls over the production 
environment are essential. The facility concerns encompass the entire building, but 
the most relevant components are those in which production materials are exposed 
to the environment. 
2.1.3.1 Warehousing 
Environmental protection of materials commences upon receipt where samples for 
release are taken from the bulk containers. Protection of the bulk materials is 
accomplished by the use of ISO 7 classifi ed environments for sampling. All samples 
should be taken aseptically, which mandates unidirectional airfl ow and full operator 
gowning. This practice is mandated by current good manufacturing practice (CGMP) 
and assures that sampling does not introduce contaminants to the materials that 
will be used in the production. Where central weighing/subdivision of active ingredients 
and excipients are performed, similar protection is provided for identical 
reasons. The expectation is that these measures reduce the potential for contamination 
ingress into materials that have yet to receive any processing at the site. Materials 
and components that are supplied sterile are received in this area, but samples 
are often packaged separately by the supplier to eliminate the need for potentially 
invasive sampling of the bulk containers. Where so - called delivery samples are used, 
it is critical that these samples are known to be fully representative of the production 
process. Additionally, where sterility or bioburden control of sampled materials 
is critical, thought must be given to the methods used to reseal the containers to 
ensure that moisture levels, bioburden levels, or in the case of sterile products sterility 
assurance are not compromised. 
2.1.3.2 Preparation Area 
The materials utilized for production of sterile processes move toward the fi lling 
area through a series of progressively cleaner environments. Typically, the fi rst step 

is transfer into an ISO 8 [Class 100,000, European Union (EU) Grade D] environment 
in which the presterilization preparation steps are performed. Wooden pallets 
and corrugated materials should always be excluded from this zone (and any classifi 
ed environment), and transfers of materials are performed in air locks designed 
to reduce the potential for particle ingress and to a lesser extent microbial ingress. 
Preparation areas provide protection to materials and components for a variety of 
activities: component washing (glass, rubber, and other package components), cleaning 
of equipment (product contact fi ll parts, process tools, etc.), and preassembly/ 
wrapping for sterilization. In some facilities, this area is also utilized to support 
compounding operations in which case process utensils, small containers, and even 
portable equipment will be cleaned and prepared for sterilization. 
Careful attention must be given to material fl ow patterns for clean and dirty 
equipment to prevent cross contamination. In larger facilities, the equipment wash 
room may be a separate room proximate to the preparations area with defi ned fl ows 
for materials and personnel. Ideally, materials should move through the facility in 
a unidirectional fashion, with no cross over of any kind. 
The preparations area typically includes storage areas where clean and wrapped 
change parts, components, and vessels can be held until required for use in the fi ll 
or compounding areas. (Just - in - time practices are desirable for all parenteral operations 
to avoid extensive and extended storage of materials in the higher classifi ed 
fi ll or compounding areas.) The preparations area is ordinarily located between the 
warehouse and the fi lling/compounding areas and connected to each of those by 
material/equipment air locks. 
Preparation areas are supplied with high - effi ciency particulate air (HEPA) fi lters 
(remote - mounted HEPAs are commonplace). The common design requirement is 
more than 20 air changes per hour, turbulent airfl ow (see below), and temperature 
and relative humidity controlled for personnel comfort. As in any clean room area 
designed for total particulate control, the air returns should be low mounted. 
Wall and ceiling surfaces should be smooth, easily cleaned, and tolerant of 
localized high humidity. Floors should be typically monolithic with integral drains 
to prevent standing water. Common utilities are water for injection, deionized 
water, compressed air, and clean/plant steam. Clean - in - place (CIP) and sterilize - 
in - place (SIP) connections may be present if the prep area supports compounding 
as well. 
Ordinarily, present within the preparation area are localized areas of ISO 5 unidirectional 
airfl ow (Class 100) utilized to protect washed components prior to sterilization 
and/or depyrogenation. These areas are not aseptic and should not be 
subjected to the more rigorous microbial expectations of aseptic processing. They 
are designed to reduce/eliminate the potential for particle contamination of 
unwrapped washed materials. Operators accessing these protective zones wear 
gloves at all times when handling materials. 
Operators in the preparations area are typically garbed in low particle uniforms 
(or suits) with shoe, hair, and beard covers. The use of latex or other gloves is 
required when contacting washed components. Sterilized gowns and three - stage 
gowning facilities are not required to enter or work in this ISO 8 environment. 
Gowns are generally donned within a single - stage airlock, which is maintained at a 
pressure slightly negative to the ISO 8 working environment. Separate personnel 
entry/exit are not typically necessary for this lower classifi ed environment. 
FACILITY DESIGN 105

106 STERILE PRODUCT MANUFACTURING 
Equipment within the preparations area varies with the practices of the fi rm and 
can include manual or ultrasonic wash/rinse sinks; single or double door automated 
parts washers; batch or continuous glass washers; stopper washers for closure components; 
CIP/SIP stations; equipment wrap areas (as described above); and staging 
areas for incoming (prewash) components, dirty equipment, and cleaned components/
equipment. An adjacent classifi ed storage area(s) may be present in larger 
facilities to accommodate the full variety of change parts and equipment that is not 
in immediate use. Where the preparations area also supports compounding, it may 
include additional equipment such as pH meters, fi lter integrity apparatus, and the 
like in support of those operations. ( Note: Where compounding requires aseptic 
conditions for rigorous control of bioburden, as is the case for unpreserved biologics 
and other contamination - sensitive products, it is best to provide separate entry for 
compounding. The moisture level and hence contamination potential in a typical 
preparation area is unsuitable for entry into an aseptic compounding area). 
Depending on the scale of the operation, the preparations area may include the 
loading areas for both sterilizers and ovens. In high - throughput operations where 
the use of tunnels for glass depyrogenation is more prevalent, glass washers and 
tunnels for each fi lling line may be in separate ISO 8 rooms accessed from the 
preparations area. 
2.1.3.3 Compounding Area 
The manufacture of parenteral solutions is ordinarily performed in ISO 7 (Class 
10,000, EU Grade C) controlled environments in which localized ISO 5 unidirectional 
fl ow hoods are utilized to provide greater environmental control during 
material addition. These areas are designed to minimize the microbial, pyrogen, and 
particle contributions to the formulation prior to sterilization. Depending upon the 
scale of manufacture, this can range from small containers (up to 200 L) (disposable 
containers are coming into use for these applications), to portable tanks (up to 
600 L) to large fi xed vessel (10,000 L or more have been used) in which the ingredients 
are formulated using mixing, heating, cooling, or other unit operations. Smaller 
vessels are placed or rolled onto scales, while fi xed vessels are ordinarily mounted 
on weigh cells. The vessels may be equipped for temperature and pressure measurement 
instruments, as mandated by process requirements. Compounding areas often 
include equipment for measuring mass and volume of liquid and solid materials 
including, for example, graduated cylinders, and scales of various ranges, transfer 
and metering pumps, homogenizers, prefi lters, and a variety of other liquid/powder 
handling equipment. Liquid handling may be accomplished by single - use fl exible 
hose, assemblies of sanitary fi ttings, or some combination thereof. A range of smaller 
vessels to be used for the addition of formulation subcomponents or excipients to 
the primary compounding tank may be required as well. Because parenteral formulations 
can include aqueous and nonaqueous vehicles, suspensions, emulsions, and 
other liquids, the capabilities of the compounding area may vary. Agitators can be 
propeller, turbine, high shear, or anchor designs depending upon the requirements 
of the products being manufactured, and it is not uncommon to fi nd examples of 
each in larger facilities. It is preferable to perform as much of the process as possible 
while the formulated liquid is nonsterile to ease sterilization requirements, although 
precautions to prevent microbial and endotoxin contamination are important risk 
abatement features. 

FACILITY DESIGN 107 
The formulation area is customarily a combination of open fl oor space, adjacent 
to three - sided booths and individual processing rooms in which the ingredients are 
handled and individual batches are produced. Walls and ceiling materials are selected 
to be impervious to liquids and chemical spills and are easy to clean. Floors in these 
areas are monolithic and should be sloped (at 1 – 3 : 100) to drains with appropriate 
design elements and control procedures to eliminate backfl ow potential (regulatory 
bans on drains in classifi ed areas are focused on protecting aseptic environments 
and are inappropriate for nonsterile compounding areas). Pit scales should be 
avoided in new installations; fl oor - mounted scales intended for cleaning underneath 
the base are preferable. 
Compounding areas are supplied with HEPA fi lters (ceiling - mounted terminal 
HEPAs are more common, though central supply is possible in areas of low contamination 
risk). The common design requirement is more than 50 – 60 air changes 
per hour, turbulent airfl ow (see below), with temperature and relative humidity for 
personnel comfort. Air returns may be at or near fl oor level, with localized extraction 
provided as necessary to minimize dusting of powder materials. Where substantial 
heat is generated from processing or sterilization, a ceiling or high wall return 
may be more appropriate. Wall and ceiling surfaces are smooth, easy to clean, and 
tolerant of localized high humidity. Floors are typically monolithic with integral 
drains to prevent standing water. Common utilities are water for injection, deionized 
water, nitrogen, compressed air, clean/plant steam, and heating and cooling media 
for the fi xed and portable tanks. Water for injection use points are often equipped 
with sanitizable heat exchangers for operator safety. 
Cleaning of the fi xed vessels and portable tanks is accomplished using either 
manual sequenced cleaning procedures or more commonly with a CIP system. 
Cleaning of other items can be accomplished in a wash area accessed from the 
compounding area or in a common wash room incorporating both fi lling and compounding 
equipment. Sterilization of the nonsterile processing equipment and vessel 
is often provided for as an option, even where it is not routinely required to control 
product bioburden. Where production volumes or physical location dictate, the 
compounding area may have a separate preparations area from that utilized to 
support fi lling operations. 
Personnel working in the compounding area typically wear a coverall (which may 
be sterilized for contamination control as required), with head/beard covers, as well 
as dust masks and sterile gloves. Additional personnel protective equipment may 
be necessary for some of the materials being processed. A fresh gown should be 
donned upon each entry into the compounding area. Separate gowning/degowning 
rooms should be provided to minimize cross - contamination potential for personnel 
working with different materials. As nonsterile compounding areas are often ISO 
6 – 7 environments but are not aseptic, the more rigorous contamination controlling 
designs required of aseptic gowning areas (see below) are somewhat reduced. 
2.1.3.4 Aseptic Compounding Area (If Present) 
Where products are fi lled using in - line fi ltration direct to the fi lling machine, an 
aseptic compounding area may not be present. In those instances the fi nal sterilizing 
fi lter will be located in the fi ll room. 
Products that are held/processed in sterilized vessels prior to fi lling require an 
aseptic compounding area. This is typically an ISO 7 in environment with localized 

108 STERILE PRODUCT MANUFACTURING 
ISO 5 unidirectional fl ow present where open - product containers or aseptic operations 
are conducted. Some products may require larger ISO 5 suites with full HEPA 
coverage rather than the more common ISO 5 clean booth design. Fixed vessels in 
this area are cleaned and sterilized in situ, while portable vessels are typically relocated 
to the wash area for cleaning. Sterilization of portable vessels may be accomplished 
at an SIP station in the aseptic core, compounding, or preparations areas. 
When accomplished outside the aseptic processing area, resterilization of the connecting 
lines may be appropriate. Filters for sterilization of solutions from compounding 
to holding vessels are typically located in this environment as well, with 
sterilization by either SIP or sterilization in an autoclave. The use of integrated, 
programmable logic controlled ( PLC) fi lter skids with automatic CIP/SIP and fi lter 
integrity testing is frequently seen for contamination sensitive products. 
Depending upon the formulations being produced, additional sterilized processing 
equipment may be present in this area for use in the process. This can include 
in - line homogenizers, static mixers, and colloid mills. Where sterile powders are 
produced, the aseptic compounding processes can include blending, milling, and 
subdivision equipment. 
Aseptic compounding areas typically require a means to introduce sterile equipment, 
tubing, and other items, so access to a sterilizer is desirable. The aseptic compounding 
area may be contiguous to the aseptic fi lling suites. If it is not, separate 
gowning areas must be provided for personnel as well as separate air locks/pass - 
throughs (see below). 
Personnel working in aseptic compounding wear full aseptic garb: sterile gown, 
hood, face mask, goggles, foot covers, and gloves. Adaptations may be necessary 
for potent/toxic compounds to assure operators are properly protected from 
hazardous materials. Gowning areas are ordinarily shared with aseptic fi lling, 
but where they are not shared a comparable design, albeit on a smaller scale, 
is appropriate. 
The facility design features match that of the aseptic fi lling room/aseptic processing 
areas described in greater detail below. Utility services would mimic those utilized 
in the nonsterile compounding area that is usually adjacent (next to or above) 
to the aseptic compounding area. Temperature and humidity should be controlled 
to similar levels as those required for aseptic fi lling. Since CIP/SIP systems tend to 
generate heat and humidity, suffi cient capacity must be available to control temperatures 
to approximately 18 – 20 ° C and < 50% relative humidity (RH). 
2.1.3.5 Aseptic Filling Rooms and Aseptic Processing Area * 
The fi lling of aseptic formulations (and many terminally sterilized products as well, 
by reason of their lesser number) is performed in an ISO 5 (Class 100) environment, 
which is accessed from an ISO 6/7 background environment in which personnel are 
present. Some measure of physical separation is provided between the ISO 5 and 
ISO 6/7 environments as a means of environmental protection as well as a reminder 
to personnel to restrict their exposure to ISO 5. 
* This section describes the conventional manned clean room; a later section in this chapter will address 
alternative aseptic processing environmental control designs with somewhat different features and 
control measures. 

FACILITY DESIGN 109 
In large operations an aseptic fi lling room is generally one of a multiple suite of 
aseptic rooms which allow simultaneous production of multiple products. The fi lling 
rooms are independent of each other; however, sharing the supporting rooms is 
common. Sterilizer unload rooms, corridors, air locks, storage rooms, lyophilizer 
loading rooms, and gowning rooms (each will be briefl y described as well) may all 
be present, and their arrangement must suit production volumes. Where shared 
common areas are required, the design should feature unidirectional materials fl ow 
to prevent cross - contamination and to minimize the potential for mix - ups. In the 
smallest facilities, only the gowning area might be separate from the fi ll room, and 
all of the supportive activities could be inclusive in a single room (however, unloading 
activities should not occur during fi lling operations). All of these aseptic processing 
areas (APAs) are built to the same design standards: smooth, impervious ceilings, 
walls and fl oors, fl ush - mounted windows, clean room door designs, coved corners, 
fi nishes capable of withstanding the aggressive chemicals utilized for cleaning and 
sanitization. Air returns throughout the APA are located at or near fl oor level. 
Unidirectional airfl ow is provided over all exposed sterile materials, that is, fi ll zone, 
sterilizer/oven/tunnel unload areas, and anywhere else sterile materials are exposed 
to the environment. Air changes in these ISO 5 environments can approach 600 per 
hour, though lesser values have proven successful. Air changes in the background 
environment vary from 60 to 120 per hour. 
The glass container fi ll rooms fi lling machines are connected to depyrogenating 
tunnels and exit ports leading to capping stations. Batch handling of glass is discouraged 
unless isolator systems are employed. In some operations, the in - feed and discharge 
of containers/components may utilize trays, tubs, or bag systems for material 
feed/discharge. Wherever possible, automation of component feeding should 
be considered to reduce contamination risk. Supportive equipment present might 
include carts, weigh stations, stoppering, crimping, sealing, and other fi ll system 
related machinery depending upon requirements. 
The product contact surfaces in this environment are typically removed for cleaning; 
however, in some installations, the sterilization, transfer, and reinstallation of 
the component feed hoppers present such diffi culty that these systems are decontaminated 
in situ with a sporicidal agent, rather than removed after each use. These 
units should still be removed for cleaning and sterilization on a validated periodic 
basis to prevent the buildup of residues that might impact their in - situ decontamination 
or create particle control problems. All other product contact surfaces should 
be sterilized prior to each use. Nonsterilized items should not be allowed to enter 
the ISO 5 portion of the fi ll zone, and sanitization is essential for all nonproduct 
surfaces in the fi ll zone, as well as the surrounding background environment. 
Discharge of sealed containers can be accomplished via a exit port or “ mouse 
hole ” that allows for the passage of the containers from the APA to the surrounding 
environment. Proper design of the mouse hole system ensures protection of the 
classifi ed fi ll area from contamination fl owing against the fl ow of the containers. In 
many instances the discharge is into a nonclassifi ed inspection area that may lead 
directly to the secondary labeling/packaging area. 
Personnel working in aseptic compounding wear full aseptic garb: sterile gown, 
hood, face mask, goggles, foot covers, and gloves. Adaptations may be necessary for 
potent/toxic compounds to assure operators are properly protected from hazardous 
materials. 

110 STERILE PRODUCT MANUFACTURING 
2.1.3.6 Capping and Crimp Sealing Areas 
The application of aluminum seals over rubber stoppers is essential to secure them 
properly. In many older facilities this was accomplished outside the aseptic processing 
area in an unclassifi ed environment. Current practice requires that air supplied 
to this activity meet ISO 5 under static conditions. The protection of crimping has 
resulted in a variety of designs to meet the requirement: Sterile crimps can be 
applied with the aseptic core on the fi lling line; sterile crimps can be applied in a 
separate crimping room accessible from the fi lling room. If the crimpling operation 
is located within the APA, it should be in a separate room maintained at a negative 
pressure differential relative to the fi lling environment. Crimping may alternatively 
be performed in a classifi ed room accessed from a controlled but unclassifi ed environment. 
In this case it is imperative to verify that the environmental controls satisfy 
regulatory expectations for all relevant markets. 
2.1.3.7 Sterilizer Unload (Cooldown) Rooms 
Sterilizers/ovens are unloaded and items staged prior to transfer to the individual 
fi ll rooms. ISO 5 air is provided over the discharge area of ovens (and autoclaves if 
items are sterilized unwrapped) to provide protection until the items are ready for 
transfer. The heat loads in this room may be such that special high - temperature 
sprinkler heads may be necessary to avoid unintentional discharge when unloading 
hot materials. This room may not be separate from the corridor used to connect the 
fi ll rooms. It is ordinarily adjacent to any aseptic storage area. 
2.1.3.8 Corridors 
Corridors serve to interconnect the various rooms that comprise the APA. Fill 
rooms, air locks, and gowning rooms are accessed from the corridor. They can also 
be utilized for modest storage as well. 
2.1.3.9 Aseptic Storage Rooms 
In general, extensive use of in - process storage areas should be avoided. It is best to 
operate the aseptic facility in a just - in - time mode in which components and 
equipment are sterilized shortly before they are required for use in the fi lling or 
compounding areas. Some limited storage is necessary for nonproduct contact 
materials such as sanitizing agents, environmental supplies and equipment, and 
other items. 
2.1.3.10 Lyophilizer Loading and Unloading Rooms 
The loading of lyophilizers is accomplished under ISO 5 environmental conditions 
within the aseptic processing area. Several possible locations are possible: within 
the aseptic fi ll room itself, in a separate room adjacent to the fi ll room, or in a separate 
room remote from the fi ll room. There are pros and cons with each of these 
selections which should be carefully considered in the facility design. There is a 

universal expectation that fi lled containers of product should be maintained under 
ISO 5 conditions during transfer and lyophilizer loading. Many modern facilities 
incorporate automatic lyophilizer loading and unloading. Automation of loading, 
unloading, and in the case of vials transfer to the crimping station greatly reduces 
contamination risk and is highly recommended. 
If manual transfer is unavoidable, location of the lyophilizer relatively close to 
the fi lling line enables protected transfer to be accomplished rather easily. Remote 
locations may require transfer of product in carts capable of providing ISO 5 quality 
air. These carts will generally require battery power in order to run the necessary 
air blowers and control systems. Alternatively, product trays could be placed in air - 
tight carriers; this activity and the sealing of the carriers would have to be accomplished 
under ISO 5 conditions. Locating the lyophilizer in the fi ll room may restrict 
the ability to unload the dryer while the fi lling line is in use, particularly if the 
lyophilizer is loaded and unloaded manually, which would increase the clean room 
personnel load and potentially increase contamination risk. 
The use of trays during lyophilization is less common, nevertheless, ring trays 
with removable bottoms are sometimes used to transfer vials to/from the lyophilizer. 
Where trays are used, they must be cleaned and sterilized prior to each batch. Large 
lyophilization facilities will sometimes use an automated loading/unloading system 
in which all shelves or a shelf at a time are processed. Regardless of the practice, 
ISO 5 conditions are required for all areas of the facility in which partially stoppered 
containers are transferred or handled. As previously mentioned, it may be possible 
in some operations to transfer containers in a manner that they are not exposed to 
the environment during transfer. 
Upon completion of the drying process, the containers will ordinarily have their 
stoppers fully seated on the container within the freeze dryer. The stoppered containers 
are then passed through a sealing station in which aluminum crimps are 
applied. This may be accomplished on the fi ll line, or using a separate crimping 
machine. Precautions will need to be taken to ensure that only fully stoppered vials 
are transferred to the crimping station. This can be accomplished by automatic 
inspection systems of various designs. It is increasingly common for product transfer 
to crimping and crimping itself to be done under unidirectional airfl ow. It should 
be noted that a crimpling station will generally not meet ISO 5 particulate air quality 
requirements when the crimper is operating since the generation of relatively high 
levels of particulate is an inherent feature of this process. 
2.1.3.11 Air Locks and Pass - Throughs 
Air locks serve as transition points between one environment and another. Ordinarily, 
they are designed to separate environments of different classifi cation: that is, 
ISO 6 from ISO 7. When this is the case, they are designed to achieve the higher of 
the two air quality levels in operation. If they are utilized for decontamination purposes 
for materials/equipment that cannot be sterilized, but must be introduced into 
the higher air quality environment, they may be fi tted with ultraviolet (UV) lights, 
spray systems, vapor phase hydrogen peroxide generators, or other devices that may 
be effectively utilized for decontamination of materials. Regardless of the design or 
the decontamination method employed, the process should be validated to ensure 
FACILITY DESIGN 111

112 STERILE PRODUCT MANUFACTURING 
consistent effi cacy. The doors at each end can be automatically interlocked or 
managed by standard operating procedure. In some instances a demarcation line is 
used to delineate the extent to which individuals from one side should access the 
air lock. It is good practice to carefully control and to minimize the time that any 
operator spends accessing an air lock, therefore transfer of materials should be 
carefully planned to minimize frequent and spontaneous access. Additionally, the 
capacity of the air lock should be carefully considered relative to the actual production 
requirements. Air locks that lack suffi cient capacity and that cannot provide 
suffi cient air exchange will be less suited to the control of contamination into more 
critical areas of the aseptic processing environment. 
A smaller scale system with comparable capabilities is the pass - through. This 
differs from the air lock primarily in dimension, as items are typically placed into the 
pass - through by personnel, whereas the air lock is customary for pallet, portable 
tanks, and larger items that are either rolled or mechanically lifted into position. 
The operation of the pass - through can be either manual or automatic with similar 
capabilities to that of the air lock described above. In general pass - throughs should 
be supplied with HEPA fi lters and should be designed to meet the air quality level of 
the higher air quality classifi cation room served. Pass - throughs should also be interlocked 
and provide adequate facilities for decontamination of materials being 
transferred. 
Air locks and pass - throughs are bidirectional and can be used for movement in 
either direction. When used as an exit route, the decontamination procedure can be 
omitted. Where production volumes warrant separate entry and exit, air locks may 
be necessary to maintain both adequate capacity and separation between clean and 
used items. In an emergency, airlocks can serve as emergency exits for personnel, in 
which case the interlocks can be overridden. 
2.1.3.12 Gowning Rooms 
The gowning area used for personnel entry/exit presents some unique problems. 
Gowning facilities must be designed to the standards of the aseptic processing area, 
yet personnel upon entry are certainly not gowned. Because ungowned staff will 
release higher concentrations of contaminants into the environment, gown rooms 
must be designed with suffi cient air exchange so that this contamination is effectively 
and promptly removed. In general, the contamination load within a gowning 
environment will require air exchange rates at the high end of recommended levels 
for a given ISO 14644 air quality classifi cation. Gowning areas are separated into 
well - defi ned zones where personnel can progress through the various stages of the 
gowning process. 
The most common approach in industry is a three - stage gowning area design in 
which three linked rooms with increasing air quality levels are utilized to effi ciently 
and safely affect clothing change. Staff should enter the fi rst state of the gowning 
room wearing plant uniforms. No articles of outerwear worn outside the facility 
should be worn to the gowning area. Therefore, a pregowning room equipped with 
lockers is required so that operators can change into dedicated plant clothing prior 
to moving to the gowning area. Generally, the pregowning locker area is not classi- 
fi ed, although entry is controlled and temperature and humidity are maintained at 
20 – 24 ° C and 50% ± 10%. The pregown area should have extensive hand - washing 

facilities equipped with antibacterial soap, warm water, and brushes for cleaning 
fi nger nails. Soap and water dispensing should be automatic and hands should be 
air rather than towel dried. The pregown area should have typical clean room wall 
and fl oor fi nishes along for frequent and rigorous cleaning and sanitization. The 
pregown area is bidirectional as it is used as both an entry and exit point. Separate 
pregown areas are required for female and male personnel. A typical complement 
of garments for exit of the pregown area includes surgical scrubs or other nonparticulate 
shedding plant uniform. Ideally, the uniform should have a high neck and 
sleeves which extend to the lower wrist. Hair covers and beard covers are donned 
in the pregown area. 
Upon entry into the fi rst - stage gowning room, which is generally designed to an 
ISO 7 air quality level, the operators often don a second hair cover, sterilized gloves, 
and a sterilized surgical mask. In the second and third stages of the gowning area 
room classifi cation is typically ISO 6 or ISO 6 followed by ISO 5 at the exit point. 
Different fi rms have different gowning sequences. However, in every case the fl ow 
of personnel and arrangement of gowning materials should be such that personnel 
fl ow is in one direction. In the last of the three gowning stages, secondary protective 
equipment can be donned, including sleeve covers and a second set of gloves. Some 
fi rms will use tape to secure the gloves to the sleeves to prevent separation. A dry 
glove decontamination point utilizing disinfectant foam is generally provided prior 
to exiting the gowning area; this should be a hands - free operation. In some facilities 
air showers, which provide a high - intensity blast of HEPA air for a predetermined 
length of time, are employed after gowning is completed. Side - by - side gowning of 
personnel should be avoided to preclude adventitious contamination. Similarly, 
personnel exiting the aseptic area should use a separate degowning area. These 
design practices are appropriate in all but the very smallest facilities where only a 
single aseptic operator is present. 
2.1.3.13 Terminal Sterilization Area 
The terminal sterilization of fi nished product containers may be performed in the 
same sterilizers utilized to supply the aseptic processing operations. The differing 
process needs of terminal sterilization will sometimes dictate the use of sterilizers 
specifi cally designed for terminal sterilization incorporating air - over pressure 
systems, internal fans, and spray cooling. Where this is the case, the terminal sterilizer 
is located proximate to the crimping/sealing areas. A double - door sterilizer 
design is preferred with staging areas for fi lled containers to be sterilized and a 
separate area for containers that have completed the process. Classifi cation of these 
areas is not required as the containers are closed throughout the sterilization process. 
The fl ooring materials in this area should be monolithic to allow for easy cleanup 
in the event of container breakage. 
2.1.3.14 Inspection, Labeling, and Packaging 
These activities are performed on fi nished product containers in unclassifi ed environments. 
The primary design requirements are straightforward: separation of products 
to prevent mix - up, adequate lighting for the processes, and control over labeling 
materials. 
FACILITY DESIGN 113

114 STERILE PRODUCT MANUFACTURING 
2.1.4 ASEPTIC PROCESSING FACILITY ALTERNATIVES 
The successful production of parenteral drugs by aseptic processing requires an 
environment in which microorganisms and particles are very well controlled. The 
means to accomplish this has undergone substantial change over the last 50 years 
(see Figure 1 ) with continuing refi nement. The earliest aseptic processing systems 
used glove boxes with minimal (if any) airfl ow and manual disinfection in which 
manual processes were performed. The availability of HEPA fi lters in the late 1950s 
led to human - scale clean rooms in which processing equipment could be installed. 
Aseptic processing changed radically once entire clean rooms became feasible. 
As it had always been recognized that personnel were the dominant source of 
contamination, the majority of designs utilized some measure of physical separation 
between the operator and the critical zone (sterile fi eld) in which the aseptic 
processing activities were performed. Separative devices (a term that is now 
embodied in ISO 14644 - 7 Separative Enclosures) of different design and varying 
capability have been successfully employed including fl exible curtains and fi xed 
plastic shields with or without integrated gloves/sleeves [9] . In the most evolved 
designs operation of the equipment is interlocked with the surrounding enclosure, 
such that equipment stops running when the doors are opened. These latter designs 
represented the pinnacle of clean room - based aseptic processing into the early 
1990s. 
Isolators represent a return to operator separation principles utilized during the 
glove box era, albeit with substantial improvements in the form of rapid transfer 
ports for material transfer, air - handling systems utilizing modern HEPA fi lters, and 
reliable decontamination systems. The salient element of all isolator designs is the 
completeness of separation between the internal and external environments. This 
single feature affords vastly superior performance relative to manned clean rooms 
in excluding personnel - derived contamination and has comparable advantages for 
the containment of potent compounds. While initial adoption of the technology was 
slowed by the novelty that isolators presented to users, much of the initial reluctance 
has been overcome [10, 11] . Isolators for aseptic processing vary in complexity, size, 
and amount of processing equipment. They can be utilized for processing ranging 
Aseptic Processing Family Tree 
Gloveboxes 
Conventional 
Cleanroom 
Barrier 
Systems 
RABs 
Closed 
Isolators 
Open 
Isolators 
BFS/FFS 
FIGURE 1 Aseptic processing family tree. 

from manual compounding of small batches to high - speed fi lling of fi nal product 
containers. Depending upon the process requirements, isolators can be utilized for 
containment of potent compounds (under negative pressure while still nonsterile) 
during the compounding, aseptic operation (under positive pressure) for preparation 
and transfer of components and aseptic containment (also under positive pressure) 
for aseptic fi lling of the potent drug solution. 
Firms that were intimidated by or unconvinced of the superiority of isolators 
developed the restricted - access barrier (RAB) system as a potentially less complex 
and less costly alternative [12] . The real - world utility of RABs systems is unknown; 
there are still relatively few installations; thus, the experience base is still emerging. 
Also unconfi rmed at this point are the actual validation and ongoing process control 
requirements which make direct comparison of project time lines and overall costs 
with isolators somewhat speculative. 
There are specialized technologies such as BFS and FFS that are appropriate for 
aseptic processing, but these are restricted to fi lling processes only. A number of 
other new technologies are being developed for use in aseptic processing, including 
vial isolators and closed vial fi lling [13 – 15] . All of these have the objective of reducing 
contamination through reduction in human involvement or increased protection 
of the container. Further advances in processing including gloveless isolator designs, 
robotics, and others are already under active development to further improve the 
safety of parenteral products. 
2.1.4.1 Expandability 
Large facilities often include design elements that facilitate later expansion of the 
facility to add additional capacity. The most common of these is extension of an 
aseptic corridor to additional fi lling suites; reservation of space for additional sterilizers; 
and allocating space for additional or oversizing initial utility systems. Obviously, 
these types of changes require careful design and must be properly managed 
during execution to avoid impact on existing operations. 
Isolation technology changes this dynamic signifi cantly by eliminating most of 
the disruption on current activities, as fabrication of the isolator occurs off - site, and 
installation can be minimally disruptive compared to what is required with a clean - 
room design. Isolators are generally installed in ISO 8 space; therefore, it is possible 
to build a rather large ISO 8 facility in which equipment can be moved, replaced, 
or reconfi gured quite easily compared to conventional human - scale zoned aseptic 
processing areas. 
2.1.5 UTILITY REQUIREMENTS 
Any utility in direct product contact is subject to formal qualifi cation through con- 
fi rmation of the quality of the delivered material at each use point. Water - for - 
injection (WFI) systems are considered the most critical of all, and the qualifi cation 
period for WFI is the longest and may be as long as 3 months. The remaining product 
contact utilities can be qualifi ed more rapidly. Nonproduct utilities requirements 
can be satisfi ed by commissioning. 
UTILITY REQUIREMENTS 115

116 STERILE PRODUCT MANUFACTURING 
2.1.5.1 Water for Injection 
The most important utility in sterile manufacturing is WFI. Not only is it a major 
component in many formulations, it is also utilized as a fi nal rinse of process equipment, 
product contact parts, utensils, and components. In some facilities it may be 
the only grade of water available and is used for initial cleaning of items as well. 
The WFI may be produced by either distillation (multiple effect or vapor compression) 
or reverse osmosis (generally in conjunction with deionization) and is ordinarily 
stored and recirculated at an elevated temperature greater than 70 ° C to prevent 
microbial growth [16, 17] . Where cold water is required, it may be supplied by use 
point heat exchangers or using a separate cold loop (usually without a storage 
capability). Point - of - use cool water drops and reduced temperature circulation 
loops are generally sterilized or high - temperature sanitized at defi ned and validated 
intervals. The design details of the WFI system varies with the incoming water 
quality, local utility costs, and operational demands. Very small operations may not 
have a WFI system and will utilize larger (5 L or larger) packages of WFI for formulation 
and cleaning. 
Other grades of water may be present in parenteral facilities for use as initial 
rinses and detergent cleaning. The water utilized for these purposes is generally of 
relatively low bioburden and is often deionized, softened, ultra - fi ltered, or in some 
instances prepared by distillation or reverse osmosis, resulting in chemical purity 
similar to, if not identical to, WFI. Systems for the preparation of this water are 
subject to qualifi cation, validation, and routine analysis to assure consistent quality. 
2.1.5.2 Clean (Pure) Steam 
Sterilizers and SIP systems in the facility are supplied with steam which upon condensation 
meets WFI quality requirements (testing steam condensate for microbial 
content is not fruitful). The steam can be produced directly from the water of suffi - 
cient purity to meet the input requirements of the steam generator. Steam generators 
are phase transition technologies that operate like a still, so it is no more 
necessary to provide these devices with WFI feed water than it would be to double 
distill WFI. (Production from WFI is certainly possible, but that is both expensive 
and an unnecessary precaution.) Modest quantities of steam can be produced from 
the fi rst effect of a multiple effect WFI still, however, with a resultant loss of WFI 
output [18] . 
2.1.5.3 Process Gases 
Air or nitrogen used in product contact is often supplied in stainless steel piping 
and ordinarily equipped with point - of - use fi lters; quite often an additional fi lter is 
placed within the distribution loop or at the entry point into a room resulting in a 
form of redundant fi ltration. Compressed air is typically provided by oil - free compressors 
to minimize potential contaminants and is often treated with a drier to 
obviate the possibility of condensation within the lines which could be a source of 
contamination. Nitrogen is supplied as a bulk cryogenic liquid. Argon and carbon 
dioxide have also been utilized as inerting gases, while propane or natural gas may 
be needed for sealing of ampoules. 

2.1.5.4 Other Utilities 
The operation of a parenteral facility often entails other utilities for the operation 
of the equipment. These include plant steam, jacket cooling water, and instrument 
air. 
2.1.6 STERILIZATION AND DEPYROGENATION 
The preparation of the drug formulation, components, and equipment entails the 
use of various sterilization/depyrogenation treatments to control bioburden, avoid 
excessive pyrogens, and to sterilize. The selection of the specifi c process must always 
fully consider the impact of the treatment on the items being sterilized/depyrogenated. 
Sterilization and heat depyrogenation processes must balance the effect of 
the treatment on the microorganism with the effect of that same treatment on the 
materials being processed. The choice of one method over another is often based 
upon achieving the desired sterilization/depyrogenation effect with minimal impact 
on the items critical quality attributes. 
2.1.6.1 Steam Sterilization 
The method of choice in nearly every instance is moist heat due to its lethality, 
simplicity, speed, and general ease of process development and validation. For the 
majority of items, this is accomplished in a double - door steam sterilizer, which is 
conventionally located between the preparations and aseptic processing (fi lling or 
compounding) areas. Steam sterilizers are routinely utilized for items such as elastomeric 
closures, process and vent fi lters, product contact parts, heat stabile environmental 
monitoring equipment, tools and utensils, hoses, sample containers, and 
other items unaffected by contact with saturated steam at commonly used sterilizing 
temperature and pressure [19] . Similar items utilized in the nonsterile compounding 
area would be processed in a similar manner. Regardless of their fi nal 
destination or usage, items for steam sterilization should be protected from poststerilization 
contamination by materials that are permeable to steam, air, or condensate 
but impenetrable by microorganisms. The wrapping materials would be 
maintained on the sterilized items until just prior to use. There are numerous 
publications that provide additional details on steam sterilization procedures 
[19 – 21] . 
Sealed containers of aqueous solutions, suspensions, and other liquids can be 
processed through steam sterilizers as well. These liquids might be used in formulation 
or cleaning procedures, and sterilization in this manner may be more effi cient 
and more reliable than sterilizing fi ltration. Larger volumes of aqueous liquids are 
often sterilized in bulk using a jacketed and agitated pressure vessel (the vessel is 
usually rated for full vacuum as well). 
Steam SIP is a widely used practice for the sterilization of equipment prior to 
the introduction of process materials and is the method of choice for holding tanks, 
process transfer lines, lyophilizers, and other large items. Conceptually, it has many 
similarities to sterilization in autoclaves but differs markedly due to the often 
custom designs of process equipment requiring SIP. Systems must be designed with 
careful consideration given to air removal and condensate draining, process sequenc- 
STERILIZATION AND DEPYROGENATION 117

118 STERILE PRODUCT MANUFACTURING 
ing, and poststerilization integrity to assure success [22] . Terminal sterilization of 
fi nished product containers is addressed later in this chapter. 
2.1.6.2 Dry - Heat Sterilization and Depyrogenation 
The use of dry heat for depyrogenation (and sterilization) is almost universal for 
glass containers. Temperatures of 250 ° C or higher are utilized to render the glass 
endotoxin free. The depyrogenation is necessary because the washing of glass to 
reduce particles can introduce unacceptable levels of gram - negative microorganisms 
whose presence could result in pyrogen formation. The depyrogenation process 
can assist in component surface treatment (siliconization is required for some formulations) 
and will also render the glass sterile as well (depyrogenation temperature 
conditions far exceed those needed for sterilization [23] ). 
Sterilization by dry heat is only infrequently used, preference being given to the 
use of steam (due to its higher speed) or dry - heat depyrogenation (affording an 
added measure of safety using the same equipment). Where it is employed temperatures 
in the range of 170 – 180 ° C are employed, and a batch oven is customarily 
used. 
Dry - heat processes are conducted in either batch ovens or continuous tunnels, 
which are also installed between preparations and aseptic processing areas. Ovens 
have lower capacity and are typically found in smaller facilities. They offer the ability 
to handle items other than fi nal product containers and thus can replace autoclaves 
in facilities where fi lling parts, feed hoppers, tools, and other items that must be 
extremely dry. Ovens should be equipped with internal HEPA fi lters, recirculating 
fans, heating/cooling coils, and a sophisticated control system [24] . Items prepared 
for dry - heat treatment in ovens are inverted or covered to protect them after exiting 
from the oven as there are no sealed protective systems suitable for the higher 
temperatures necessary for dry - heat depyrogenation or sterilization. Oven discharge 
is typically into a cool - down area (usually the same as that used for the sterilizer), 
though in small facilities it might discharge directly into the fi ll room. Unless ovens 
are used in conjunction with isolators, they require direct operator intervention to 
transfer containers to the fi lling line and to charge the line with depyrogenated glass. 
This constitutes a risky intervention which should be avoided. For this reason, batch 
glass processing is rare in all but the lowest throughput facilities. 
Dry - heat tunnels are typically utilized where the production volumes are higher 
and allow for continuous supply of depyrogenated glass to the aseptic fi ll room. 
Tunnels are operated at high temperatures ( > 300 ° C) to increase processing speed 
and include a cooling zone that facilities discharge at or near room temperature. 
Typically, heating of the glass to 300 ° C or more for 3 or more minutes will result in 
much greater than the three - log endotoxin reduction required in current industry 
standards. The air inside the tunnel is HEPA fi ltered, and newer designs allow for 
dry - heat sterilization of the cooling zone as an added protective measure. Tunnels 
must be positioned with some care as they ordinarily will terminate into a fi ll room. 
A pressure differential between the cooling zone of the tunnel and the fi ll room is 
critical for proper operation of the tunnel. The pressure differential must conform 
to the requirements stipulated by the tunnel manufacturer. It is not necessary to 
have a > 12.5 PA (particulate air) differential between the in - feed side of the heating 
zone of the tunnel and the exit side of the cooling zone. It has been suggested by 

some that, since the in - feed side of the tunnel is typically in ISO 7 or 8 space, a 
greater differential is required; however, this is not true since the cooling zone is 
ISO 5, and the heating zone is certain to be sterile and is also ISO 5 in terms of 
particulate air quality. Their in - feed is often direct from a glass washer, which may 
be remote from the main preparations area utilized for washing, wrapping, and 
sterilizer loading. 
2.1.6.3 Gas and Vapor Sterilization 
The sterilization of materials using noncondensing gases (ethylene oxide, chlorine 
dioxide, or ozone) or condensing vapors such as hydrogen peroxide is a supplementary 
process intended for items that cannot be exposed to heat. The utilization of gas/ 
vapor designs is coming into increased use as a supportive technology for isolation 
technology for presterilized items such as syringes and stoppers that must be introduced 
into the isolators aseptic zone. Air locks using these agents can be utilized in 
similar fashion for the supply of materials to manned clean rooms. Control over agent 
concentration or injection mass, relative humidity, and temperature may be required 
for these systems. There are different types of vapor processes available, and users 
should generally follow the cycle development strategy suggested by the manufacturer 
of the equipment they have chosen. Specifi c temperature and humidity ranges 
may be required for some vapor processes to assure appropriate effi cacy [25, 26] . 
2.1.6.4 Radiation Sterilization 
The use of radiation within a parenteral facility would have been considered unthinkable 
prior to the start of the twenty - fi rst century. While . irradiation is typically a 
contracted service provided off - site, electron beam sterilization advances can make 
the installation of an in - house (and generally an in - line) system a real possibility. 
An in - line system would be utilized similarly to the gas/vapor systems described 
above for treatment of external surfaces for entry into either a clean room or 
isolator - based aseptic processing facility. The use of this same technology for terminal 
sterilization is also possible [1] . Association for the Advancement of Medical 
Instrumentation (AAMI)/ISO 11137 provides widely accepted guidance on the 
development and validation of radiation sterilization processes. 
2.1.6.5 Sterilization by Filtration 
Filters are utilized to sterilize liquids and gases by passage through membranes that 
retain microorganisms by a combination of sieve retention, impaction, and attractive 
mechanisms [27] . In contrast with the other forms of sterilization that are destructive 
of the microorganisms, fi lters rely on separation of the undesirable items (microorganisms 
as well as nonviable particles) from the fl uid. Because fi ltration requires 
passage of the fl uid from the “ dirty ” (upstream) side of fi lter to the clean (downstream) 
side of the fi lter, the downstream piping and equipment must be both 
“ clean ” and sterile prior to the start of the fi ltration process. This will ordinarily 
require the use of SIP procedures or sterilization followed by aseptic assembly. 
Sterilizing fi ltration of parenterals is a complex and often inadequately considered 
subject, and numerous controls are required on the fi lter, fl uid, and sterilizing/ 
STERILIZATION AND DEPYROGENATION 119

120 STERILE PRODUCT MANUFACTURING 
operating practices employed. PDA Technical Reports 26 and 40 can be instructive 
in understanding the relevant concerns [28, 29] . 
2.1.7 FACILITY AND SYSTEM: QUALIFICATION AND VALIDATION 
Facilities for the manufacture of sterile products require the qualifi cation/validation 
of the systems/equipment and procedures utilized for that production. Each system 
described above and others with a direct/indirect impact on the quality of the products 
being produced should be placed into operation using a defi ned set of practices. 
The general approach is described below, and best practices include the development 
of traceable documentation from project onset. The preferred approach begins 
during a project ’ s conceptual design phase where provisions for meeting the CGMP 
expectations and user requirement specifi cations establishing the technical basis for 
the processes are fi rst defi ned. This is commonly followed by the validation master 
planning exercise in which the user requirement specifi cations are used as a basis 
for the development of acceptance criteria for process control studies. This effort 
should be accompanied by an analysis of risk that considers product attributes, 
target patient population, as well as technical and compliance requirements. Detailed 
design follows in which the specifi cs of the various systems are refi ned. Construction 
of the facility and fabrication of the process equipment follows and a variety of 
controls are necessary during these activities to satisfy user requirements for compliance 
of the various elements of the facility. Typically, factory acceptance testing 
(FAT) will be done on all key process equipment, usually at the manufacturer ’ s 
plant site; much of the information gathered during FAT can be referenced in the 
qualifi cation activities to follow. Physical completion is followed by a well - defi ned 
step termed commissioning in which construction and fabrication errors and omissions 
are addressed. Site acceptance testing of installed process equipment may be 
done in parallel with facility commissioning. Formal qualifi cation of the facility 
ensues in which the installed systems and equipment are evaluated for their conformance 
to the design expectations. The very last steps in this process are variously 
termed performance qualifi cation. Detailed discussion of these subjects is not possible 
within the constraints of this chapter, however the qualifi cation/validation of 
equipment, systems, and processes has been extensively addressed in the literature 
[30] . 
2.1.8 ENVIRONMENTAL CONTROL AND MONITORING 
Confi rmation of appropriate conditions for aseptic processing and its supportive 
activities is required by regulation. In the highest air quality environment utilized 
for aseptic processing, ISO 5, there is a general expectation that the air and surfaces 
be largely free of microbial contamination and the number of particles be within 
defi ned limits (less than 3500 particles greater than 0.5 . m/m 3 ). Proving the complete 
absence of something is an impossible requirement, so the usual expectation is that 
99+% of all samples taken from this most critical environment be free of detectable 
microorganisms. The minimum monitoring expectations for these environments as 
defi ned by the regulators are consistently attainable in nearly all instances, 

especially those with lesser expectations. This is accomplished by proper design, 
periodic facility disinfection, and measures to control the ingress of microorganisms 
and particles for materials entering each environment from adjacent less clean areas 
[31] . 
2.1.8.1 Sanitization and Disinfection 
Disinfection is customarily performed by gowned personnel during nonoperating 
periods using such agents as phenolics, quaternary ammonium compounds, aldehydes, 
and other nonsporicidal agents. The frequency of treatment varies with the 
ability of the facility to maintain the desired conditions between disinfection. Sporicidal 
agents such as dilute hydrogen peroxide or bleach are reserved for those 
occasional periods when control over the spore population warrants and is often 
employed after lengthy maintenance shutdowns or at the end of construction. Isolation 
technology replaces the manual disinfection with reproducible decontamination 
with a sporicidal agent and thus assures a superior level of environmental 
control as compared to manned environments. The manual treatments fall short of 
this level of control due to the uncertainties of the manual procedure and recontamination 
of the environment as a consequence of the very personnel and activities 
utilized to disinfect it. To mitigate these weaknesses, automatic sporicidal disinfection 
of manned clean spaces has been developed by multiple vendors. Disinfection 
of the less critical environments is accomplished in the same manner albeit on a less 
frequent interval befi tting their higher allowable levels of microorganisms. 
2.1.8.2 Monitoring 
Aseptic environments are subject to a variety of monitoring systems including air, 
surface, and personnel monitoring for viable microorganisms and for nonviable 
particles. Environmental monitoring programs are often developed during the qualifi 
cation of a new facility using a multiphase approach. Methods for the monitoring 
and expectations for performance have been extensively discussed in the literature 
and will only be addressed briefl y in the context of this chapter [1, 2, 31, 32] . In 
general, the frequency and intensity of monitoring and concern for cleanliness 
increases as the product progresses from preparation steps (typically in ISO 7/8 
environments) to more important activities (nonsterile compounding in ISO 6) and 
ultimately into the aseptic core (aseptic compounding and fi lling in ISO 5). Sampling 
site and time selection should be a balance between the need to collect meaningful 
data and avoidance of sampling interventions that could adversely (and inadvertently) 
impact product quality. Microbiological sampling must always be done by 
well - trained staff utilizing careful aseptic technique. This will both minimize risk to 
the product and also improve the reliability of the data by reducing the likelihood 
of false - positive results. 
Air Sampling The relative cleanliness of air in the most critical environment is 
assessed using passive sampling systems such as settle plates or estimated volumetrically 
using active air samplers. Active air samplers should be designed to be isokinetic 
in operation to avoid disruptions to unidirectional airfl ow. Considerable 
variability has been reported among the several sampling methods employed for 
ENVIRONMENTAL CONTROL AND MONITORING 121

122 STERILE PRODUCT MANUFACTURING 
active air sampling, and there are also reports that active air sampling may have 
advantages in terms of sensitivity. Passive sampling using settle plates can be a useful 
adjunct in critical areas with limited access and where an active sampler might 
interfere with airfl ow or entail a worrisome intervention risk. It must be recognized 
that attempts to support the “ sterility ” of the cleanest aseptic environments (those 
in ISO 5) by aggressive sampling may have exactly the opposite effect. Sampling 
too frequently will increase process contamination risk by causing critical interventions 
that are best avoided within these very clean environments. As personnel are 
the greatest single source of microbial contamination and conduct the sampling, 
sampling intensity should be carefully considered. There is no value to taking air 
samples beyond those required to assess the relative cleanliness level within the 
environment. 
Surface Sampling Surfaces in the classifi ed environments are monitored using a 
variety of methods but most commonly with contact plates (on smooth surfaces) or 
swabs (for irregular surfaces). Surface sampling in aseptic environments (ISO 5/6) 
is typically performed after the completion of the process to avoid the potential for 
adventitious contamination of the production materials as a consequence of sampling 
activities during the process. Fortunately, studies indicate that contamination 
does not build up during typical processing operations in modern clean rooms. 
Sampling with these materials may leave a trace of media or water on the sampled 
surface, and cleaning of the surface immediately after sampling is commonplace. 
Sampling of product contact surfaces (i.e., fi ll needles, feeder bowls, etc.) should only 
be performed after completion of the process, and the results of this testing should 
not be considered as an additional sterility test on the products. As in any form of 
manual environmental sampling, the risk of contamination by samplers during the 
processing of a sample makes the data less than completely reliable. Sampling of 
surfaces such as walls and fl oors should not be overdone because with good attention 
to aseptic technique they should be of little concern relative to actual process 
risk. Sampling on these surfaces is probably most useful in assessing ongoing changes 
in microfl ora and to confi rm the adequacy of the disinfection program. 
Personnel Sampling The monitoring of personnel gown surfaces is an adaptation 
of surface sampling in which samples are taken from surfaces on the operator. In 
ISO 5 environments, this ordinarily entails the gloved hands and perhaps forearms. 
As with any other sampling of a critical surface (the gloved hand is often in closest 
proximity to sterile product contact surfaces and sterilized components), the sampling 
should be performed at the conclusion of the aseptic activity. Sampling during 
the midst of the process risks contamination of the product and should be avoided. 
Sampling of other aseptic gown surfaces is ordinarily restricted to gowning certifi cation 
or postmedia fi ll testing, where more aggressive sampling can sometimes be 
informative. Whenever a gowned individual is sampled, the sample should be taken 
in the background environment (not ISO 5), and the individual should immediately 
exit and regown before continuing any further activity in the aseptic core area. 
Sampling of personnel in less critical environments can be useful; however, meeting 
regulatory expectations in these areas is ordinarily straightforward. Recommended 
contamination levels often distinguish among the different room classifi cation levels 
found within clean rooms. While this may seem reasonable, it is not completely 

logical since operators often move frequently between these different levels of classifi 
cation during the conduct of their work. 
Total Particulate Monitoring Confi rming the ability of the facility ’ s heating, ventilation, 
and air - conditioning (HVAC) system to maintain the appropriate conditions 
throughout (to the extent practical) the classifi ed environments is most easily 
accomplished using electronic total particle counters that can provide near immediate 
feedback on conditions during production operations. Total particle samples can 
be taken automatically, using permanently installed probes oriented into the unidirectional 
airfl ow. As such, they can be positioned proximate to critical activities to 
reaffi rm the continued quality of the air in the vicinity of the sterile materials and 
surfaces. Manual total particulate air sampling can be a dangerous intervention and 
therefore if required should be timed so as to minimize risk to product. Attempts 
to correlate total particle counts with microbial counts have proven diffi cult. Correlations 
are only meaningful when the source of foreign material is personnel since 
people are the only source of airborne contamination within an aseptic processing 
area. When personnel are the only source of particulate, the ratio between viable 
and nonviable particles have been consistently found to be > 1000 : 1, which means 
that in ISO 5 environments even relatively large total particulate count excursions 
would typically contribute microbial contamination that fell far below the limit of 
detection. Process equipment can and often does contribute airborne particulate 
matter but not detectable levels of microbial contamination. Also, microbial sampling 
is highly variable with respect to sensitivity, accuracy, precision, and limit of 
detection making correlations, particularly in rooms of highest air quality. So, it 
might seem logical to think that particle excursions are indicative of coincident 
microbial excursions especially in the cleaner environments (ISO 5) where the 
aseptic process takes place. 
It is common practice for fi rms to interrupt their aseptic processes when atypical 
total particulate excursions are observed so that the scientists and engineers can 
determine the source of the foreign material. Monitoring frequency and expectations 
in the less critical environments is always reduced relative to the critical aseptic 
environments. 
Where fi rms have introduced unidirectional air systems in preparations and 
compounding areas for particle control, there is often the temptation to expect these 
areas to meet the same microbial limits that these locations might attain in the 
aseptic core. This temptation should be resisted to avoid unnecessary sampling and 
deviations associated with expecting these environs to meet the conditions of aseptic 
areas where sanitization frequency, background environment, and most importantly 
personnel gowning are far superior to that found in the less clean locales [33] . 
Housekeeping An important component of environmental control are the housekeeping 
activities utilized to clean the facility external to the controlled environments. 
Aseptic operations utilize a series of protective environments to protect the 
sterile fi eld. Controls on the surrounding unclassifi ed areas are an important part 
of the overall control scheme for sterile manufacturing. These unclassifi ed areas 
support sterile operations in a variety of ways, and it is important to conduct activities 
therein that assist in the environmental control. Routine housekeeping, periodic 
sanitization, and even occasional environmental monitoring may be appropriate to 
ENVIRONMENTAL CONTROL AND MONITORING 123

124 STERILE PRODUCT MANUFACTURING 
assure that microbial and particle loads on items, equipment, and personnel entering 
the classifi ed environments is appropriately controlled. 
2.1.9 PRODUCTION ACTIVITIES 
The preparation of sterile materials requires execution of a number of supportive 
processes that together constitute the manufacturing process. They are intended to 
control bioburden, reduce particle levels, remove contaminants, sterilize, and/or 
depyrogenate. Nearly all of these activities occur within the controlled environments 
and are subject to qualifi cation/validation. 
2.1.9.1 Material and Component Entry 
Prior to the start of any production activity, materials and components must be 
transferred from a warehouse environment into a classifi ed environment. For most 
items this will necessitate removal from boxes or cartons, transfer to a nonwooden 
pallet, and passage through an air lock which serves as the transfer system between 
the controlled and uncontrolled environments. Often components are contained 
within plastic bags within a box or carton, and in some cases there are multiple bag 
layers to facilitate disinfection and passage through air locks into different zones of 
operation within the aseptic area. The fi rm may utilize an external disinfection 
of the materials in conjunction with this transfer. The concern is for minimization 
of particles and bioburden on these as yet unprocessed items in order to protect the 
controlled environment. 
Raw materials may be weighed in a weigh area in which they are transferred to 
plastic bags and/or noncorrugate containers prior to the transfer. The weighing area 
provides ISO 7 or better conditions, and may be a dedicated portion of the warehouse 
proper; in a central weighing/dispensing area; or in a location contiguous to 
the compounding area. Sterile ingredients are never opened anywhere other than 
an aseptic environment and must be handled aseptically at all times including sampling 
and processing of samples. 
2.1.9.2 Cleaning and Preparation 
Once the container component items have been introduced into the preparations 
area, they must be readied for sterilization/depyrogenation. For many items this 
consists of washing/rinsing processes designed to remove particles and reduce bioburden 
and endotoxin levels. The application of silicone suspensions for glass or 
closure materials is sometimes employed to provide lubrication allowing smoother 
feeding of components or dispensing (elimination of product accumulation on vial). 
Following the cleaning, items for sterilization are dried, wrapped, and staged/stored 
for steam sterilization. Washed containers are either placed in trays or boxes for 
depyrogenation in ovens or are directly loaded into dry - heat tunnels. It is common 
practice to protect all washed items with ISO 5 air from the completion of washing, 
through either wrapping or placement into a sterilizer or oven for passage into the 
aseptic area. The intention is to avoid foreign matter that could result in contamination 
of product. 

It is increasingly common for components to be supplied by the vendor in a 
ready - to - sterilize condition (washed and pretreated as necessary). Some items are 
available in a ready - to - use confi guration with the supplier providing sterile and 
pyrogen - free components. The use of supplier - prepared items eliminates the need 
for preparation activities at the fi ll site and requires modifi cation of material in - feed 
practices relative to on - site prepared items. 
The process equipment (portable tanks, valves, fi ll needles, etc.) and consumable 
materials (fi lters, hoses, gaskets, etc.) are prepared using a variety of methods. Portable 
tanks are subjected to CIP (and perhaps SIP as well) in the preparation area. 
Smaller items are disassembled (if necessary) and cleaned either manually or in a 
cabinet washer. After cleaning they are wrapped and staged/stored prior to sterilization. 
Tubing should not be reused; its preparation typically consists of fl ushing with 
WFI followed by cutting to the required length. It is best to preassemble fi ll sets 
with tubing, fi lters, and fi ll needles/pumps and then wrap them in preparation for 
sterilization. This process obviates poststerilization assembly steps and therefore 
mitigates contamination risk. These steps may be performed in ISO 5 environments 
to reduce total particulate contamination on the items. 
There are items that must be transferred into the aseptic processing area that 
cannot be treated within a sterilizer/oven. These include portable tanks, electronic 
equipment, and containers of sterile materials (ready - to - use items, sterile powders, 
environmental monitoring media, etc.). Air locks, pass - throughs, and similar designs 
are employed in which the exterior surfaces of the items are disinfected. The disinfection 
process may be completed by personnel outside and/or inside the aseptic 
area depending upon the specifi cs of the design. 
At the completion of the cleaning process, the items should be free of contaminating 
residues including traces of prior products, free of endotoxin, and well - controlled 
in terms of total particulate and microbial levels. This level of control would be 
appropriate regardless of whether the items, equipment, or components are to be 
sterilized or not. Sterilization, other than by relatively high temperature dry heat, has 
only a modest impact on endotoxin levels; cleaning provides the only means to 
control endotoxin for materials and equipment that is sterilized by other means. 
2.1.9.3 Compounding 
Fixed equipment in the compounding area (nonaseptic or aseptic) is cleaned in 
place. This eliminates traces of prior products, particles, and pyrogens. Sterilization 
in place is required for the aseptic fi xed equipment and is sometimes employed for 
the nonaseptic equipment as well as a bioburden control measure. Fixed transfer 
lines must be cleaned and sterilized as well, and this is accomplished independently 
or in conjunction with the vessels. The reuse of hoses and tubing is discouraged as 
cleaning and extractables cannot be confi rmed beyond a single use. 
The preparation of the product is performed within a classifi ed environment with 
careful attention to the batch record, especially for time limits and appropriate 
protection of materials during handling to guard against all forms of contamination. 
This is proper for nonsterile compounding to minimize contamination prior to fi ltration/
sterilization and is required for aseptic compounding activities. Barrier designs 
and other means of physically separating the worker from the product are recommended 
as a minimum even in nonaseptic compounding. As compounding may 
PRODUCTION ACTIVITIES 125

126 STERILE PRODUCT MANUFACTURING 
expose the worker to a variety of potent/toxic materials, the use of personnel protective 
equipment may be required. In extreme cases, the use of containment system 
may be required to protect the compounding operator. 
Where the compounding is nonaseptic, careful control over the environment, 
materials, and equipment is still appropriate to reduce viable/nonviable levels and 
to reduce the potential for endotoxin. Time limits should be imposed on manufacturing 
operations for additional control over microorganisms and thus microbial 
toxins. 
Once the materials have been sterilized, interventions near either the formulation 
or product contact surfaces/parts should be minimized. Direct handling of these 
materials should only be done with sterilized tools or implements; nonsterile objects, 
such as operator gloves, should never directly contact a sterilized surface. Sampling, 
fi lter integrity testing, process connection, and other activities should all be designed 
to eliminate the need for personnel exposure to sterile items. 
Aseptic compounding is often a required activity for sterile products that cannot 
be fi lter sterilized. The preparation of the sterile solids for use in these formulations 
is outside the scope of this chapter, but it is often acknowledged as the most diffi cult 
of all pharmaceutical processes to properly execute. Handling these materials at the 
fi ll site is performed using ISO 5 environments, and the use of closed systems is 
preferred [34] . 
2.1.9.4 Filling 
Aseptic fi lling is performed in ISO 5 environments, and a variety of approaches are 
utilized with the technology choice largely dependent upon the facility design, batch 
size, and package design. Older plants utilize manned clean rooms in which 
aseptically gowned personnel operate the fi lling equipment: performing the setup, 
supplying components, making any required adjustments, and conducting the environmental 
monitoring. As human operators are directly or indirectly responsible 
for essentially all microbial contamination, aseptic fi lling operations are increasingly 
designed to minimize the potential for operator contamination to enter the critical 
environment. Barriers of various sophistication and effectiveness are employed to 
increase the protection afforded to sterile materials. The most evolved of the clean - 
room designs are RAB systems in which personnel interventions are restricted to 
defi ned locations. Many newer facilities utilize isolation technology in which the 
fi lling environment is fully enclosed and personnel contamination is completely 
avoided. 
Filling designs for syringes and ampoules differ only with respect to the details 
of component handling and closure design. However, it is wise not to underestimate 
the infl uence of both component quality and component handling reliability on 
contamination control in aseptic processing. Components that minimize the need 
for intervention and equipment that is rather tolerant of component variability will 
result in better contamination control performance. Aside from these distinctions, 
the range of fi lling technologies previously described is also possible. 
The fi lling of plastic containers is accomplished using two very different 
approaches. Pre - formed containers can be sterilized in bulk, introduced into the 
aseptic suite via air locks, oriented (unscrambled), and fi lled. Blow-fi ll - seal prepares 
sterile bottles (most often LDPE) on line just prior to fi lling and sealing. 

Filling of suspensions, emulsions, and other liquids may require slightly different 
fi lling designs to assure uniformity of dose in each container. Ointments and creams 
are sometimes fi lled at elevated temperatures to improve their fl ow properties 
through the delivery and fi lling equipment. These are ordinarily fi lled into presterilized 
plastic tubes that have largely replaced aluminum tubes for these formulations. 
Powders are typically fi lled in vials using equipment specifi cally engineered for that 
purpose. 
An inerting gas (typically nitrogen, but other gases can be utilized) may be added 
to the headspace of the container to protect formulations that are oxygen sensitive. 
If the product is particularly sensitive to oxygen, purging may be done in the empty 
container prior to fi lling and again immediately after fi lling. Products may also be 
fi lled in an isolator under a nitrogen atmosphere if required. Products that require 
inert gas purging will also generally require inert gas for pressurization of tanks to 
provide motive force to drive the product through the fi lter(s) and into the fi lling 
reservoir. 
2.1.9.5 Stoppering and Crimping 
If the product is not freeze dried, the primary closure or “ stopper ” is applied shortly 
after completion of the fi lling process to better assure the sterility of the contents. 
When the product is to be lyophilized, the stopper may be partially inserted after 
fi lling and be fully seated after completion of the lyophilization cycle. Alternatively, 
the container could be left open and a stopper applied after completion of the 
drying. 
Crimping is the act of securing the closure to the vial. It must be performed with 
suffi cient uniform downward force to assure the container is properly secured. Too 
little downward force results in inadequately secured closures, while excessive force 
can result in container breakage. The force contributed by the crimp roller may be 
controllable as well. 
Applying the closure to syringes, ampoules, and other containers usually differs 
in methodology from the approaches used for vials, but the objective is identical to 
secure the container ’ s contents fully assuring the product ’ s critical quality attributes 
(especially sterility) are maintained throughout its shelf life. 
2.1.9.6 Lyophilization 
Lyophilization (or freeze - drying) is a process utilized to convert a water - soluble 
material fi lled into a container to a solid state by removal of the liquid while frozen. 
The process requires the use of deep vacuums and careful control of temperatures. 
By conducting the process under reduced pressure, the water in the container converts 
from ice directly to vapor as heat is applied and is removed from the container 
by the vacuum. The dissolved solids in the formulation cannot undergo this phase 
change and remain in the container. At the completion of the cycle, the container 
will be returned to near atmospheric pressure; stoppers are applied or fully seated 
and crimped as described above. Lyophilization is particularly common with biological 
materials whose stability in aqueous solution may be relatively poor. The time 
period in solution and the temperature of the solution are kept at a specifi ed low 
temperature to prevent product degradation [35] . 
PRODUCTION ACTIVITIES 127

128 STERILE PRODUCT MANUFACTURING 
As partially stoppered but unsealed containers must be transferred to the 
lyophilizer from the fi ll line, various designs have been utilized to protect the containers 
during this transit. Among the common alternatives utilized are the 
following: 
• Placement of the lyophilization in the wall of the fi ll room to allow for direct 
loading 
• Battery - operated unidirectional airfl ow carts to a remote lyophilizer 
• ISO 5 – protected conveyors with single shelf loading 
• Transfer utilizing isolator technology 
The use of trays for supporting the containers during the transfer, loading, lyophilization, 
and unloading steps was at one time common. The major problem with the 
use of trays for this purpose was the heat/handling - related distortion of the tray 
bottom that impacted the uniformity of the heating process in the freeze dryer. This 
was overcome by the use of trays with bottoms that were removed after loading 
and reinserted after completion of the drying. The current preference is for the 
placement of the containers directly on the shelf eliminating the trays entirely. This 
is accomplished by single height loading/unloading of the individual shelves with 
various pusher designs. 
The use of thermocouples to monitor product temperature inside selected 
vials with the lyophilizer is still the prevalent practice. The utility of this data is 
questionable and the current trend is to eliminate this “ requirement ” as soon as 
possible to better assure sterility of the unsealed vials by eliminating placement of 
the thermocouples. 
The lyophilizer chamber and condenser should be cleaned with a CIP system 
after each batch to prevent cross - contamination and, after cleaning, both should be 
sterilized. If a slot door loading system is utilized, periodic opening of a full door in 
the lyophilizer may be required to remove stoppers and glass that may have 
fallen. 
2.1.10 PERSONNEL 
Aseptic processing in the pharmaceutical industry is almost entirely dependent 
upon the profi ciency of the personnel assigned to this most critical of all activities. 
The operators must be able to consistently aseptically transfer sterile equipment 
and materials in a manner that avoids contamination of those materials [1] . This is 
no mean feat given the contamination continuously released by personnel and the 
prevailing need for personnel for execution of the process activities. 
Personnel profi ciency in aseptic operations must be fi rmly established before 
they are allowed to conduct critical aseptic process steps. Operators must master a 
number of relevant skills in order to be declared competent. The usual progression 
is from classroom training (CGMP, microbiology, sterilization, etc.) to relevant 
practical exercises (aseptic media transfers, aseptic gowning rehearsals) and ultimately 
to the core aseptic skills required (aseptic gowning certifi cation, aseptic 

assembly/technique) using a growth medium. Through this approach the operator 
gradually acquires the necessary skills to be a fully qualifi ed member of the production 
staff. Training/qualifi cation of personnel is an ongoing requirement and must 
be repeated periodically to assure the skills are maintained. Continuing evaluation 
of operator qualifi cation is accomplished using written examinations, practical challenges, 
documented observation, and participation in process simulation trials. 
There is general acknowledgment of the risk associated with heavy reliance on 
personnel for aseptic processing. This has fostered much of the innovative designs 
for aseptic fi lling such as RABS and isolators where personnel are largely removed 
from the critical environment. The future will undoubtedly witness aseptic technologies 
where human interaction with sterile materials has been eliminated. 
2.1.11 ASEPTIC PROCESSING CONTROL AND EVALUATION 
The preparation of any pharmaceutical product requires controls over the production 
operations to assure the end result is a product that meets the required quality 
attributes. The methods utilized for this control are supported by formalized validation 
studies in which proof of consistency is demonstrated by appropriately designed 
experiments. The defi nition of appropriate operating parameters is the primary 
objective of the development activities and is further confi rmed during scale - up to 
commercial operations. The validation supports that the routine controls applied to 
the process are appropriate to assure product quality [36] . This is typically accomplished 
in formalized validation activities in which expanded sampling/testing of the 
product materials is performed to substantiate their uniformity and suitability for 
use [30] . 
2.1.11.1 In - Process Testing 
The sampling and testing of in - process materials during the course of the manufacturing 
process can confi rm that essential conditions have been provided. This is 
appropriate in preparation, compounding, and fi lling activities. Sampling in preparation 
processes can confi rm the absence of particles, proper siliconization levels, and 
cleanliness of equipment to assure that production items and equipment are suitable 
for use. Samples for microbiological quality, must, as previously mentioned, always 
be done by fully gowned staff under ISO 5 conditions using excellent aseptic techniques. 
During compounding, in - process testing can confi rm proper pH, dissolution 
of materials, bioburden, and potency prior to fi lling. Filling operations can be monitored 
for fi ll volume (weight), headspace oxygen, and particles. These activities can 
all be automated to reduce interventions. These are typical examples of in - process 
controls utilized to assure acceptability of the process while it is underway. In the 
event of an abnormal result, corrective measures could be applied before further 
processing. The validation effort supports that these control measures are suffi cient 
to assure product quality, when met during production operations. The sample 
intervals, sizes, and locations for in - process testing are chosen to enhance the validation. 
The tolerance limits are usually tightened relative to the release requirements 
to further assure that no out - of - tolerance materials are produced. 
ASEPTIC PROCESSING CONTROL AND EVALUATION 129

130 STERILE PRODUCT MANUFACTURING 
2.1.11.2 End - Product Testing 
Upon completion of the process, samples are taken to establish that the batch meets 
the fi nal product specifi cations defi ned for release. Predefi ned sampling plans are 
utilized to obtain representative samples of the entire batch, the prior validation 
effort having assured through an expanded sampling effort that the process provides 
a uniform product. End - product sampling often suffers from the inability to link an 
anomalous result with a specifi c portion/segment of the batch. If the validation is 
insuffi ciently rigorous, an out - of - specifi cation result will ordinarily result in rejection 
of the batch and little opportunity to take effective corrective action. 
The FDA has been supportive of the use of process analytical technologies 
(PATs) as an improvement on end - product testing [37] . These are intended to act 
as on - line indicators of critical product attributes enabling immediate corrective 
action and preventing the production of off - specifi cation materials. This approach 
is common in the continuous process industries where feedforward controls are 
often employed. Their application to the more batch - oriented pharmaceutical/biotechnology 
industry is an acknowledgment that this approach can assure product 
quality more fully than a sampling - based approach. The PAT applications are still 
relatively few in number, but their utility in lieu of traditional quality methods is 
certainly promising. 
The preceding relates solely to product quality attributes, based upon chemical 
or physical requirements. Assurance of sterility, the most critical of all the quality 
components for an aseptically fi lled sterile product relies on the following: 
• The validation of the various sterilization processes for preparation of materials, 
equipment, and formulations 
• The design of the aseptic manufacturing process and facility 
• The establishment and maintenance of a proper processing environment 
• Most importantly, the profi ciency of the operating personnel directly involved 
with the aseptic process 
There is no direct means to evaluate the cumulative capability of these measures. 
We infer success in aseptic processing through the evaluation of indirect measures 
of performance: air pressure differentials, total particle counts, viable monitoring 
results, and end - product sterility testing. The enormous challenge of aseptic processing 
is that none of the in - process or end - product testing results can prove that the 
attribute of sterility is attained with a high degree of certainty. Therefore, we rely 
on validation and the demonstration of a validated state of control to infer the 
adequacy of our contamination control efforts. 
2.1.11.3 Process Simulations 
An indirect means of assessing a facility ’ s aseptic processing performance is the 
process simulation (or media fi ll) test [38] . This test substitutes a growth medium 
for the product in the process from the point of sterilization through to closure of 
the product container. The expectation is that successful handling of the growth 
media through the operating steps provides assurance that product formulations 
handled in a similar fashion would also be successful [39] . Process simulations 

culminate in the incubation of the media - fi lled containers with success defi ned as a 
limited number of contaminated units in a larger number of fi lled units. The result 
is a contamination rate for the media fi ll, and not a direct indication of the level of 
sterility assurance afforded to aseptically processed materials using the same procedures. 
At the present time, the level of sterility provided to aseptically processed 
materials cannot be measured. The FDA and EMEA have harmonized their expectations 
relative to process simulation performance, but they have also asserted that 
the goal in every process simulation is zero contamination [1, 2] . This formalized 
expectation and recognition that patient safety should always be preeminent have 
resulted in substantial improvements in aseptic processing technology over the last 
20 years. 
2.1.12 TERMINAL STERILIZATION 
Terminal sterilization is a process by which product is sterilized in its fi nal container. 
Terminal sterilization is the method of choice for products that are suffi ciently 
stabile when subjected to a compatible lethal treatment. Because the process utilized 
is expected to be lethal to the microorganisms present, is highly reproducible, 
and generally readily validated, there is a clear preference for its use [1, 40, 41] . 
The predominant method for terminal sterilization is moist heat, and a substantial 
percentage of sterile products are processed in this manner. (Estimates range 
from 5 to 15% of all sterile products are terminally sterilized.) The sterilization often 
requires the attainment of a balance between sterility assurance and degradation of 
the material ’ s essential properties [42] . The overkill sterilization method is preferred 
for heat - resistant materials, and may be usable for terminal sterilization where the 
formulation can tolerate substantial heat input. The bioburden/biological indicator 
approach uses less heat input but requires increased control over the titer and 
resistance of the bioburden organisms present. 
The large - volume parenteral (LVP) industry sometimes uses dedicated nonaseptic 
fi lling systems for its containers prior to subjecting them to terminal treatments. 
These LVP systems may approach the aseptic designs described earlier, but they are 
not supported by the same levels of environmental monitoring nor process simulation. 
Application of terminal sterilization at small volume parenteral producers may 
be done after the product is aseptically fi lled, although this practice is usual only 
where the fi rm produces predominantly aseptically fi lled products and would not 
have a fi lling system dedicated to terminally sterilized formulations. Product 
that will be subject to terminal sterilization may be fi lled under clean conditions 
with reduced environmental monitoring and control. However, control of total 
particulate levels requires unidirectional airfl ow for critical fi lling or assembly 
processes. 
Terminal sterilization is most commonly accomplished by moist heat. Terminal 
sterilization by other means is certainly possible, and a very limited number of parenteral 
drugs are treated with dry heat or radiation after fi lling. There is growing 
interest in the use of radiation, including low - energy E - beam, as a terminal treatment 
suggesting more products will be processed in this manner. 
Although there are numerous advantages to terminal sterilization, there can be 
very good reasons for aseptically fi lling products that are stabile enough to be com- 
TERMINAL STERILIZATION 131

132 STERILE PRODUCT MANUFACTURING 
patible with a sterilization process. For example, multichamber containers that 
cannot withstand terminal sterilization may provide a very important safety benefi t 
to the patient by reducing aseptic admixture or reconstitution in the clinic. These 
aseptic activities when conducted in clinics are generally not able to be done within 
anything like the controls required in industrial aseptic processing. It is often benefi - 
cial to discuss processing technology choices with regulatory authorities early in the 
development of a new product. 
2.1.13 CONCLUSION 
The manufacture of parenteral drugs by aseptic processing has long been considered 
a diffi cult technical challenge. These products require careful control and stringent 
attention to detail to assure their safety. Aseptic processing done with discipline and 
taking advantage of the numerous technical developments that have occurred over 
the years results in sterile products that can be administered with complete confi - 
dence. The wider adaptation of advanced aseptic processing will result in further 
evolutionary improvements in aseptic processing. The industry is at the beginning 
of an era in which human - scale aseptic processing will be completely replaced by 
separative technologies and process automation. Additionally, improved in - process 
controls are likely to be implemented making validation easier and easing the compliance 
burden. 
APPENDIX 
Parenteral Drug Association, Bethesda, Maryland 
TM 1: Validation of Steam Sterilization Cycles, 1978 
TR 3: Validation of Dry Heat Processes used for Sterilization & Depyrogenation, 
1981 
TR 7: Depyrogenation, 1985 
TR 11: Sterilization of Parenterals by Gamma Irradiation, 1988 
TR 13: Fundamentals of an Environmental Monitoring Program, 2001 
TR 22: Process Simulation Testing for Aseptically Filled Products, 1996 
TR 26: Sterilizing Filtration of Liquids, 1998 
TR 28: Process Simulation Testing for Sterile Bulk Pharmaceutical Chemicals, 
2006 
TR 34: Design & Validation of Isolator Systems for the Manufacture & Testing 
of Health Care Products, 2001 
TR 36: Current Practices in the validation of Aseptic Processing, 2002 
TR 40: Sterilizing Filtration of Gases, 2005 
International Society For Pharmaceutical Engineering, Tampa, Florida 
Baseline Guide, Vol. 3: Sterile Manufacturing Facilities, 1999 
Baseline Guide, Vol. 4: Water and Steam Systems, 2001 
Baseline Guide, Vol. 5: Commissioning and Qualifi cation, 2001 

REFERENCES 
1. U.S. Food and Drug Administration (FDA) ( 2004 ), Guideline on sterile drug products 
produced by aseptic processing, FDA, Washington, DC. 
2. European Union (EU) (2006), Annex 1—Sterile medicinal products—draft revision. 
3. International Organization for Standardization (ISO) , international standard 14644 1 - 3. 
4. U.S. Food and Drug Administration (FDA) ( 2004 ), Pharmaceutical CGMPs for the 
twenty - fi rst century —A risk-based approach, FDA, Washington, DC. 
5. International Conference on Organization (ICH) ( 2005 ), Draft consensus guideline 
quality risk management Q9, draft. 
6. Whyte , W. , and Eaton , T. ( 2004 ), Microbiological contamination models for use in risk 
assessment during pharmaceutical production , Eur J Parenteral Pharm Sci , 9 ( 1 ). 
7. Whyte , W. , and Eaton , T. ( 2004 ), Microbial risk assessment in pharmaceutical cleanrooms , 
Eur J Parenteral Pharm Sci , 9 ( 1 ). 
8. Agalloco , J. , and Akers , J. ( 2006 ), Simplifi ed risk analysis for aseptic processing: The 
Akers - Agalloco method , Pharm Technol , 30 ( 7 ), 60 – 76 . 
9. International Organization for Standardization (ISO) ( 2004 ), Cleanrooms and associated 
controlled environments — Part 7: Separative devices (clean air hoods, gloveboxes, isolators 
and mini - environments), ISO 14644 - 7 . 
10. Agalloco , J. ( 2006 ), Thinking inside the box: The application of isolation technology for 
aseptic processing , Pharm Technol ., p. S8 – 11 . 
11. Lysford , J. , and Porter , M. ( 2003 ), Barrier isolators history and trends , Pharm Eng , 23 ( 2 ), 
58 – 64 . 
12. ISPE ( 2005 ), Restricted access barrier systems (RABS) for aseptic processing, ISPE defi - 
nition, Aug. 16. 
13. Wikol , M. ( 2004 ), GoreTM vial isolator, ISPE presentation, Feb. 12. 
14. Py , D. ( 2004 ), Development challenges for intact sterile fi lling, PDA presentation, 
Mar. 9. 
15. Thilly , J. ( 2004 ), CVFL technology from lab scale to industry, PDA presentation, Mar. 8. 
16. ISPE (2001), Water and Steam Systems Baseline® guide. 
17. ISPE ( 1999 ), Sterile Manufacturing Facilities Baseline ® guide. 
18. ISPE (2001), Water and Steam Systems Baseline® guide. 
19. PDA ( 2006 ), Technical Monograph 1, Industrial moist heat sterilization in autoclaves, 
draft 17. 
20. Perkins , J. ( 1969 ), Principles and Methods of Sterilization in Health Sciences , Charles 
Thomas , Springfi eld, IL . 
21. Phillips , G. B. , and Morrissey , R. F. ( 1993 ), Sterilization Technology: A Practical Guide for 
Manufacturers and Users of Health Care Products , Van Nostrand Reinhold , New York . 
22. Agalloco , J. ( 1998 ), Sterilization in place technology and validation , in Agalloco , J. , and 
Carleton , F. J. , Eds., Validation of Pharmaceutical Processes: Sterile Products , Marcel 
Dekker , New York . 
23. PDA ( 1981 ), Technical Report 3, Validation of dry heat processes used for sterilization 
and depyrogenation. 
24. Case , L. , and Heffernan , G. ( 1998 ), Dry heat sterilization and depyrogenation: Validation 
and monitoring , in Agalloco , J. , and Carleton , F. J. , Eds., Validation of Pharmaceutical 
Processes: Sterile Products , Marcel Dekker , New York . 
25. Burgess , D. , and Reich , R. ( 1993 ), Industrial ethylene oxide sterilization , in Phillips , G. B. , 
and Morrissey , R. F. Eds., Sterilization Technology: A Practical Guide for Manufacturers 
and Users of Health Care Products , Van Nostrand Reinhold , New York . 
REFERENCES 133

134 STERILE PRODUCT MANUFACTURING 
26. Sintim - Damao , K. ( 1993 ), Other gaseous sterilization methods , in Phillips , G. B. , and Morrissey 
, R. F. Eds., Sterilization Technology: A Practical Guide for Manufacturers and Users 
of Health Care Products , Van Nostrand Reinhold , New Youk . 
27. Meltzer , T. , Agalloco , J. , et al. ( 2001 ), Filter integrity testing in liquid applications ; Revisited, 
Part 1, Pharm Technol , 25 ( 10 ), and Part 2, Pharm Technol , 25 ( 11 ). 
28. PDA (1998), Technical Report 26, Sterilizing fi ltration of liquids. 
29. PDA (2005), Technical Report 40, Sterilizing fi ltration of gases. 
30. Agalloco , J. , and Carleton , F. J. , Eds. ( 1998 ), Validation of Pharmaceutical Processes: Sterile 
Products , Marcel Dekker , New York . 
31. PDA (2001), Technical Report 13, Fundamentals of an environmental control program. 
32. USP . 1116 . ( 2005 ), Microbiological control and monitoring environments used for the 
manufacture of healthcare products , Pharm Forum , 31 ( 2 ), Mar. – Apr. 
33. Agalloco , J. ( 1996 ), Qualifi cation and validation of environmental control systems , PDA 
J Pharm Sci Technol , 50 ( 5 ), 280 – 289 . 
34. PDA ( 2006 ), Technical Report 28, Process simulation testing for sterile bulk pharmaceutical 
chemicals. 
35. Trappler , E. ( 1998 ), Validation of lyophilization , in Agalloco , J. , and Carleton , F. J. , Eds., 
Validation of Pharmaceutical Processes: Sterile Products , Marcel Dekker , New York . 
36. Chapman , K. G. ( 1984 ), The PAR approach to process validation , Pharm Technol , 8 ( 12 ), 
22 – 36 . 
37. Food and Drug Administration (FDA) ( 2004 ), PAT guidance for industry — A framework 
for innovative pharmaceutical development, manufacturing, and quality assurance, FDA, 
Washington, DC. 
38. PDA ( 1998 ), Technical Report 22, Process simulation testing for aseptically fi lled 
products. 
39. Agalloco , J. , and Akers , J. ( 2006 ), Aseptic processing for dosage form manufacture: Organization 
& validation , in Carleton , F. J. , and Agalloco , J. P. , Eds., Validation of Pharmaceutical 
Processes: Sterile Products , Marcel Dekker , New York . 
40. Food and Drug Administration (FDA) ( 1991 ), Use of aseptic processing and terminal 
sterilization in the preparation of sterile pharmaceuticals, FR 56, 354 – 358 . 
41. PIC/S41. ( 1999 ), Decision trees for the selection of sterilisation methods 
(CPMP/QWP/054/98). 
42. PDA ( 2006 ), Technical Monograph 1, Industrial moist heat sterilization in autoclaves, 
draft 17. 
ADDITIONAL READINGS 
Akers , J. ( 2001 ), An overview of facilities for the control of microbial agents , in Block , S. S. , 
Ed., Disinfection, Sterilization and Preservation , 5th ed, Lippincott, Williams and Wilkins , 
Philadelphia , pp. 1123 – 1138 . 
Akers , J. , and Agalloco , J. ( 1997 ), Sterility and sterility assurance , J Pharm Sci Technol 51 , 
72 – 77 . 
Cole , J. C. ( 1990 ), Pharmaceutical Production Facilities — Design and Application , Ellis 
Norwood , Chicester . 
Institute of Environmental Science and Technology (IEST) ( 1995 ), Compendium of standards, 
practices, and similar documents relating to contamination control, CC009/ 
IESCC009.2, IEST, Mt. Prospect, IL. 

Ljungvist , B. , and Reinmueller , B. ( 1995 ), Ventilation and Airborne Contamination in Clean 
Rooms , Pharmacia A/B , Stockholm . 
Reinmuller , B. ( 2000 ), Microbiological risk assessment of airborne contaminants in clean 
zones, Bulletin No. 52, Royal Institute of Technology/Building Services and Engineering, 
Stockholm. 
United States Pharmacopoeia/National Formulary ( 2006 ), 29, Chapter 1116, Microbial evaluation 
of clean rooms, Rockville, Maryland, pp. 2969 – 2976 . 
ADDITIONAL READINGS 135


FACILITY 
SECTION 3


139 
3.1 
FROM PILOT PLANT TO 
MANUFACTURING: EFFECT OF 
SCALE - UP ON OPERATION OF 
JACKETED REACTORS 
B. Wayne Bequette 
Rensselaer Polytechnic Institute, Troy, New York 
Contents 
3.1.1 Motivation 
3.1.2 Background 
3.1.2.1 Pharmaceutical Process Development 
3.1.2.2 Batch Reactors 
3.1.2.3 Reaction Calorimetry 
3.1.3 Laboratory Vessels and Reaction Calorimeters 
3.1.3.1 Material and Energy Balances 
3.1.3.2 Estimating Fluid Properties and Heat Transfer Coeffi cients from Calorimeter 
Data 
3.1.3.3 Estimating Heat Flows 
3.1.3.4 Relating Heat Flows and Conversion 
3.1.3.5 Semibatch Reactions 
3.1.3.6 Rapid Scale - Up Relationships 
3.1.3.7 Strategy under a Cooling System Failure 
3.1.4 Heat Transfer in Process Vessels 
3.1.4.1 Heat Transfer Relationships 
3.1.4.2 Effect of Reactor Type, Jacket Heat Transfer Fluid, and Reactor Fluid 
Viscosity 
3.1.4.3 Pilot - and Production - Scale Experiments 
3.1.5 Dynamic Simulation Studies 
3.1.6 Summary 
References 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

140 EFFECT OF SCALE-UP ON OPERATION OF JACKETED REACTORS 
3.1.1 MOTIVATION 
There are many phases of process development between the discovery of an active 
pharmaceutical ingredient and the design, construction, and operation of a manufacturing 
process to produce a drug. A sequence of reactions and separations that 
is successful at the bench scale may lead to a process that is unsafe, is diffi cult to 
operate, or produces unsatisfactory product at the manufacturing scale. A manufacturing 
process typically has a large sequence of steps, involving several different unit 
operations (heat exchangers, reactors, separators, etc.), and a complete review of the 
design and scale - up of these unit operations would constitute a chemical engineering 
curriculum; thus, the focus of this chapter is the scale - up of jacketed batch 
chemical reactors from the laboratory to the pilot plant and manufacturing. These 
reaction vessels often serve many functions, including mixing, heating, cooling, 
distillation, and crystallization. 
Temperature control for laboratory reactors is typically easy because of high heat 
transfer area – reactor volume ratios, which do not require large driving forces (temperature 
differences) for heat transfer from the reactor to the jacket. Pilot - and 
full - scale reactors, however, often have a limited heat transfer capability. A process 
development engineer will usually have a choice of reactors when moving from the 
laboratory to the pilot plant. Kinetic and heat of reaction parameters obtained from 
the laboratory reactor, in conjunction with information on the heat transfer characteristics 
of each pilot plant vessel, can be used to select the proper pilot plant 
reactor. 
Similarly, when moving from the pilot plant to manufacturing, a process engineer 
will either choose an existing vessel or specify the design criteria for a new reactor. 
A necessary condition for operation with a specifi ed reactor temperature profi le is 
that the required jacket temperature is feasible. We have therefore chosen to focus 
on heat transfer – related issues in scale - up. Clearly there are other scale - up issues, 
such as mixing sensitive reactions. See Paul [1] for several examples of mixing scale - 
up in the pharmaceutical industry. 
In this chapter we discuss important issues as we move from laboratory to pilot 
plant and manufacturing. A review of batch process operation and pharmaceutical 
research is covered in Section 3.1.2 , followed by laboratory vessels and reaction 
calorimetry in Section 3.1.3 . In Section 3.1.4 heat transfer in process vessels is presented, 
including the effect of reactor type and heat transfer fl uid on the vessel heat 
transfer capability. In Section 3.1.5 dynamic behavior based on simulation studies 
is discussed. 
3.1.2 BACKGROUND 
3.1.2.1 Pharmaceutical Process Development 
Anderson [2] presents a wide range of topics on pharmaceutical process development, 
including a number of different problems related to process scale - up, such as 
solvent and reagent selection, purifi cation, and limitations to various operations. He 
notes that most reactors used for scale - up operations are selected for fl exibility in 
running many different processes, especially for pilot plants and multiproduct manufacturing 
plants. 

BACKGROUND 141 
Pisano [3] discusses the management of process development projects in the 
pharmaceutical industry. Case studies are used to illustrate the effect of resource 
allocation decisions at different stages of a project. While there has been a focus on 
product development in the pharmaceutical industry, clearly process development 
plays an important role in getting a product to market and lowering the long - term 
product manufacturing costs. 
3.1.2.2 Batch Reactors 
Batch processes present challenging control problems due to the time - varying 
nature of operation. Chylla and Haase [4] present a detailed example of a batch 
reactor problem in the polymer products industry. This reactor has an overall heat 
transfer coeffi cient that decreases from batch to batch due to fouling of the heat 
transfer surface inside the reactor. Bonvin [5] discusses a number of important 
topics in batch processing, including safety, product quality, and scale - up. He notes 
that the frequent repetition of batch runs enables the results from previous runs to 
be used to optimize the operation of subsequent ones. 
LeLann et al. [6] discuss tendency modeling (using approximate stoichiometric 
and kinetic models for a reaction) and the use of model predictive control (linear 
and nonlinear) in batch reactor operation. Studies of a hybrid heating – cooling 
system on a 16 - L pilot plant are presented. 
Various aspects of the effect of process scale - up on the safety of batch reactors 
have been discussed by Gygax [7] , who presents methods to assess thermal runaway. 
Shukla and Pushpavanam [8] present parametric sensitivy and safety results for 
three exothermic systems modeled using pseudohomogenous rate expressions from 
the literature. Caygill et al. [9] identify the common factors that cause a reduction 
in performance on scale - up. They present results of a survey of pharmaceutical and 
fi ne chemicals companies indicating that problems with mixing and heat transfer 
are commonly experienced with large - scale reactors. 
3.1.2.3 Reaction Calorimetry 
The microanalytical methods of differential thermal analysis, differential scanning 
calorimetry, accelerating rate calorimetry, and thermomechanical analysis provide 
important information about chemical kinetics and thermodynamics but do not 
provide information about large - scale effects. Although a number of techniques are 
available for kinetics and heat - of - reaction analysis, a major advantage to heat fl ow 
calorimetry is that it better simulates the effects of real process conditions, such as 
degree of mixing or heat transfer coeffi cients. 
Regenass [10] reviews a number of uses for heat fl ow calorimetry, particularly 
process development. The hydrolysis of acetic anhydride and the isomerization of 
trimethyl phosphite are used to illustrate how the technique can be used for process 
development. Kaarlsen and Villadsen [11, 12] provide reviews of isothermal reaction 
calorimeters that have a sample volume of at least 0.1 L and are used to measure 
the rate of evolution of heat at a constant reaction temperature. Bourne et al. [13] 
show that the plant - scale heat transfer coeffi cient can be estimated rapidly and 
accurately from a few runs in a heat fl ow calorimeter. 
Landau et al. [14] use a heat fl ow calorimeter to investigate feasible pilot plant 
operating conditions for the production of a pharmaceutical intermediate. They 

142 EFFECT OF SCALE-UP ON OPERATION OF JACKETED REACTORS 
determine kinetic and heat fl ow parameters using the calorimeter and further estimate 
heat transfer parameters for a pilot - scale reactor. Simulation studies are used 
to fi nd the required jacket temperature for desired batch reactor temperature pro- 
fi les. Semibatch operation is shown to be safer than normal batch operation. Landau 
[15] provides a detailed review of reaction calorimetry, including mathematical 
expressions for energy balances, and a number of application examples. 
3.1.3 LABORATORY VESSELS AND REACTION CALORIMETERS 
As reviewed in Section 3.1.2.3 , reaction calorimeters can be used to better understand 
and characterize scale - related process phenomena, such as mixing and heat 
transfer. A heat fl ow calorimeter, the Mettler RC1e, is shown in Figure 1 . A 
schematic of a similar calorimeter system is shown in Figure 2 [16] . A heat fl ow 
calorimeter can be used to estimate: 
• Physical parameters (heat capacity) 
• Reaction rate constants 
• Heat transfer coeffi cients (overall, U or, or fi lm, h i ) 
3.1.3.1 Material and Energy Balances 
The overall energy balance for a process with no reaction has the form 
Energy accumulation energy in heat transfer from jacket 
energy i 
= 
+ n by calibration probe energy lost by ambient heat transfer . 
FIGURE 1 Mettler RC1e heat fl ow calorimeter system ( www.mt.com ). 

FIGURE 2 Schematic of HEL SIMULAR reaction calorimeter. From ref. 16 . 
Additional heater 
F3 F2 F1 
Stirrer 
Tamb 
Qadd 
n 
Condenser 
Tw,in 
TR pHR pR 
Tj,out 
Tj,in 
Tw,out and mw 
Inert gas 
venting 
Circulation 
thermostat, 
heater, chiller 
Liquid 
surface 
Oil jacket 
Outlet valve Scale Scale Scale 
which is shown mathematically as 
( ) ( ) ( ) mc 
dT 
dt 
UAT T q k T T p r j = . . + . . cal loss amb (1) 
where ( mc p ) r 
is the reactor thermal capacitance, T is the reactor temperature, T j is 
the jacket temperature, U is the overall heat transfer coeffi cient, A is the area for 
heat transfer, q cal is the heat fl ow from the calibration probe, and the fi nal term 
accounts for heat loss from the reactor system. The thermal capacitance is composed 
of the fl uid in the vessel as well as the inert components in contact with the fl uid, 
including the vessel wall, agitator (stirrer), and sensors (e.g., thermocouple), as 
shown in the equation 
( ) mc V c m c p r p v pv = + . (2) 
where V is the volume of liquid, . is the liquid density, c p is the liquid heat capacity, 
m v is the mass of the vessel wall and other inerts, and c pv is the average heat capacity 
of the vessel wall and inerts. The inert contributions and heat transfer to the ambient 
can be found from extensive calibration studies. For small - scale reactors, such as 
reaction calorimeters, the thermal mass of the inerts can be signifi cant. The thermal 
capacitance ratio, sometimes called the Lewis number, is given as 
. 
. . 
= =+ 
( ) mc 
V c 
m c 
V c 
p r 
p 
v pv
p 
1 (3) 
which can be on the order of 1.5 – 2 for a small - scale reactors and adiabatic calorimeters 
but is often 1.05 – 1.10 for small pilot plant reactors and less than 1.02 for 
manufacturing - scale reactors. 
LABORATORY VESSELS AND REACTION CALORIMETERS 143

144 EFFECT OF SCALE-UP ON OPERATION OF JACKETED REACTORS 
3.1.3.2 Estimating Fluid Properties and Heat Transfer Coeffi cients from 
Calorimeter Data 
In a heat fl ow calorimeter, a feedback controller is used to maintain a constant 
desired reactor temperature by adjusting the jacket temperature. From (1), with a 
constant calibration probe heat fl ow, at steady state ( dT / dt = 0), the overall heat 
transfer coeffi cient can be found from 
UA 
q k T T 
T Tj 
= 
. . 
. 
cal loss amb ( ) 
(4) 
Also, the fl uid heat capacity can be found by ramping up the reactor temperature 
and using 
( ) 
( ) ( ) 
mc 
UAT T q k T T 
dT dt p r 
j =
. . + . . cal loss amb 
/ 
(5) 
and solving for c p from (2), assuming that the reactor inert component contributions 
are known from previous studies. An example calibration study is shown in Figure 
3 , where a constant heat fl ow is applied from 35 to 42 min, enabling the heat transfer 
coeffi cient to be estimated from the temperature difference using Equation (4) . 
Then, the heat capacity is estimated from the temperature ramp applied between 5 
and 20 min. It should be noted that the heat transfer coeffi cient and heat capacity 
of the fl uid may vary with concentration and temperature. Typically, calibration 
experiments are performed before and after the reaction; then the heat transfer 
coeffi cient and heat capacity are assumed to vary linearly with conversion or batch 
time. For polymerization reactions in particular, the viscosity can increase tremendously 
with conversion, causing a substantial decrease in the heat transfer coeffi - 
FIGURE 3 Example reaction calorimetry study without reaction. The overall heat transfer 
coeffi cient area can be found during the steady - state temperature difference and known calibration 
probe heat fl ow, between 35 and 42 min. The heat capacity can then be found from 
the temperature ramp between 5 and 20 min. 
RC1 Calibration profiles 
Determine UA 
Determine cp 
Time, min Temperature, °C 
28 
26 
24 
22 
20 
20 30 40 50 60 
18
0 10 
reactor 
jacket

cient. Reaction experiments can be run at several temperatures to fi nd the functional 
relationship with temperature. 
Since the heat transfer area as a function of liquid volume is known, the overall 
heat transfer coeffi cient U can be calculated from (4). The overall heat transfer 
coeffi cient is calculated as 
1 1 
U h 
x
k i 
g
g 
= + (6) 
where the jacket side resistance is negligible. The glass vessel heat transfer resistance 
( x g / k g , thickness/thermal conductivity) can be used to fi nd the reactor fl uid heat 
transfer coeffi cient ( h i ). 
3.1.3.3 Estimating Heat Flows 
The reaction heat fl ow can be found by rearranging (1), with the calibration heat 
probe replaced by the reaction heat fl ow, to fi nd 
q mc 
dT 
dt 
UA T T k T T r p r j = + . + . ( ) ( ) ( ) loss amb (7) 
The total heat released during the reaction can be found by integrating (7), 
Q qdt r 
tf 
tot = .0 (8) 
or, represented as a scaled (per - unit mass) total heat release, 
Q 
Q
V 
Q
m tot 
tot tot = = 
. 
(9) 
The molar heat of reaction can be found from 
.H 
Q 
n rxn 
tot 
rxn 
= 
. 
(10) 
where n rxn is the molar amount reacted. 
As a “ fi rst - pass ” calculation, if it is assumed that the dominant heat transfer 
resistance is on the reactor side, then the overall heat transfer coeffi cient ( U ) from 
(4) can be used for scale - up. 
3.1.3.4 Relating Heat Flows and Conversion 
The reaction heat fl ows are directly related to the conversion of reactants [14] . 
Consider a fi rst - order reaction of a limiting reactant, with the rate expression 
dC 
dt 
kC = . (11) 
LABORATORY VESSELS AND REACTION CALORIMETERS 145

146 EFFECT OF SCALE-UP ON OPERATION OF JACKETED REACTORS 
where C is the molar concentration of the reactant. The heat fl ow is 
q 
dC 
dt 
H V kC H V r =( ) = . . . rxn rxn (12) 
with an initial heat fl ow of 
q kC H V r0 0 = . . rxn (13) 
dividing (12) by (13), we fi nd the relationship between concentration and heat 
fl ow: 
C 
C 
q 
q 
r 
r 0 0 
= (14) 
For an isothermal reaction, the solution to (11) is 
C 
C 
e kt 
0 
= . (15) 
so, the heat fl ow for an isothermal reaction is 
q 
q 
e r 
r 
kt 
0 
= . (16) 
Thus, the reaction rate constant k can be estimated from the reaction heat fl ow 
without making any concentration measurements. Assuming an Arrhenius rate 
expression 
k Ae E RT = . 
0 
/ (17) 
the rate constant at several temperatures can be used to estimate the frequency 
factor ( A 0 ) and activation energy ( E ). (See ref. 14 for an example application.) 
3.1.3.5 Semibatch Reactions 
For extremely exothermic reactions it is necessary to slowly add the feed over time, 
that is, operate in a semibatch fashion. The heat fl ow for a semibatch reaction can 
be found from 
q UAT T mc 
dT 
dt 
m c T T r j p r f pf f = . + + . ( ) ( ) ( )  (18) 
where  mf is the mass fl ow rate of the feed stream. If the reactor temperature is 
maintained constant, this reduces to 
q UAT T mc T T r j f pf f = . + . ( ) ( )  (19) 

For reactions with essentially instantaneous kinetics, the reaction rate is limited by 
the feed addition rate. For other reactions, particularly if the reactor is operated at 
too low of a temperature, a reactant concentration can “ build up, ” eventually reaching 
an unsafe level that could lead to a rapid temperature rise and explosion. It is 
important for these reactions to monitor the heat fl ow to confi rm that the reactant 
concentration is not increasing to unacceptable levels. 
3.1.3.6 Rapid Scale - Up Relationships 
Lacking knowledge of the larger scale reactor, it is tempting to simply assume that 
only the area for heat transfer varies upon scale - up. A natural parameter is the 
cooling time , 1 defi ned as 
. 
. . 
co= = 
( ) mc 
UA 
V c 
UA 
p r p (20) 
The heat transfer area varies with the square of the vessel diameter, and the volume 
varies with the cube of the vessel diameter. Thus the area – volume ratio ( A / V ) varies 
with volume as 
A
V V 
~ 
1
1/3 
(21) 
The inverse cooling time relationship for scale - up from volume V 1 to V 2 is 
UA 
V c 
UA 
V c 
V
V p p . . . . 
.
. . 
.
. . 
= .
. . 
.
. . 
( ) 2 1 
1
2 
1/3 
(22) 
The required reactor - jacket temperature difference on scale - up, with a constant 
Lewis number, is 
[ ] [ ] T T T T 
V
V j j . = . ( ) 2 1 
2
1 
1/3 
(23) 
so the temperature difference can increase dramatically when a process is scaled up 
several orders of magnitude. Reactor - jacket temperature difference constraints can 
be particularly important for glass - lined vessels, where the limit is often 75 ° C. 
3.1.3.7 Safety under a Cooling System Failure 
In the event of a cooling system failure it can be assumed that the reactor operates 
adiabatically. The adiabatic temperature rise can be found from 
1 The notion of cooling time can be understood by writing (1) and assuming no calibration energy or heat 
loss. Then (1) becomes . co ( dT / dt ) = . ( T . T j ). If a constant temperature difference T . T j is applied, it 
will take . co time units for the reactor temperature to change by the temperature difference. 
LABORATORY VESSELS AND REACTION CALORIMETERS 147

148 EFFECT OF SCALE-UP ON OPERATION OF JACKETED REACTORS 
.T 
Q
mcp r 
ad 
tot =
( ) 
(24) 
and the fi nal temperature is 
T T T final initial ad = +. (25) 
As long as the fi nal temperature is less than some critical “ onset ” temperature where 
a secondary decomposition reaction occurs, then the process can safely handle a 
cooling system failure. If a batch reactor temperature cannot be assured to remain 
less than the onset temperature after a cooling system failure, then a semibatch 
operation should be used. As noted in Section 3.1.3.5 , it is necessary to assure that 
reactant concentration is not increasing above an onset concentration where a 
similar decomposition could occur with a cooling system failure. 
3.1.4 HEAT TRANSFER IN PROCESS VESSELS 
Based on initial heat fl ow calorimetry studies, a process development engineer must 
choose the appropriate reactor vessels for pilot plant studies. A pilot plant typically 
has vessels that range from 80 to 5000 L, some constructed of alloy and others that 
are glass lined. In addition some vessels may have half - pipe coils for heat transfer, 
while others have jackets with agitation nozzles. A process drawing for a typical 
glass - lined vessel is shown in Figure 4 . In Sections 3.1.4.1 and 3.1.4.2 we review 
fundamental heat transfer relationships in order to predict overall heat transfer 
coeffi cients. In Section 3.1.4.3 we review experimental techniques to estimate heat 
transfer coeffi cients in process vessels. 
3.1.4.1 Heat Transfer Relationships 
Reactor - Side Coeffi cient The reactor - side heat transfer coeffi cient is calculated 
as 
h a
k
D i 
i
i 
i i = Re Pr . . 0 67 0 33 (26) 
where a is the agitation constant (0.33), k i is the fl uid thermal conductivity, Re i is 
the Reynolds number, and Pr i is the Prandtl number, 
Rei 
ag i 
i 
D N 
= 
2 . 
. 
(27) 
Pri 
i pi 
i 
c 
k 
= 
. 
(28) 

FIGURE 4 Typical 300 - or 500 - gal jacketed vessel ( www.pfaudler.com ). 
SRW 3525 drive 
Lubricated dry 
mechanical seal 
Drive nozzle 
face 
E
10. 
5. 
6. 
3. 18. 
12. 
3. Legs (four) 
45. Leg circle 
54. O.D. 
48. I.D. 
13. 
(3. Nozs.) (4. Nozs.) 
A 
B 
C 
D 
F 
Optional side 
supports 
Fin Battle w/RTD 
Temperature Sansor 
23. Span 
Cryo-Lock CBT 
2. Cplgs. (Two) 
1/2. Cplg. 
1/2. Cplg. 
(4) 3/4. dia. holes 
equally spaced on 
a 10. BC 
14 1/4. (6. Noz.) 
4 1/4. 
3. Noz. 
1 1/2. Cplgs. 
13 1/4. 
1 1/2. agit. 
nozs. 
(Offset) 
and N is the agitator rotation rate. It should be noted that the fi lm heat transfer 
coeffi cient varies inversely with the viscosity, that is, 
hi 
i 
~ . 
1
0 33 . 
(29) 
Reactions where the viscosity increases substantially with conversion, such as some 
polymerization reactions, can be particularly diffi cult to control upon scale - up. 
Jacket - Side Coeffi cient Here the calculations are shown for a jacket equipped 
with agitation nozzles that greatly increase the jacket fl uid velocity. The jacket “ swirl 
velocity ” v j is calculated (iteratively) from the nonlinear algebraic relationship 
[17] 
m v v 
fL 
D 
v 
A n n j 
e 
j 
f ( ) . =( ).. . 
.. . 
4 
2
2 
. (30) 
HEAT TRANSFER IN PROCESS VESSELS 149

150 EFFECT OF SCALE-UP ON OPERATION OF JACKETED REACTORS 
where  mn the is the nozzle mass fl ow rate, v n is the nozzle velocity, the friction factor 
is 
f = 
. 2 0 023 
0 2 
. 
Re . 
(31) 
the jacket - side fi lm coeffi cient is 
h 
k
D j 
j
e 
j j = 0 027 0 8 0 33 . Re Pr . . (32) 
and the Reynolds and Prandtl numbers are 
Rej 
e j j 
j 
D v 
= 
. 
. 
(33) 
Prj 
j pj 
j 
c 
k 
= 
. 
(34) 
Overall Coeffi cient The overall heat transfer coeffi cient is found from the sum of 
the resistances, 
1 1 1 
U h h 
x
k 
x
k 
ff ff 
i j 
m
m 
g
g 
i j = + + + + + (35) 
which includes reactor fi lm, jacket fi lm, vessel metal, vessel glass, and fouling factors 
for both the reactor and jacket sides. 
3.1.4.2 Effect of Reactor Type, Jacket Heat Transfer Fluid, and Reactor 
Fluid Viscosity 
Here we present examples of how the reactor type and heat transfer fl uid affect the 
heat transfer coeffi cient. When the reactor fl uid has a low viscosity, the dominant 
heat transfer resistance tends to be on the jacket side. When the reactor fl uid has a 
high viscosity, however, the dominant resistance is typically on the reactor side. 
Parameter values for the studies are presented in Figures 5 – 7 and are given in the 
literature [18] . 
The overall heat transfer coeffi cient is much higher for an alloy reactor/half - pipe 
jacket than for a glass - lined carbon steel reactor/agitation nozzle jacket, as shown 
in Figure 5 , where Syltherm is the heat transfer fl uid. Syltherm has a signifi cantly 
lower heat transfer coeffi cient than an ethylene glycol mixture, as shown in Figure 
6 , but is capable of operating over a wider range of temperatures. The reactor fl uid 
viscosity has a tremendous effect on the overall heat transfer coeffi cient, as shown 
in Figure 7 . This can be particularly important in polymerization reactions where 
viscosity increases with conversion. 

FIGURE 5 Overall heat transfer coeffi cient for 500 - gal reactors. Comparison of alloy half 
pipe with glass - lined carbon steel (GLCS). Syltherm is the heat transfer fl uid. ( From ref. 18 , 
with permission .) 
–50 0 50 100 150 200 250 
20 
30 
40 
50 
60 
70 
80 
Jacket temperature °C 
Overall U, English units 
Half pipe 
Jacket w/nozzles 
FIGURE 6 Overall heat transfer coeffi cient for 500 - gal GLCS reactor. Comparison of Syltherm 
with Glycol. ( From ref. 18 , with permission .) 
–50 0 50 100 150 200 250 
15 
20 
25 
30 
35 
40 
45 
50 
55 
60 
Jacket temperature °C 
Overall U, English units 
Syltherm 
Glycol 
3.1.4.3 Pilot - and Production - Scale Experiments 
The relationships shown in Section 3.1.3 are also pertinent to large - scale reactors. 
By using different solvents and volumes of solvent, pilot and production reactor 
heat transfer characteristics can be determined from a series of experiments. A 
primary limitation, compared to reaction calorimeter characterization, is that a calibration 
probe is rarely available. Thus, heat - up and cool - down studies, performed 
HEAT TRANSFER IN PROCESS VESSELS 151

152 EFFECT OF SCALE-UP ON OPERATION OF JACKETED REACTORS 
FIGURE 7 Overall heat transfer coeffi cient for 500 - gal GLCS reactor with glycol heat 
transfer fl uid. Comparison of effect of reactor - side viscosity. 
–20 0 20 40 60 80 100 120 
10 
15 
20 
25 
30 
35 
40 
45 
50 
55 
60 
Jacket temperature °C 
Overall U, English units 
500 gal. GLCS. 1 cp vs. 3000 cp 
1 cP 
3000 cP 
by varying the jacket temperature and observing the changes in the reactor temperature 
(for solvents with known heat capacity), are used to characterize the 
reactor. The inverse cooling time, 
UA 
mc 
dT dt 
T T p r j ( ) 
= 
.
/ 
(36) 
can be estimated from the temperature data collected from a heat - up/cool - down 
study. A characteristic example for a pilot - scale reactor is shown in Figure 8 . The 
FIGURE 8 Temperature profi les (jacket inlet, jacket outlet, and reactor) for a pilot plant 
reactor. ( From ref. 19 .) 
Time, min 
0 20 40 60 80 100 120 140 160 180 
100 
80 
60 
40 
20
0 
temperature, °C

FIGURE 9 Cooling time estimates based on data presented in Figure 8 . ( From ref. 19 .) 
0 10 
28 
27 
26 
25 
24 
23 
22 
21 
20 
19 
20 30 
Jacket temperature, °C 
mCp/UA, min 
40 50 60 70 80 90 
cool-down 
heat-up 
resulting cooling time estimates are shown in Figure 9 . Notice that the overall heat 
transfer coeffi cient is clearly a function of the jacket temperature. The reduced heat 
transfer at the lower jacket temperatures is due to the strong relationship between 
viscosity and temperature for the 40% glycol solution used in the jacket. The discontinuity 
in the cooling time estimate at around 45 ° C may be due to two factors. 
One factor is the assumption of no heat loss from the vessel, which would tend to 
lower the UA estimates during the heat - up phase. Another factor is the assumption 
that the metal and glass inerts in the reactor are at the temperature of the reactor; 
in practice it might be a better assumption that the reactor wall in particular is at a 
temperature that is intermediate between the jacket and reactor temperatures. 
The fl uid and inert thermal masses can be independently estimated by conducting 
experiments with a number of different solvent amounts. From the cooling time 
expression 
( ) mc 
UA 
m c 
UA 
V c 
UA 
p r v pv p = + 
. 
(37) 
writing this as a function of the reactor fl uid volume, 
( ) mc 
UA 
m c 
UA 
c 
UA 
V p r v pv p = + . 
. 
(38) 
and conducting experiments at a number of different fl uid volumes or, equivalently, 
masses ( V . ), 
( ) mc 
UA 
m c 
UA 
c 
UA 
V p r v pv p = + .. (39) 
the linear regression can be used to fi nd the slope and intercept and thus estimate 
the UA and m v c pv terms [19] . This approach is shown in Figure 10 for a jacket tem- 
HEAT TRANSFER IN PROCESS VESSELS 153

154 EFFECT OF SCALE-UP ON OPERATION OF JACKETED REACTORS 
FIGURE 10 Linear regression to estimate thermal mass and UA . ( From ref. 19 .) 
200 250 300 350 
Mass of water, kg 
30 
28 
26 
24 
22 
20 
18 
mCp/UA, min 
perature of 60 ° C (based on a total of eight experiments at fi ve different reactor 
fl uid volumes). 
3.1.5 DYNAMIC SIMULATION STUDIES 
Older pilot plant and manufacturing processes often used steam for heating and 
water for cooling, with a switch - and - purge strategy between the two modes. Recent 
process designs have two heat transfer fl uid systems (hot and cold heat transfer 
fl uids) that are used for most of the heating and cooling needs. In addition, some 
vessels may have nitrogen coolers for cryogenic operation. 
A simplifi ed schematic for a jacket heat transfer service is shown in Figure 11 
[18] . Here, two separate heat transfer fl uid headers are used, and the control valve 
is on the outlet stream to reduce the temperature shocks that might occur if a single 
FIGURE 11 Characteristic pilot plant vessel control strategy. Slave (secondary) controller 
based on jacket outlet temperature is shown. The control valve is on the outlet stream to 
minimize temperature gradients (when switching from hot to cold fl uids) that would be 
imposed if the valve was on the inlet. ( From ref. 18 , with permission .) 
TC1 
TC2 
From hot HT 
fluid system 
From cold HT 
fluid system 
To cold HT 
fluid system 
To hot HT 
fluid system

control valve was on the inlet stream. Depending on the range of temperatures, 
either ethylene glycol or a proprietary fl uid such as Syltherm is used. Depending 
on whether heating or cooling is needed, either the hot or cold process control 
valve is open. Similarly, on – off valves return fl uid to the appropriate distribution 
system. 
Although the heat transfer fl uid can be used over a wide range of temperatures, 
the fi lm heat transfer coeffi cient is a strong function of temperature due to viscosity 
effects. The “ cooling time ” of a large reactor operating at a low temperature can be 
substantially longer than that of a small reactor operating at a high temperature 
due to this strong temperature effect. Simulation studies can be used to: 
• Understand the effect of heat transfer fl uid 
• Understand possible performance limitations due to scale and operating 
conditions 
• Test the effect of specifi ed temperature gradient constraints 
• Assist with controller design and selection of tuning parameters for system 
start - up 
Various levels of models can be used to describe the behavior of pilot - scale jacketed 
batch reactors. For online reaction calorimetry and for rapid scale - up, a simple 
model characterizing the heat transfer from the reactor to the jacket can be used. 
Another level of modeling detail includes both the jacket and reactor dynamics. 
Finally, the complete set of equations simultaneously describing the integrated 
reactor/jacket and recirculating system dynamics can be used for feedback control 
system design and simulation. The complete model can more accurately assess the 
operability and safety of the pilot - scale system and can be used for more accurate 
process scale - up. 
In the simulation studies that follow, it is assumed that the reactor and jacket are 
well mixed, resulting in differential equations for the material and energy balances 
[18] . The reactor shell (including a glass lining, if used) and reactor internals (agitator 
and baffl es) are at the same temperature as the reactor, so their “ thermal mass ” 
is included in the reactor energy balance. Similarly, the jacket shell is at the jacket 
temperature, with an associated thermal mass. The heat transfer area A is proportional 
to the reactor liquid level (between volumes associated with the minimum 
and maximum heat transfer area); also, the reactor shell thermal mass varies linearly 
with the liquid level. Heat transfer coeffi cients are calculated using the relationships 
presented in Section 3.1.4 ; see Garvin [20] or Dream [21] for detailed examples. 
Parameters, viscosity in particular, are a function of temperature. 
We focus on the effect of reactor size and material of construction on the expected 
dynamic behavior of the reactors. Details on the model development and simulation 
environment are presented elsewhere [18] . Figure 12 illustrates that a vessel can 
have signifi cantly different dynamic behavior depending on whether it is being 
heated or cooled (for illustrative purposes, the freezing point of water is neglected 
in this simulation). The increase in reactor temperature results in a much faster 
response than a decrease for two reasons: (i) the jacket heat transfer fl uid has a 
much higher viscosity (resulting in a lower overall heat transfer coeffi cient) at low 
temperatures and (ii) the fl uid fl ow rate/jacket temperature gain is proportional to 
DYNAMIC SIMULATION STUDIES 155

156 EFFECT OF SCALE-UP ON OPERATION OF JACKETED REACTORS 
FIGURE 12 Comparison of responses for ± 30 ° C reactor temperature setpoint changes at 
t = 10 min; 500 - gal GLCS fi lled with water (1925 kg). 
0 20 40 60 80 100 120 140 160 180 200 
–40 
–20
0 
20 
40 
60 
80 
100 
Time, min 
Temperature, °C 
FIGURE 13 Comparison of temperature responses for 30 ° C batch setpoint change; 500 - gal 
GLCS, water (1925 L) vs. organic (1700 L). 
0 20 40 60 80 100 120 140 160 180 200 
20 
30 
40 
50 
60 
70 
80 
90 
100 
Time, min 
Temperature, °C 
Water 
Organic 
the difference between the jacket temperature and make - up fl uid temperature, 
which becomes small at low jacket temperatures. Notice that the initial response for 
the temperature increase is constrained by the ramp limit of 5 ° C/min on the jacket 
temperature. The temperature response of an organic solvent is much faster than 
water because of the heat capacity difference, as shown in Figure 13 . The previous 
plots were for simple heating/cooling applications (ref. 18 presents further studies 
for cryogenic and semibatch systems). 

3.1.6 SUMMARY 
In this chapter we have presented an overview of scale - up considerations involved 
as one moves from bench - scale reaction calorimetry to larger scale pilot plant and 
production reactors. Our focus has been on heat transfer and single - phase processes, 
addressing primarily the problem that the heat transfer area per unit reactor volume 
decreases with scale. Clearly, there are many challenging problems associated with 
multiphase vessels, with evaporation/distillation and crystallization as obvious 
examples, but these topics are beyond the scope of this chapter. 
REFERENCES 
1. Paul , E. L. ( 1988 ), Design of reaction systems for specialty organic chemicals , Chem. Eng. 
Sci. , 43 ( 8 ), 1773 – 1782 . 
2. Anderson , N. G. ( 2000 ), Practical Process Research and Development , Academic , New 
York . 
3. Pisano , G. P. ( 1997 ), The Development Factory , Harvard Business School , Boston . 
4. Chylla , R. W. , and Hasse , D. R. ( 1993 ), Temperature control of semi - batch polymerization 
reactors , Comp. Chem. Eng. , 17 ( 3 ), 257 – 264 . 
5. Bonvin , D. ( 1998 ), Optimal operation of batch reactors — A personal view , J. Proc. Cont. , 
8 ( 5 – 6 ), 355 – 368 . 
6. LeLann , M. V. , Cabassud , M. , and Casamatta , G. ( 1999 ), Modeling, optimization and 
control of batch chemical reactors in fi ne chemical production , Annu. Rev. Control , 23 , 
25 – 34 . 
7. Gygax , R. W. ( 1990 , Feb.), Scale - up principles for assessing thermal runaway risks , Chem. 
Eng. Prog. , 86 ( 2 ), 53 – 60 . 
8. Shukla , P. K. , and Pushpavanam , S. ( 1994 ), Parametric sensitivity, runaway, and safety in 
batch reactors: Experiments and models , Ind. Eng. Chem. Res. , 33 ( 12 ), 3202 – 3208 . 
9. Caygill , G. , Zanfi r , M. , and Gavrildis , A. ( 2006 ), Scalable reactor design for pharmaceuticals 
and fi ne chemicals production. 1: Potential scale - up obstacles , Org. Proc. Res. Dev. , 
10 ( 3 ), 539 – 552 . 
10. Regenass , W. ( 1985 ), Calorimetric monitoring of industrial chemical processes , Thermochim. 
Acta , 95 , 351 – 369 . 
11. Kaarlsen, L. G. , and Villadsen, J. (1987), Isothermal reaction calorimeters—I. A literature 
review , Chem. Eng. Sci. , 42 ( 5 ), 1153 – 1164 . 
12. Kaarlsen , L. G. , and Villadsen , J. ( 1987 ), Isothermal reaction calorimeters — II. Data treatment 
, Chem. Eng. Sci. , 42 ( 5 ), 1165 – 1173 . 
13. Bourne , J. R. , Buerli , M. , and Regenass , W. ( 1981 ), Heat transfer and power measurements 
in stirred tanks using heat fl ow calorimetry , Chem. Eng. Sci. , 36 , 347 – 354 . 
14. Landau , R. N. , Blackmond , D. G. , and Tung , H. - H. ( 1994 ), Calorimetric investigation of an 
exothermic reaction: Kinetic and heat fl ow modeling , Ind. Eng. Chem. Res. , 33 , 814 – 820 . 
15. Landau, R. N. (1996), Expanding the role of reaction calorimetry , Thermochim. Acta , 289 , 
101 – 126 . 
16. Obenndip , D. A. , and Sharratt , P. N. ( 2006 ), Towards an information - rich process development. 
Part I: Interfacing experimentation with qualitatitive/semiquantitative modeling , 
Org. Proc. Res. Dev. , 10 ( 3 ), 430 – 440 . 
REFERENCES 157

158 EFFECT OF SCALE-UP ON OPERATION OF JACKETED REACTORS 
17. Bolliger , D. H. ( 1982 ), Assessing heat transfer in process - vessel jackets , Chem. Eng. , Sept. 
20 , 95 – 100 . 
18. Bequette , B. W. , Holihan , S. , and Bacher , S. ( 2004 ), Automation and control issues in the 
design of a pharmaceutical pilot plant , Control Eng. Practice , 12 , 901 – 908 . 
19. Zima , A. , Spencer , G. , and Bequette , B. W. ( 1996 ), Model development for batch reactor 
calorimetry and control, Preprint, presented at the AIChE Annual Meeting, Chicago, IL, 
Nov. 1996. 
20. Garvin , J. ( 1999 ), Understand the thermal design of jacketed vessels , Chem. Eng. Prog. , 
95 ( 6 ), 61 – 68 . 
21. Dream , R. F. ( 1999 ), Heat transfer in agitated jacketed vessels , Chem. Eng. , Jan., 90 – 96 . 

159 
3.2 
PACKAGING AND LABELING 
Maria In e s Rocha Miritello Santoro and Anil Kumar Singh 
University of S a o Paulo, S a o Paulo, Brazil 
Contents 
3.2.1 Introduction 
3.2.2 Packaging Materials 
3.2.2.1 General Considerations 
3.2.2.2 Glass as packaging material 
3.2.2.3 Plastic as Packaging Material 
3.2.2.4 Metal as Packaging Material 
3.2.2.5 Applications: Some Examples 
3.2.3 Quality Control of Packaging Material 
3.2.3.1 General Considerations 
3.2.3.2 Packaging Components 
3.2.3.3 Inhalation Drug Products 
3.2.3.4 Drug Products for Injection and Ophthalmic Drug Products 
3.2.3.5 Liquid - Based Oral Products, Topical Drug Products, and Topical Delivery 
Systems 
3.2.3.6 Solid Oral Dosage Forms and Powders for Reconstitution 
3.2.4 Importance of Proper Packaging and Labeling 
3.2.5 Regulatory Aspects 
3.2.5.1 General Considerations 
3.2.5.2 Food, Drug and Cosmetic Act 
3.2.5.3 New Drugs 
3.2.5.4 Labeling Requisites 
3.2.5.5 Prescription Drugs 
3.2.5.6 Drug Information Leafl et 
3.2.5.7 Other Regulatory Federal Laws 
3.2.5.8 Fair Packaging and Labeling Act 
3.2.5.9 United States Pharmacopeia Center for the Advancement of Patient 
Safety 
3.2.5.10 National Agency of Sanitary Vigilance (ANVISA, Brazil) 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

160 PACKAGING AND LABELING 
3.2.5.11 International Committee on Harmonization (ICH) 
3.2.5.12 European Union Regulatory Bodies 
References 
3.2.1 INTRODUCTION 
The packaging of a pharmaceutical product fulfi ls a variety of roles, such as product 
presentation, identifi cation, convenience, and protection until administration or use. 
Selection of packaging requires a basic knowledge of packaging materials, the environmental 
conditions to which the product will be exposed, and the characteristics 
of the formulation. Several types of packaging are used to contain and protect the 
pharmaceutical preparations, such as the primary packaging around the product and 
secondary packaging such as a carton and subsequent transit cases [1] . 
The principal objective of the modern pharmaceutical industry is to manufacture 
pharmaceutical preparations presenting high quality, identity, purity, effectiveness, 
and innocuity in order to guarantee the satisfaction and safety of patients. The 
development of a new drug must involve the synthesis of a molecule, determination 
of its pharmacological activity, industrial - scale production, and its commercialization 
to guarantee quality of the fi nal product. 
Packaging system development, including primary and secondary packaging 
components, is of critical importance. The material should be selected based on the 
characteristics of pharmaceutical product and dosage form. After the production 
phase, packaging must be planned according to regulatory requirements and its 
quality should be controlled according to the specifi cations. 
Commercially, the packaging material is used as a barrier to protect the pharmaceutical 
preparations against external factors that can degrade them and consequently 
decrease their effectiveness and increase toxic effects. 
Once the type of packaging material is decided based on such factors as size, 
shape, capacity, and physicochemical properties, all these data, including quality 
control tests, should be included in the specifi cation of the products in order to 
assure the therapeutic effectiveness during its shelf life. 
Several types of materials are in use in the preparation of containers and closure 
systems: glass, plastics, metals, and combinations of these materials. However, care 
should be taken in the selection of appropriate material. These materials should not 
present any physical or chemical reactivity that could modify drug activity, quality, 
purity, or physical characteristics of the drug and pharmaceutical preparations. Any 
minor modifi cation in the pharmacopeial specifi cation is acceptable if it does not 
present a threat to patient ’ s health. 
The aim of this chapter is to discuss the importance of the packaging and labeling 
of pharmaceutical preparations. The role of packaging and labeling in the pharmaceutical 
industry has grown substantially over the past decade. The total packaging 
operation is part of any drug development program. Pharmaceutical products generally 
require a standard of packaging which is superior to that of most other products 
in order to support and comply with their main requirements, such as effi cacy, integrity, 
purity, safety, and stability. 

PACKAGING MATERIALS 161 
For these reasons packaging technology should be based on the understanding 
of pharmaceutical products, characteristics of formulations, and dosage forms, 
including the physical and chemical properties of the drug substance. 
In the past, packaging concerns often arose only during the later steps of product 
development. Today, packaging is integrated with the development step and is 
among the earliest considerations of new pharmaceutical preparations being studied. 
Labels of products can vary from the simple to the extremely complex. But, even 
at the most basic level, product identifi cation should meet regulatory requirements. 
More complex are the labels that make use of bar code technologies. New components 
such as microchips, biosensors, and deoxyribonucleic acid (DNA) arrays are 
making possible the development of new technologies leading to fi nished products 
individually packed that require specialized packaging materials and design expertise. 
The challenge now is to maintain low packaging cost, that is, always integrated 
into the cost of the product itself. 
Packaging in the post – World War II period benefi ted immensely from the commercialization 
of plastics, which were little known or used in prior years. Since then, 
the packaging industry has openly adopted plastics as a powerful new tool in the 
development of new packaging forms and functions. 
Quality control of a packaging component starts at the design stage. All aspects 
of package development that may give rise to quality problems must be identifi ed 
and minimized by good design. Identifying and correcting mistakes in packaging 
will avoid product recall and rejection of pharmaceutical preparations [2, 3] . 
3.2.2 PACKAGING MATERIALS 
3.2.2.1 General Considerations 
Packaging refers to all the operations, including fi lling and labeling, through which 
a bulk product should pass to become a fi nished product. Usually, sterile fi lling is 
not considered part of the packing process, although the bulk product is contained 
in a primary container. 
A packaging component means any single part of a container closure system. 
Typical components are containers (e.g., ampules, vials, bottles), container liners 
(e.g., tube liners), closures (e.g., screw caps, stoppers), closure liners, stopper overseals, 
container inner seals, administration ports [e.g., on large - volume parenterals 
(LVPs)], overwraps, administration accessories, and container labels [4] . 
A primary packaging component is one that is or may be in direct contact with 
the dosage form. A secondary packaging component is one that is not and will not 
be in direct contact with the dosage form [4] . 
A container closure system refers to the sum of packaging components that 
together contain and protect the dosage form. This includes primary packaging 
components and secondary packaging components, if the latter are intended to 
provide additional protection to the drug product. A packaging system is equivalent 
to a container closure system [4] . 
The role of packaging material on the overall perceived and actual stability of 
the dosage form is well established. Packaging plays an important role in quality 
maintenance, and the resistance of packaging materials to moisture and light can 

162 PACKAGING AND LABELING 
signifi cantly affect the stability of drugs and their dosage forms. It is crucial that 
stability testing of dosage forms in their fi nal packaging be performed. The primary 
role of packaging, other than its esthetic one, is to protect the dosage forms from 
moisture and oxygen present in the atmosphere, light, and other types of exposure, 
especially if these factors affect the overall quality of the product on long - term 
storage [5] . 
The compliance packaging such as for fi xed - dose combination pills and unit 
dosage form packaging is a therapy - related intervention and is designed to facilitate 
medication regimens and so potentially improve adherence. Compliance packaging 
can be defi ned as a prepackaged unit that provides one treatment cycle of the 
medication, to both the pharmacist and the patient, in a ready - to - use package. This 
innovation type of packaging is usually based on blister packaging that contain unit 
therapeutic dose for one time use. The separate dosage units and separate days are 
usually indicated on the dosage cards to help remind the patient when and how 
much of the medication to take, for example, blister packed oral dosage forms with 
drug information leafl ets and contraceptive pills [6, 7] . 
The selection of packaging material for any pharmaceutical product is as important 
as proper pharmaceutical dosage form. To guarantee the safe and adequate 
delivery of drug product to the patient and improve patient compliance, the manufacturer 
should consider the following factors: 
1. Compatibility and safety concerns raised by the route of administration of the 
drug product and the nature of the dosage form (e.g., solid or liquid based) 
2. Kinds of protection the container closure system should provide to the dosage 
form (e.g., photosensitive, hygroscopic, easily oxidized drug products) 
3. Potential effect of any treatment or handling that may be unique to the drug 
product in the packaging system 
4. Patient compliance to the treatment and ease of drug administration 
5. Safety, effi cacy, and quality of drug product throughout its shelf - life 
The acquisition, handling, and quality control of primary and secondary packaging 
materials and of printed materials should be accomplished in the same way as 
that for the raw materials. The printed materials should be stocked in a reserved 
place so the possibility of unauthorized access is avoided. The labels and other 
rejected printed materials should be stored and transported with proper identifi cation 
before being destroyed. There should be a destruction record of the printed 
materials. Each batch of printed material and packaging material should receive a 
specifi c reference number for identifi cation. 
The identifi cation affi xed on the containers, on the equipment, in the facilities, 
and on the product containers should be clear, without ambiguity, and in a format 
approved by the company and contain the necessary data. Besides the text, differentiated 
colors indicating its condition could be used (e.g., in quarantine, approved, 
rejected, and cleaned). 
The packing materials should attend to the specifi cations, giving emphasis to the 
compatibility of the same with the pharmaceutical product that it contains. The 
material should be examined with relation to visible physical and critical defects as 
well as the required specifi cations. 

PACKAGING MATERIALS 163 
3.2.2.2 Glass as Packaging Material 
A packaging system found acceptable for one drug product may not be appropriate 
for another. Each application should contain enough information to show that each 
proposed container closure system and its components are suitable for the intended 
use. 
Nonsterile Products 
Solids Some topical drug products such as powders may be considered for marketing 
in glass bottles with appropriate dispenser. These topical drug products may be 
sterile and could be subject to microbial limits. 
The most common glass - packed solid oral dosage forms are oral powders and 
granules for reconstitution. A typical solid oral dosage form container closure system 
is a glass bottle (although plastic bottles are also used) with a screw - on or snap - off 
closure. A typical closure consists of a metal cap, often with a liner and frequently 
with an inner seal. 
The dry powders that are reconstituted in their marketed container need not be 
sterile; however, the possibility of an interaction between the packaging components 
and the reconstituting fl uid can’t be discarded. Although the contact time will be 
relatively short when compared to the component/dosage form contact time for 
liquid - based oral dosage forms, it should still be taken into consideration when the 
compatibility and safety of the container closure system are being evaluated. 
Powders for oral administration that are reconstituted in their market container, 
however, have an additional possibility of interaction between the packaging components 
and the reconstituting fl uid. Although the contact time will be relatively 
short when compared to the component/dosage form contact time for liquid - based 
oral dosage forms, it should still be taken into consideration when the compatibility 
and safety of the container closure system are being evaluated. 
Nonsolids For nonsterile products the preservative provides some protection, but 
continual microbial challenge will diminish the effi cacy of the preservative, and 
spoilage or disease transmission may occur [8] . 
Antimicrobial preservatives such as phenylmercuric acetate are known to partition 
into rubbers during storage, thus reducing the formulation concentration below 
effective antimicrobial levels [9] . A complication of modern packaging is the need 
for the application of security seals to protect against deliberate adulteration and 
maintain consumer confi dence. 
Sterile Products The sterile dosage forms share the common attributes that they 
are generally solutions, emulsions, or suspensions and are all required to be sterile. 
Injectable dosage forms represent one of the highest risk drug products (Table 1 ). 
Any contaminants present (as a result of contact with a packaging component or 
due to the packaging system ’ s failure to provide adequate protection) can be rapidly 
and completely introduced into the patient ’ s general circulation. Injectable drug 
products may be liquids in the form of solutions, emulsions, or suspensions or dry 
solids that are to be combined with an appropriate vehicle to yield a solution or 
suspension. 

164 PACKAGING AND LABELING 
Although ophthalmic drug products can be considered topical products, they 
have been grouped here with injectables because they are required to be sterile and 
the descriptive, suitability, and quality control information is typically the same as 
that for an injectable drug product. 
The potential effects of packaging component/dosage form interactions are 
numerous. Hemolytic effects may result from a decrease in tonicity and pyrogenic 
effects may result from the presence of impurities. The potency of the drug product 
or concentration of the antimicrobial preservatives may decrease due to adsorption 
or absorption. 
A cosolvent system essential to the solubilization of a poorly soluble drug can 
also serve as a potent extractant of plastic additives. 
A disposable syringe may be made of plastic, glass, rubber, and metal components, 
and such multicomponent construction provides a potential for interaction 
that is greater than when a container consists of a single material. 
Injectable drug products require protection from microbial contamination (loss 
of sterility or added bioburden) and may also need to be protected from light or 
exposure to gases (e.g., oxygen). 
Performance of a syringe is usually addressed by establishing the force to initiate 
and maintain plunger movement down the barrel and the capability of the syringe 
to deliver the labeled amount of the drug product. 
Solids For solids that must be dissolved or dispersed in an appropriate diluent 
before being injected, the diluent may be in the same container closure system (e. 
g., a two - part vial) or be part of the same market package (e.g., a kit containing a 
vial of diluent). 
Sterile powders or powders for injection may need to be protected from exposure 
to water vapor. For elastomeric components, data showing that a component meets 
the requirements of U.S. Pharmacopeia (USP) elastomeric closures for injections 
will typically be considered suffi cient evidence of safety. 
Nonsolids The package must prevent the entry of organisms; for example, packaging 
of sterile products must be absolutely microorganism proof — hence the continued 
use of glass ampules. Liquid injections are classifi ed as small - volume parenterals 
(SVPs), if they have a solution volume of 100 mL or less, or as LVPs, if the solution 
volume exceeds 100 mL [10] . Liquid - based injectables may need to be protected 
from solvent loss. 
An SVP may be packaged in a vial or an ampule. An LVP may be packaged in 
a vial, a glass bottle or, in some cases, as a disposable syringe. Packaging material 
for vials, and ampules are usually composed of type I or II glass. Stoppers and septa 
in cartridges, and vials are typically composed of elastomeric materials. 
Pharmaceuticals may interact with packaging and containers, resulting in the loss 
of drug substances by adsorption onto and absorption into container components 
and the incorporation of container components into pharmaceuticals. Diazepam in 
intravenous fl uid containers and administration sets exhibited a loss during storage 
due to adsorption onto glass [11, 12] . 
Glass surfaces are also known to adsorb drug substances. Chloroquine solutions 
in glass containers decreased in concentration owing to adsorption of the drug onto 
the glass [13] . 

PACKAGING MATERIALS 165 
Rubber closures are also known to absorb materials, including drugs. Absorption 
of preservatives such as chlorocresol into the rubber closures of injectable formulations 
has been studied extensively [13] . 
The water permeability of rubber closures used in injection vials is considered 
an important parameter in assessing the closures, but quantitative prediction of 
water permeability through rubber closures is diffi cult because the diffusion coeffi - 
cient of water is dependent on relative humidity [14] . 
Liquid - based oral drug products are usually dispensed in glass bottles (sometimes 
in plastic), often with a screw cap with a liner, and possibly with a tamper - resistant 
seal or an overcap that is welded onto the bottle. The same cap liners and inner 
seals are sometimes used with solid oral dosage forms. A laminated material can be 
used to overwrap glass bottles for extra safety. 
The USP - grade glass packaging components are chemically resistant and can be 
considered suffi cient evidence of safety and compatibility. In some cases (e.g., for 
some chelating agents), a glass packaging component may need to meet additional 
criteria to ensure the absence of signifi cant interactions between the packaging 
component and the dosage form. 
Several ophthalmic preparations are commercialized in glass containers. Although 
the risk factors associated with ophthalmic preparations are generally considered 
to be lower than for injectables, any potential for causing harm to the eyes demands 
caution. 
A large - volume intraocular solution (for irrigation) may be packaged in a polyolefi 
n (polyethylene and/or polypropylene) container. 
The liquid - based oral dosage forms may be marketed in multiple - unit bottles. 
The dosage form may be used as is or admixed fi rst with a compatible diluent or 
dispersant. Liquid - based oral drug products in glass container must meet the requirements 
for USP containers. Glass containers are accepted as suffi cient evidence of 
safety and compatibility. Performance is typically not a factor for liquid - based oral 
drug products but should be considered while treating pressurized liquid - based oral 
drug products (e.g., elixir spray). 
Topical dosage forms such as unpressurized sprays, lotions, ointments, solutions, 
and suspensions may be considered for marketing in glass bottles with appropriate 
dispenser. Some topical drug products, especially ophthalmic, are sterile or may be 
subject to microbial limits. In these cases, packaging material and handling should 
be done as those for injectables. 
3.2.2.3 Plastic as Packaging Material 
For plastic components, data from USP biological reactivity tests will typically be 
considered suffi cient evidence of safety. Whenever possible, extraction studies 
should be performed using the drug product. If the extraction properties of the drug 
product vehicle may reasonably be expected to differ from that of water (e.g., due 
to high or low pH or to a solubilizing excipient), then drug product should be used 
as the extracting medium. If the drug substance signifi cantly affects extraction characteristics, 
it may be necessary to perform the extractions using the drug product 
vehicle. If the total extract signifi cantly exceeds the amount obtained from water 
extraction, then an extraction profi le should be obtained. It may be advisable 
to obtain a quantitative extraction profi le of an elastomeric or plastic packaging 

166 PACKAGING AND LABELING 
component and to compare this periodically to the profi le from a new batch of the 
packaging component. Extractables should be identifi ed whenever possible. 
Nonsterile Products 
Solids The most common solid oral dosage forms are capsules and tablets. A 
typical solid oral dosage forms container closure system is a plastic, usually high - 
density polyethylene (HDPE), bottle with a screw - on or snap - off closure and a 
fl exible packaging system such as a pouch or a blister package. A typical closure 
consists of a cap, often with a liner, frequently with an inner seal. If used, fi llers, 
desiccants, and other absorbent materials are considered primary packaging 
components. 
A change in the selection of packing materials combined with a change in storage 
conditions or conditions during administration of the drug products may provoke 
stability problems. 
Many studies have been conducted on predicting the role of packaging in moisture 
adsorption by dosage forms. Adsorption of moisture by tablets contained in 
polypropylene fi lms was successfully modeled from storage temperature and the 
difference in water vapor pressure between the inside and outside of the packaging 
[15] . 
Chemical and physical degradation of packaged dosage forms caused by moisture 
adsorption has been predicted from the moisture permeability of the packaging. For 
example, strength changes of lactose – corn starch tablets in strip packaging [16] and 
discoloration of sugar - coated tablets of ascorbic acid [17, 18] were predicted using 
the moisture permeability coeffi cient of the packaging. 
Typical fl exible forms of packaging containing solid oral dosage forms are the 
blister package and the pouch. A blister package usually consists of a lidding material 
and a forming fi lm. The lidding material is usually a laminate which includes a 
barrier layer (e.g., aluminum foil) with a print primer on one side and a sealing agent 
(e.g., a heat - sealing lacquer) on the other side. 
The sealing agent contacts the dosage form and the forming fi lm. The forming 
fi lm may be a single fi lm, a coated fi lm, or a laminate. A pouch typically consists of 
fi lm or laminate which is sealed at the edges by heat or adhesive. 
Solid oral dosage forms generally need to be protected from the potential adverse 
effects of the following: 
1. Water vapor (e.g., moisture may affect the decomposition rate of the active 
drug substance or the dissolution rate of the dosage form) 
2. Incident light (e.g., in case of photosensitive products) 
3. Reactive gases (e.g., oxygen could provoke oxidative reactions) 
Carefully selected packaging material may help protect drug products. For 
example, a blister or pouch and use of secondary packing may be used to protect 
pack photosensitive material, especially when a dark polymeric fi lm with a covering 
lid made of aluminum is used for blister packing. Blister packaging using multilayer 
HDPE material and selection of an adequate sealing technique may help prevent 
moisture in the blister system. However, plastics and glass for packaging of solid 
oral dosage forms and for powders for reconstitution should meet the requirements 

PACKAGING MATERIALS 167 
of the USP container test. Incorporating oxygen adsorbents such as iron powder 
into packaging units can reduce the effect of oxygen. Protection from light can be 
achieved using primary packaging (packaging that is in direct contact with the 
dosage forms) and secondary packaging made of light - resistant materials. May be 
involved in the photolytic degradation kinetics. The velocity of the photochemical 
reaction may be affected not only by the light source, intensity, and wavelength of 
the light but also by the size, shape, composition, and color of the container. 
Great effort should be taken to stabilize a formulation in such a way that the 
shelf life becomes independent of the storage conditions. The photostability of drugs 
and excipients should be evaluated at the formulation development stage in order 
to assess the effects of packaging on the stability of the fi nal product. Molsidomine 
tablet preparations in inadequate primary containers (blister) without secondary 
containers when exposed to irradiation may produce morpholine. These results 
illustrate the importance of packaging for the stability of molsidomine [19] . 
Three standard tests for water vapor permeation have been established by the 
USP for use with solid oral dosage forms. 
1. Polyethylene containers (USP . 661 . ) [10] 
2. Single - unit containers and unit - dose containers for capsules and tablets (USP 
. 671 . ) 
3. Multiple - unit containers for capsules and tablets (USP . 671 . ) [10] 
The cotton and rayon used as fi llers in solid oral dosage form containers may not 
meet pharmacopeial standards, but through appropriate tests and acceptance criteria 
for identifi cation and moisture content, their adequacy should be shown. For 
example, rayon has been found to be a potential source of dissolution problems for 
gelatin capsules and gelatin - coated tablets. 
Desiccants are often used to eliminate moisture in packaging when the moisture 
resistance of the packaging is not suffi cient to prevent exposure. The utility of desiccants 
has been assessed based on a sorption – desorption moisture transfer model 
[20] . 
Desiccants or other absorbent materials are primary packaging component. The 
components should differ in shape and/or size from the tablets or capsules with 
which they are packaged. Their composition should be provided and their inertness 
should be proved through appropriate tests, and acceptance criteria should be 
established. 
A topical powder product may be marketed in a sifter - top container made of 
fl exible plastic tubes or as part of a sterile dressing (e.g., antibacterial product). The 
topical formulations in a collapsible tube can be constructed from low - density polyethylene 
(LDPE), with or without a laminated material. Normally, there is no 
product contact with the cap during storage. Thus usually there is no cap liner, 
especially in collapsible polypropylene screw caps. Normally separate applicator 
devices are made from LDPE. Product contact is possible if the applicator is part 
of the closure, and therefore an applicator ’ s compatibility with the drug product 
should be established, as appropriate (e.g., vaginal applicators). 
Nonsolids Typical liquid - based oral dosage forms are elixirs, emulsions, extracts, 
fl uid extracts, solutions, gels, syrups, spirits, tinctures, aromatic waters, and suspen

168 PACKAGING AND LABELING 
sions. These products are usually nonsterile but typically need to be protected from 
solvent loss, microbial contamination, and sometimes exposure to light or reactive 
gases (e.g., oxygen). 
The presence of a liquid phase implies a signifi cant potential for the transfer of 
materials from a packaging component into the dosage form. 
The higher viscosity of semisolid dosage forms and transdermal systems may 
cause the rate of migration of leachable substances into these dosage forms to be 
slower than for aqueous solutions. Due to extended contact, the amount of leachables 
in these drug products may depend more on a leachable material ’ s affi nity for 
the liquid/semisolid phase than on the rate of migration. 
In addition to absorption onto and absorption into containers, transfer of 
container components into pharmaceuticals may affect the perceived stability/ 
quality of drug dosage forms. Adsorption of volatile components from rubber 
closures onto freeze - dried parenterals during both dosage form processing and 
storage brought about haze formation upon reconstitution [21 – 23] . Leaching 
of dioctyl phthalate, a plasticizer used especially in polyvingl chloride (PVC) 
plastics, into intravenous solutions containing surfactants was observed [24, 25] . 
Plastics contain additives to enhance polymer performance. PVC may contain 
phthalate diester plasticizer, which can leach into infusion fl uids from packaging 
[26] . 
The liquid - based oral dosage forms may be marketed in multiple - unit bottles or 
in unit - dose or single - use pouches or cups. The dosage form may be used as is or 
admixed fi rst with a compatible diluent or dispersant. A liquid - based oral drug 
pouch may be a single - layer plastic or a laminated material. The pouches may use 
an overwrap, which is usually a laminated material. 
For LDPE components, data from USP container tests are typically considered 
suffi cient evidence of compatibility. The USP general chapters do not specifi cally 
address safety for polyethylene (HDPE or LDPE), polypropylene (PP), or laminate 
components. 
In such cases, an appropriate reference to the indirect food additive regulations 
[27] is typically considered suffi cient. This reference is considered valid for liquid - 
based oral dosage forms which the patient will take only for a relatively short 
time. 
For liquid - based oral drug products which the patient will continue to take for 
an extended period, that is, months or years, and is expected to extract greater 
amounts of substances from plastic packaging components than from water (presence 
of cosolvents), additional extractable information may be needed to address 
safety issues. 
Topical dosage forms such as creams, emulsions, gels, lotions, ointments, pastes, 
and powders may be marketed in plastic materials. Topical dosage formulations are 
for local (not systemic) effect and are generally applied to the skin or oral mucosal 
surfaces. Some vaginal and rectal creams and nasal, otic, and ophthalmic solutions 
may be considered for topical drug products. 
A rigid bottle, a collapsible tube, or a fl exible pouch made of plastic material may 
be used for liquid - based topical product. These preparations are marketed in a 
single - or multiple - unit container. For example, dissolved drug (or any substance, 
e.g., benzocaine) may diffuse in the suppository base and can, for instance, partition 
into polyethylene linings of the suppository wrap. 

PACKAGING MATERIALS 169 
Topical delivery systems are self - contained, discrete dosage forms that are 
designed to deliver drug for an extended period via intact skin or body surface, for 
example, transdermal, ocular, and intrauterine. 
These systems may be constructed of a plastic or polymeric material loaded with 
active ingredients or a coated metal. Each of these systems is generally marketed 
in a single - unit soft blister pack or a preformed tray with a preformed cover or 
overwrap. The compatibility and safety for topical delivery systems are addressed 
in the same manner as for topical drug products. Performance and quality control 
should be addressed for the rate - controlling membrane. 
Sterile Products 
Nonsolids An SVP may be packaged in a disposable cartridge, a disposable syringe, 
or a fl exible bag made of polymeric plastic. Flexible bags are typically constructed 
with multilayered plastic (Table 2 ). 
An LVP may be packaged in a vial, a fl exible bag, or, in some cases, a disposable 
syringe. Packaging material for cartridges, syringes, vials, and ampules are usually 
composed of polypropylene (Table 2 ). 
Stoppers and septa in cartridges and syringes are typically composed of elastomeric 
materials. An overwrap may be used with fl exible bags to retard solvent loss 
and to protect the fl exible packaging system from rough handling. 
Diazepam in intravenous fl uid containers and administration sets exhibited 
a loss during storage due to adsorption onto and absorption into plastics 
[11, 12] . 
Absorption of clomethiazole edisylate and thiopental sodium into PVC infusion 
bags was observed [28] . 
The pH dependence of adsorption/absorption of acidic drug substances such as 
warfarin and thiopental and basic drug substances such as chlorpromazine and diltiazem 
indicates that only the un - ionized form of the drug substance is adsorbed 
onto or absorbed into PVC infusion bags [29] . 
The absorption was correlated to the octanol – water partition coeffi cients of the 
drugs, suggesting that prediction of absorption from partition data is possible [30, 
31] . Polymers such as Nylon - 6 (polycaprolactam) are known to adsorb drug substances 
such as benzocaine [32] . 
The ophthalmic drug products are usually solutions marketed in a LDPE bottle 
with a dropper built into the neck. A few solution products use a glass container 
due to stability concerns regarding plastic packaging components. 
3.2.2.4 Metal as Packaging Material 
Metal tubes constructed of a single material are the packaging material of choice 
for topical dosage forms and may be tested readily for stability with a product. Tubes 
with a coating, however, present additional problems. The inertness of coating material 
must be established through adequate tests and guarantee that it completely 
covers underlying material. The coating material must be resistant to creaking, 
leaking, leaching, and solvent erosion. For example, frequently used aluminum tubes 
have demonstrated reactivity with fatty alcohol emulsions, mercurial compounds, 
and preparations with pH below 6.5 and above 8.0. Nonreactive, epoxy linings have 
been found to make aluminum tubes resistant to attack [6] . 

170 PACKAGING AND LABELING 
TABLE 2 Parenteral Drug Administration Devices 
Sterile Device Plastic Material 
Containers for blood products Polyvinyl chloride 
Disposable syringes Polycarbonate, polyethylene, polypropylene 
Irrigating solution containers Polyethylene, polypropylene, polyvinyl chloride 
Intravenous infusion fl uid containers Polyethylene, polypropylene, polyvinyl chloride 
Administration sets Nylon (spike), polyvinyl chloride (tubing), 
polymethylmethacrylate (needle adapter), 
polypropylene (clamp) 
Catheter Tefl on, polypropylene, thermoplastic elastomers 
Source : From ref. 6 . 
Some examples of plastic additives and parenteral drug administration devices used 
as packaging materials for sterile products can be seen in Tables 1 and 2 . 
Ophthalmic ointments are marketed in a metal tube with an ophthalmic tip. 
Ophthalmic ointments that are reactive toward metal may be packaged in a tube 
lined with an epoxy or vinyl plastic coating. 
Metal containers, pressurized or not, may also be used for topical drug products. 
Topical dosage forms include aerosols, emulsions, gels, powders, and solutions 
and may be marketed in metallic fl asks, pressurized or not. Topical dosage formulations 
are for local (not systemic) effect and are generally applied to the skin or 
oral mucosal surfaces. Some vaginal and rectal creams and nasal and otic spray 
drug products may be considered for marketing in metallic containers for topical 
use. 
A number of topical products marketed as a pressurized aerosol may be dispensed 
in a metallic bottle with a screw cap. Topical dosage forms in aluminum tubes 
usually include a liner. A tube liner is frequently a lacquer or shellac whose composition 
should be stated. A metallic pressurized packaging system for a liquid - 
TABLE 1 Plastic Additives 
Type Purpose Examples 
Lubricants Improve processability Stearic acid paraffi n waxes, polyethylene 
(PE) waxes 
Stabilizers Retard degradation Epoxy compounds, organotins, mixed 
metals 
Plasticizers Enhance fl exibility, 
resiliency, melt fl ow 
Phthalates 
Antioxidants Prevent oxidative 
degradation 
Hindered phenolics (BHT), aromatic 
amines, thioesters, phosphites 
Antistatic agents Minimize surface static 
charge 
Quaternary ammonium compounds 
Slip agents Minimize coeffi cient of 
friction, especially 
polyolefi ns 
Dyes, pigments Color additives 
Source : From ref. 6 . 

PACKAGING MATERIALS 171 
based topical product may deter solvent loss and may provide protection from light 
when appropriate. 
The droplet size of topical aerosol sprays does not need to be carefully controlled, 
and the dose usually is not metered as in inhalers. The spray may be used to apply 
the drug to the skin (topical aerosol) or mouth (lingual aerosol) and the functionality 
of the sprayer should be addressed. The drug product has no contact with the 
cap and short - term contact with the nozzle. A topical aerosol may be sterile or may 
conform to acceptance criteria for microbial limits. However, the physical stability 
of aerosols can lead to changes in total drug delivered per dose and total number 
of doses that may be obtained from the container. 
3.2.2.5 Applications: Some Examples 
Many research papers in the scientifi c literature present studies showing the importance 
of the effect of packaging materials on the stability of pharmaceutical and 
cosmetic preparations: 
1. Santoro and co - workers [33] presented results of the stability of oral rehydration 
salts (ORSs) in different types of packaging materials. The objective of the 
research was to give guidance on the adequate choice of packaging material presenting 
the indispensable characteristics in order to protect ORS preparation. This 
pharmaceutical preparation is essential to children living in developing countries 
with tropical climate and its distribution is one of the programs of the World Health 
Organization (WHO) [34] . 
It has been proved in several research papers that water is the most important 
factor in the component ’ s degradation of ORSs. To proceed with the study, the 
pharmaceutical formulation was prepared by a pharmaceutical manufacture. The 
batch was packed in six types of packaging material. After storage of samples for 
36 weeks maintained at ambient temperature, at ambient temperature and 76% 
relative humidity, and at 40 ° C with 80% relative humidity, analyses of water determination 
were made at different intervals of time. Water determination was performed 
by loss on drying at 50 ° C and Karl Fisher methods. 
The studied ORS preparation contained anhydrous glucose (20 g), sodium chloride 
(3.5 g), trisodium dehydrate citrate (2.9 g), and potassium chloride (1.5 g) 
According to the results, the packaging material that better protected the ORS 
preparation is the one constituted of polyester (18 g), aluminum (35%), and polyethylene 
(50 g). 
2. The effect of packaging materials on the stability of sunscreen emulsions was 
also studied by Santoro and co - workers [35, 36] . The purpose of the research was 
to study the stability of an emulsion containing UVA, UVB, and infrared sunscreens 
after storage in different types of packaging materials (glass and plastic fl asks, plastic 
and metallic tubes). The samples, emulsions containing benzophenone - 3 (B - 3), octyl 
methoxycinnamate (OM), and Phycocorail , were stored at 10, 25, 35, and 45 ° C and 
representative samples were analyzed after 2, 7, 30, 60, and 90 days. Stability studies 
were conducted by analyzing samples at predetermined intervals by high - 
performance liquid chromatography (HPLC) along with periodic rheological 
measurements. 
The proposed HPLC method enabled the separation and quantitative determination 
of B - 3 and OM present in sunscreens. The method was successfully applied in 

172 PACKAGING AND LABELING 
the stability studies of the emulsions. The method is simple, precise, and accurate; 
there was no interference from formulation components. The sample emulsions 
stored at different temperatures presented similar rheological behavior, at least 
during the period of the study (three months). Most of the samples showed a pseudoplastic 
non - Newtonian thixotropic profi le. There were no signifi cant changes in 
the physical and chemical stability of emulsions stored in different packaging material. 
The studied glass and plastic packaging materials were found adequate for 
storing referred solar protector emulsions. 
3. Sarbach and co-workers [48] , studied the effect of plastics packaging materials 
on parenteral pharmaceutical products. Compatibility studies of these containers 
with different contents are required for drug registration. The authors demonstrated 
the migration phenomena which occurred between a trilaminated fi lm and a parenteral 
solution of metronidazole at 0.5%. The main migration products found in 
the solution were e - caprolactam and a phthalic derivative. The authors also separated 
several unidentifi ed compounds probably coming from the polyurethane 
adhesive. 
4. Molsidomine is sensitive to light and shows a fast decomposition in solutions 
and in tablets. Thoma and co - workers [37] showed the importance of light - resistant 
packaging material for photolabile pharmaceuticals. They irradiated molsidomine 
preparations over a period of 72 h in a light cabinet according to storage at daylight 
for about 4 – 6 weeks. Losses of 23 – 90% in tablets and 43 – 60% in solutions were 
found. The photodegradation could be overcome by selection of suitable packaging 
materials, colorants or vanillin. The degradation product morpholine after dansylation 
was determined by HPLC and showed contents of 0.10 – 0.67 mg in tablets and 
0.10 – 0.38 mg/mL in solution after irradiation. 
These examples, among many others described in the scientifi c literature, illustrate 
the importance of proper selection of packaging material for the stability and 
effectiveness of pharmaceutical dosage forms. 
3.2.3 QUALITY CONTROL OF PACKAGING MATERIAL 
3.2.3.1 General Considerations 
Several regulatory agencies as well as private agencies [Food and Drug Administration 
(FDA), British Pharmacopoeia, WHO, USP] [4, 10, 34, 38] have issued guidelines 
on the safety evaluation of materials and container closure systems. However, 
the ultimate proof of the safety and suitability of a container closure system and 
the packaging process is established by full shelf life stability studies. An important 
step in such evaluations is characterization of the packaging materials and the 
chemicals that can migrate or extract from container closure system components to 
the drug product. This extractable material belongs to diverse chemical classes that 
can migrate from polymeric materials, such as antioxidants, contaminants, lubricants, 
monomers, plasticizers, and preservatives. Such basic information is critical to understanding 
the biological safety and suitability of a container. 
Establishing the safety of container closure systems is of key importance to the 
medical and pharmaceutical industries (Table 3 ). It is no less important than the 
contents themselves. The FDA ’ s document “ Guidance on Container Closure Systems 
for Packaging Human Drugs and Biologics ” makes this point clear [4] . 

The FDA ’ s guidance document requires the evaluation of four attributes to 
establish suitability: protection, compatibility, safety, and performance/drug delivery. 
The document also provides a structured approach to ranking packaging concerns 
according to the route of drug administration and likelihood of packaging component 
– dosage form interaction. A container closure system acceptable for one drug 
product cannot be assumed to be appropriate for another. Each product should have 
suffi cient information to establish that a container and its components are right for 
their intended use [4] . 
To establish suitability, all four attributes must be evaluated and be shown to 
pose no concern to the drug product or to product performance. Suitability refers 
to the tests used for the initial qualifi cation of the container closure system with 
regard to its intended use. The guidance defi nes what tests must be done to evaluate 
each of the attributes of suitability. 
While the tests and methods described in Table 4 allow one to provide data that 
the container closure system is suitable for its intended use, an application must also 
describe the quality control (QC) measures that will be used to ensure consistency 
in the packaging components. The principal considerations for the QC measures are 
the physical characteristics and the chemical composition. By choosing two or three 
of the tests done in the initial suitability study, a QC program can be established 
that will ensure the consistency of the container closure system (Table 4 ). 
Protection A container closure system should provide the dosage form with adequate 
protection from factors (e.g., temperature, light) that can cause a degradation 
in the quality of that dosage form over its shelf life. Common causes of such degradation 
are exposure to light, loss of solvent, exposure to reactive gases (e.g., oxygen), 
absorption of water vapor, and microbial contamination. 
A container intended to provide protection from light or offered as a light - resistant 
container must meet the requirements of the USP . 661 . light transmission test. 
The procedure requires the use of a spectrophotometer, with the required sensitivity 
TABLE 3 Examples of Packaging Concerns for Common Classes of Drug Products 
Degree of Concern 
Associated with 
Route of 
Administration 
Likelihood of Packaging Component – Dosage Form Interaction 
High Medium Low 
Highest Inhalation aerosols and 
solutions; injection; 
injectable suspensions 
Sterile powders 
and powders 
for injections 
and inhalation 
powders 
High Ophthalmic solutions and 
suspensions; transdermal 
ointments and patches; 
nasal aerosols and sprays 
Low Topical solutions and 
suspensions; topical and 
lingual aerosols; oral 
solutions and suspensions 
Topical powders; 
oral powders 
Oral tablets and 
oral (hard and 
soft; gelatin) 
capsules 
QUALITY CONTROL OF PACKAGING MATERIAL 173

174 PACKAGING AND LABELING 
and accuracy, adapted for measuring the amount of light transmitted by the plastic 
material used for the container. 
The ability of a container closure system to protect against moisture can be 
ascertained by performing the USP . 661 . water vapor permeation test. The USP 
sets limits on the amount of moisture that can penetrate based upon the size and 
composition of the plastic components [HDPE, LDPE, or polyethylene terephthalate 
(PET)]. 
Evaluating the integrity of the container can be done in several ways. Two of the 
most common tests are dye penetration and microbial ingress. Container closure 
systems stored in a dye solution and exposed to pressure and vacuum cycles are 
examined for dye leakage into the container. The microbial ingress is similar in 
fashion but determines the microbial contamination of the contents when soaked 
in a media contaminated with bacteria. Other quantitative tests that can be run are 
vacuum/pressure decay, helium mass spectrometry, and gas detection. 
Compatibility Packaging components that are compatible with a dosage form will 
not interact suffi ciently to cause unacceptable changes in the quality of either the 
dosage form or the packaging component. A leachability study designed to evaluate 
the amount and/or nature of any chemical migrating from the plastic material to 
the drug product should be considered. The study should evaluate substances that 
migrate into the drug product vehicle for the length of shelf life. The drug product 
should be evaluated at regular intervals, such as at one, three, or six months or one 
or two years, until the length of the shelf - life claim has been met. 
Analytical techniques such as liquid chromatography/mass spectrometry (LC/ 
MS) to evaluate nonvolatile organics, gas chromatography/mass spectrometry (GC/ 
MS) to evaluate semivolatile organics, and inductively coupled plasma (ICP) spectroscopy 
to detect and quantitate inorganic elements should be a part of this 
study. 
Unknown impurities and degradation products can be identifi ed using liquid or 
gas chromatography along with MS. Information or substances identifi ed from 
extractable chemical evaluation can be used to help prepare standards specifi c for 
the plastic container being studied during leachability studies. Development and 
validation of the selective analytical method should be thoroughly studied before 
its application in the detection of leachable chemicals in active drug substance and 
drug product. 
Organoleptic and chemical changes such as precipitates, discoloration, strange 
odor, and pH modifi cation are signs of degradation of drug product. Changes in the 
physical characteristics of the container, such as brittleness, should be evaluated 
using thermal analysis and hardness testing. An infrared spectroscopic scan can fi ngerprint 
the materials and also provide proof of identity. Spectrophotometry and 
LC with ultraviolet detection can be used for the analysis of drug product stored at 
different stress conditions. These tests can be used for the quality control of drug 
product as well as for conducting stability studies on different products stored in 
the same container material. 
Safety Packaging components should be constructed of materials that will not 
leach harmful or undesirable amounts of substances to which a patient will be 
exposed when being treated with the drug product. This consideration is especially 

important for those packaging components which may be in direct contact with the 
dosage form, but it is also applicable to any component from which substances may 
migrate into the dosage form (e.g., an ink or adhesive). Determining the safety of 
a packaging component is not a simple process, and a standardized approach has 
not been established. However, an extraction study should be one of the fi rst 
considerations. 
A good knowledge regarding possible extractable material could help analysts 
develop specifi c and selective methods to identify extractables from container 
closure components under various control extraction study conditions. 
Precise information on the synthesis of the polymer and descriptions of the 
monomers used in the polymerization, the solvents used in the synthesis, and the 
special additives that have been added during material production as well as knowledge 
of degradation products that may be released into the drug product are also 
important. 
Some potential extractable chemicals from packaging materials are water soluble, 
while others are soluble only in nonpolar environments. The USP includes physicochemical 
tests for plastics based on water extracts, while water, alcohol, and hexane 
extracts are required for polyethylene containers under controlled temperature and 
time parameters (70 ° C for 24 h for water and alcohol and 50 ° C for 24 h for hexane). 
The USP physicochemical tests for extractables should be a part of all suitability 
programs, regardless of the criticality of the drug dosage form. USP elastomeric 
closures for injections should also be a part of the extractables study to establish 
safety. These USP tests, which have evolved over many years, are relevant, sensitive, 
rapid, and inexpensive. They help establish material safety. 
TABLE 4 Properties of Suitability Concerns and Interactions 
Attributes Concerns and Interactions Proposed Methods 
Protection Exposure to light, moisture, 
microbial ingress, and 
oxidation from presence 
of oxygen 
USP . 661 . light transmission and water 
vapor permeation, container integrity 
(microbial ingress, dye penetration, 
helium leak) 
Compatibility Leachable induced 
degradation, absorption 
or adsorption of drug, 
precipitation, change in 
pH, discoloration, 
brittleness of packaging 
materials 
Leachability study (migration of chemicals 
into drug product) using LC/MS, GC/MS, 
ICP/AA, pH, appearance of drug and 
container, thermal analysis (DSC, TGA), 
and infrared (IR) 
Safety No leached harmful or 
undesirable amounts of 
substances to expose 
patients treated with drug 
Extraction study (USP physicochemical 
tests – plastics), USP elastomeric closures 
for injections, toxicological evaluation, 
USP biological reactivity and complies 
with CFR, additives and purity 
Performance Container closure system 
functionality, drug 
delivery 
Functionality (improved patient compliance 
or use), delivery (transfer dose in right 
amount or rate) 
Abbreviations : DSC, differential scanning calorimetry; ICP, Inductively coupled plasma spectrometer; 
AA, Atomic absorption. 
Source : From ref. 39 . 
QUALITY CONTROL OF PACKAGING MATERIAL 175

176 PACKAGING AND LABELING 
The safety of material can be guaranteed by using appropriate analytical methods 
and instrumentation to identify and quantitate extracted chemicals. Liquid and gas 
chromatography and MS are powerful analytical tools that can separate and quantitate 
volatile and nonvolatile chemicals along with useful structural information. 
The mass spectrum or fragmentation pattern acquired for each molecule makes 
these excellent and effective tools for identifying unknown impurities or degradation 
products. 
Toxicological evaluation of identifi ed and unidentifi ed impurities from a container 
can help improve the safety index of drug products. The toxicological evaluation 
should take into consideration container closure system properties, drug 
product formulation, dosage form, route of administration, and dose regimen. A 
close correlation between chemical and toxicological information can provide better 
control on safety and compatibility of containers and closures. 
Performance The fourth attribute of the suitability of the container closure system, 
performance and drug delivery, refers to its ability to function in the manner for 
which it was designed. There are two major considerations when evaluating performance. 
The fi rst consideration is functionality that may improve patient compliance, 
[e.g., a two - chamber vial or intravenous (IV) bag], or improve ease of use (e.g., a 
cap that contains a counter, a prefi lled syringe). The second consideration is drug 
delivery, which is the ability of the packaging system to deliver the right amount or 
rate (e.g., a prefi lled syringe, a transdermal patch, a metered tube, a dropper or spray 
bottle, a dry - powder inhaler, and a metered - dose inhaler). 
3.2.3.2 Packaging Components 
Quality control refers to the tests typically used and accepted to establish that, after 
the application is approved, the components and the container closure system continue 
to possess the characteristics established in the suitability studies. 
To ensure consistency, protection, compatibility, safety, and performance of the 
packaging components, it is necessary to defi ne QC measures that will be used to 
ensure consistency in the packaging components. These controls are intended to 
limit unintended postapproval variations in the manufacturing procedures or materials 
of construction for a packaging component and to prevent adverse effects on 
the quality of a dosage form. 
The USP tests and studies for establishing suitability and QC of container closure 
system and for associated component materials are summarized in Table 5 . 
Hydrolysis and oxidation are the two main routes of degradation for the majority 
of drugs. To gain more information, the drug could be subjected to a range of temperature 
and relative humidity conditions. In addition, photostability studies could 
be conducted by exposure to artifi cial or natural light conditions. Elevated temperature, 
humidity, and light stress the drug and induce rapid degradation. Harmonized 
guidelines are available for new drug substances and products and may provide 
useful information to characterize degradation processes and selection of appropriate 
packaging material. 
The primary packaging must physically protect the product from the mechanical 
stresses of warehousing, handling, and distribution. Mechanical stress may take a 

TABLE 5 U.S. Pharmacopeia General Tests and Assays 
Chapter Topic 
. 1 . Injections 
. 51 . Antimicrobial preservatives — effectiveness 
. 61 . Microbial limit tests 
. 71 . Sterility tests 
. 87 . Biological reactivity tests, in vitro 
. 88 . Biological reactivity tests, in vivo 
. 161 . Transfusion and infusion assemblies 
. 381 . Elastomeric closures for injections, biological test procedures, physicochemical 
test procedures 
. 601 . Aerosols 
. 661 . Containers: light transmission; chemical resistance — glass containers; biological 
tests — plastics and other polymers; physicochemical tests — plastics; 
containers for ophthalmics — plastics; polyethylene containers; polyethylene 
terephthalate bottles and polyethylene; terephthalate G bottles; single - unit 
containers and unit - dose containers for nonsterile; solid and liquid dosage 
forms; customized patient medication packages 
. 671 . Containers — permeation: multiple - unit containers for capsules and tablets; 
single - unit containers and unit - dose containers for capsules and tablets 
. 691 . Cotton (or the monograph for purifi ed rayon USP) 
. 771 . Ophthalmic ointments 
. 1041 . Biologics 
. 1151 . Pharmaceutical dosage forms 
Source : From ref. 10 . 
variety of forms, from impact through vibration in transit and compression forces 
on stacking. 
The demands for mechanical protection will vary with product type: Glass 
ampules will require greater protection than plastic eye drop bottles, for example. 
Other protection is required from environmental factors such as moisture, temperature 
changes, light, gases, and biological agents such as microorganisms and, importantly, 
humans. 
The global market for medicinal products requires that the products are stable 
over a wide range of temperatures ranging from subzero in the polar region, 15 ° C 
in temperate zones, up to 32 ° C in the tropics. Along with this temperature variation, 
relative humidity can vary from below 50% to up to 90%, a feature that the packaging 
should be able to resist if necessary. The majority of packaging materials (including 
plastics) are to some degree permeable to moisture and the type of closure 
employed, such as screw fi ttings, may also permit ingress of moisture. The susceptibility 
of the product to moisture and its hygroscopicity will have to be considered 
and may require packaging with a desiccant or the use of specialized strip packs 
using low - permeability materials such as foil. 
Temperature fl uctuations can lead to condensation of moisture on the product 
and, with liquids, formation of a condensate layer on top of the product. This latter 
problem is well known and can lead to microbiological spoilage as the condensate 
is preservative free. 
QUALITY CONTROL OF PACKAGING MATERIAL 177

178 PACKAGING AND LABELING 
If the product is sensitive to photolysis, then opaque materials may be required. 
Most secondary packaging materials (e.g., cartons) do not transmit light, but in 
some cases specialized primary packaging designed to limit light transmission is 
employed. 
The package must also prevent the entry of organisms; for example, packaging 
of sterile products must be microorganism proof — hence the continued use of glass 
ampules. For nonsterile products the preservative provides some protection, but 
continual microbial challenge will diminish the effi cacy of the preservative, and 
spoilage or disease transmission may occur. 
The packaging material must not interact with the product either to adsorb substances 
from the product or to leach chemicals into the product. Plastics contain 
additives to enhance polymer performance. PVC may contain phthalate diester 
plasticizer, which can leach into infusion fl uids from packaging. Antimicrobial preservatives 
such as phenylmercuric acetate are known to partition into rubbers and 
plastics during storage, thus reducing the formulation concentration below effective 
antimicrobial levels. 
A complication of modern packaging is the need for the application of security 
seals to protect against deliberate adulteration and maintain consumer confi dence. 
The active products used must also be stability tested in the proposed packaging 
material. 
The FDA guidance for industry suggests considering consistency in physical and 
chemical composition. Using a few simple tests, the quality of components and ultimately 
the container closure system can be monitored. 
Physical Characteristics The physical characteristics of interest include dimensional 
criteria (e.g., shape, neck fi nish, wall thickness, design tolerances), physical 
parameters critical to the consistent manufacture of a packaging component (e.g., 
unit weight), and performance characteristics (e.g., metering valve delivery volume 
or the ease of movement of syringe plungers). Unintended variations in dimensional 
parameters, if undetected, may affect package permeability, drug delivery performance, 
or the adequacy of the seal between the container and the closure. Variation 
in any physical parameter is considered important if it can affect the quality of a 
dosage form. 
Physical considerations such as water vapor transmission to evaluate seal integrity, 
thermal analysis such as DSC to monitor melting point and glass transitions of 
plastics, and IR scanning to prove identity should be part of an ongoing quality 
control monitoring program. 
Chemical Composition The chemical composition of the materials of construction 
may affect the safety of a packaging component. New materials may result in new 
substances being extracted into the dosage form or a change in the amount of known 
extractables. The chemical composition may also affect the compatibility, functional 
characteristics, or protective properties of packaging components by changing rheological 
or other physical properties (e.g., elasticity, resistance to solvents, or gas 
permeability). 
The chemical composition should also be evaluated by performing the simple but 
informative USP physicochemical tests using water, drug product vehicle, and 
alcohol extractions of plastic components. Specifi cations should be set for nonvola

tile residue (total extractables) during the initial suitability tests and then used to 
monitor the level of polar and nonpolar extractables as part of a quality control 
plan. 
A change in the composition of packaging raw material or a change in formulation 
is considered a change in the specifi cations. Due care must be taken to guarantee 
the safety, compatibility, and performance of a new dosage form in a new 
packaging system. 
The use of stability studies for monitoring the consistency of a container closure 
system in terms of compatibility with the dosage form and the degree of protection 
provided to the dosage form is essential. Except for inhalation drug products, for 
which batch - to - batch monitoring of the extraction profi le for the polymeric and 
elastomeric components is routine, no general policy concerning the monitoring of 
a packaging system and components with regard to safety is available. 
Secondary packaging components are not intended to make contact with the 
dosage form. Examples are cartons, which are generally constructed of paper or 
plastic, and overwraps, fabricated from a single layer of plastic or from a laminate 
made of metal foil, plastic, and/or paper. In special cases, secondary packaging components 
provide some additional measure of protection to the drug product. In such 
cases it could be considered a potential source of contamination and the safety of 
the raw materials should be taken into consideration. 
3.2.3.3 Inhalation Drug Products 
Inhalation drug products include inhalation aerosols (metered - dose inhalers); inhalation 
solutions, suspensions, and sprays (administered via nebulizers); inhalation 
powders (dry - powder inhalers); and nasal sprays. The carboxymethylcellulose 
(CMC) and preclinical considerations for inhalation drug products are unique in 
that these drug products are intended for respiratory tract compromised patients. 
This is refl ected in the level of concern given to the nature of the packaging components 
that may come in contact with the dosage form or the patient (Table 4 ). 
In October 1998, the FDA issued guidance for industry regarding container 
closure systems such as metered - dose inhaler (MDI) and dry - powder Inhaler (DPI) 
drug products. 
3.2.3.4 Drug Products for Injection and Ophthalmic Drug Products 
Injectable dosage forms are sterile and represent one of the highest risk drug products. 
Injectable drug products may be liquids in the form of solutions, emulsions, 
and suspensions or dry solids that are to be combined with an appropriate vehicle 
to yield a solution or suspension. 
Cartridges, syringes, vials, and ampules are usually composed of type I or II glass 
or polypropylene frequently used to deliver SVP and LVPs. Flexible bags are typically 
constructed with multilayered plastic. Stoppers and septa in cartridges, syringes, 
and vials are typically composed of elastomeric materials. An overwrap may be used 
with fl exible bags to retard solvent loss and to protect the fl exible packaging system 
from rough handling. 
Injectable drug products require protection from microbial contamination (loss 
of sterility or added bioburden) and may also need to be protected from light or 
QUALITY CONTROL OF PACKAGING MATERIAL 179

180 PACKAGING AND LABELING 
exposure to gases (e.g., oxygen). Liquid - based injectables may need to be protected 
from solvent loss, while sterile powders or powders for injection may need to be 
protected from exposure to water vapor. 
For elastomeric components, data showing that a component meets the requirements 
of USP elastomeric closures for injections should typically be performed to 
assure safety. For plastic components, USP biological reactivity tests are recommended 
to assure evidence of safety. Whenever possible, the extraction studies 
described in USP should be performed using the drug product. Extractables should 
be identifi ed whenever possible. For a glass packaging component, data from USP 
“ Containers: Chemical resistance — Glass containers ” will typically be considered 
suffi cient evidence of safety and compatibility. In some cases (e.g., for some chelating 
agents), a glass packaging component may need to meet additional criteria to 
ensure the absence of signifi cant interactions between the packaging component 
and the dosage form. 
The performance of a syringe is usually addressed by establishing the force to 
initiate and maintain plunger movement down the barrel and the capability of the 
syringe to deliver the labeled amount of the drug product. 
Ophthalmic drug products are usually solutions marketed in a LDPE bottle with 
a dropper built into the neck or ointments marketed in a metal tube lined with an 
epoxy or vinyl plastic coating with an ophthalmic tip. 
Since ophthalmic drug products are applied to the eye, compatibility and safety 
concerns should also address the container closure system ’ s potential to form substances 
which irritate the eye or introduce particulate matter into the product (USP 
. 771 . , ophthalmic ointments). 
3.2.3.5 Liquid - Based Oral Products, Topical Drug Products, and 
Topical Delivery Systems 
The presence of a liquid phase implies a signifi cant potential for the transfer of 
materials from a packaging component into the dosage form. 
Liquid - Based Oral Drug Products Typical liquid - based oral dosage forms are 
elixirs, emulsions, extracts, fl uid extracts, solutions, gels, syrups, spirits, tinctures, 
aromatic waters, and suspensions. These products are usually nonsterile but may be 
monitored for changes in bioburden or for the presence of specifi c microbes. 
A liquid - based oral drug product typically needs to be protected from solvent 
loss, microbial contamination, and sometimes exposure to light or reactive gases 
(e.g., oxygen). For glass components, data showing that a component meets the 
requirements of USP “ Containers: Glass containers ” are accepted as suffi cient evidence 
of safety and compatibility. For LDPE components, data from USP container 
tests are typically considered suffi cient evidence of compatibility. 
The USP general chapters do not specifi cally address safety for polyethylene 
(HDPE or LDPE), PP, or laminate components. A patient ’ s exposure to substances 
extracted from a plastic packaging component (e.g., HDPE, LDPE, PP, laminated 
components) into a liquid - based oral dosage form is expected to be comparable to 
a patient ’ s exposure to the same substances through the use of the same material 
when used to package food [27] . 

Topical Drug Products Topical dosage forms include aerosols, creams, emulsions, 
gels, lotions, ointments, pastes, powders, solutions, and suspensions. These dosage 
forms are generally intended for local (not systemic) effect and are generally applied 
to the skin or oral mucosal surfaces. Topical products also include some nasal and 
otic preparations as well as some ophthalmic drug products. Some topical drug 
products are sterile and should be subject to microbial limits. 
A rigid bottle or jar is usually made of glass or polypropylene with a screw cap. 
The same cap liners and inner seals are sometimes used as with solid oral dosage 
forms. A collapsible tube is usually constructed from metal or is metal lined from 
LDPE or from a laminated material. 
Topical Delivery Systems Topical delivery systems are self - contained, discrete 
dosage forms that are designed to deliver drug via intact skin or body surface, 
namely transdermal, ocular, and intrauterine. 
Each of these systems is generally marketed in a single - unit soft blister pack or 
a preformed tray with a preformed cover or overwrap. Compatibility and safety for 
topical delivery systems are addressed in the same manner as for topical drug products. 
Performance and quality control should be addressed for the rate - controlling 
membrane. Appropriate microbial limits should be established and justifi ed for each 
delivery system. 
3.2.3.6 Solid Oral Dosage Forms and Powders for Reconstitution 
The most common solid oral dosage forms are capsules and tablets. A typical container 
closure system is a plastic (usually HDPE) or a glass bottle with a screw - on 
or snap - off closure and a fl exible packaging system, such as a pouch or a blister 
package. If used, fi llers, desiccants, and other absorbent materials are considered 
primary packaging components. 
Solid oral dosage forms generally need to be protected from the potential adverse 
effects of water vapor, light, and reactive gases. For example, the presence of moisture 
may affect the decomposition rate of the active drug substance or the dissolution 
rate of a dosage form. The potential adverse effects of water vapor can be 
determined with leak testing on a fl exible package system (pouch or blister package). 
Three standard tests for water vapor permeation have been established by the USP, 
namely polyethylene containers (USP . 661 . ), single - unit containers and unit - dose 
containers for capsules and tablets (USP . 671 . ), and multiple - unit containers for 
capsules and tablets (USP . 671 . ). 
3.2.4 IMPORTANCE OF PROPER PACKAGING AND LABELING 
The Poison Prevention Packaging Act ( www.cpsc.gov/businfo/pppa.html) requires 
special packaging of most human oral prescription drugs, oral controlled drugs, 
certain normal prescription drugs, certain dietary supplements, and many over - the - 
counter (OTC) drug preparations in order to protect the public from personal injury 
or illness from misuse of these preparations. 
IMPORTANCE OF PROPER PACKAGING AND LABELING 181

182 PACKAGING AND LABELING 
In many countries there are very strict regulations for packaging of many drug 
substances. Nevertheless, special packaging is not required for drugs dispensed 
within a hospital setting for inpatient administration. Manufacturers and packagers 
of bulk - packaged prescription drugs do not have to use special packaging if the drug 
will be repackaged by the pharmacist. 
Various types of child - resistant packages are covered in ASTM International 
standard D - 3475. Medication errors linked to poor labeling and packaging can be 
controlled through the use of error potential analysis. The recognition that a drug 
name, label, or package may constitute a hazard to safety typically occurs after the 
drug has been approved for use and is being marketed. Calls for change almost 
always result from accumulating reports of serious injuries associated with the use 
of a drug. 
Numerous reports of medication errors are being reported, some of which have 
resulted in patient injury or death. In a number of these reports, a medication was 
mistakenly administered either because the drug container (bag, ampule, prefi lled 
syringe and bottle) was similar in appearance to the intended medication ’ s container 
or because the packages had similar labeling. Obviously, the severity of such errors 
depends largely on the medication administered. 
The problem of medical errors associated with the naming, labeling, and packaging 
of pharmaceuticals is being very much discussed. Sound - alike and look - alike 
drug names and packages can lead pharmacists and nurses to unintended interchanges 
of drugs that can result in patient injury or death. Simplicity, standardization, 
differentiation, lack of duplication, and unambiguous communication are 
human factors that are relevant to the medication use process. These factors have 
often been ignored in drug naming, labeling, and packaging. 
The process for naming a marketable drug is always lengthy and complex and 
involves submission of a new entity and patent application, generic naming, brand 
naming, FDA — or other corresponding organization all over the world — review, and 
fi nal approval. Drug companies seek the fastest possible approval and may believe 
that the incremental benefi t of human factor evaluation is small. Very often, the 
drug companies are resistant to changing, for example, brand names. Although a 
variety of private - sector organizations in many countries have called for reforms in 
drug naming, labeling, and packaging standards, the problem remains. 
Drug names, labels, and packages are not selected and designed in accordance with 
human factor principles. FDA standards or other corresponding organizations in 
other countries do not require application of these principles, the drug industry has 
struggled with change, and private - sector initiatives have had only limited success. 
A number of factors can contribute to the mistaking of one medication for 
another. Failure to read the package label is one cause. Another if a medication is 
stored in the wrong location or if clinicians select the medication based solely on 
the appearance of its package. Also, confusion can occur between medications with 
names that look alike or sound alike or between premixed medications packaged 
in similar - looking containers. Another potential source for confusion with premixed 
medications is the presence of different concentrations of the same medication in 
a particular location (e.g., a package with 100 mg/mL concentration of a drug could 
be mistaken for one with 10 mg/mL concentration). 
Daily, physicians, nurses, and pharmacists base medical decisions on the information 
provided by a drug product ’ s labeling and packaging. Unfortunately, poor 

labeling and packaging have been linked all too often to medication errors. To help 
practitioners avoid errors, drug manufacturers should present information in a clear 
manner that can be grasped quickly and easily. 
To determine what presentation is most clear, manufacturers should invite and 
consider the input of physicians, nurses, and pharmacists, because they work with 
these products every day and are more likely than label and package designers to 
discover potential problems. Such input provides the basis for failure and effect 
analysis (FMEA), also known as error potential analysis or error prevention analysis. 
FMEA is a systematic process that can predict how and where systems might 
fail. Using FMEA, health care practitioners examine a product ’ s packaging or labeling 
in order to identify the ways in which it might fail. A number of steps to reduce 
confusion and improve the readability of a drug product ’ s label have already been 
determined through the use of FMEA. 
The fi rst step is to reduce label clutter. Only essential information, such as the 
brand and generic names, strength or concentration, and warnings, should appear 
prominently on the front label. Numerous deaths have been prevented through the 
addition of a warning to concentrated vials of injectable potassium chloride, for 
example. Another step includes the use of typeface to enhance distinctive portions 
of look - alike drug names on look - alike packaging. 
Medication errors are also associated with poor product packaging design. Unfortunately, 
medication errors linked to poor labeling and packaging are sometimes 
used in the health care environment to justify the damage. Participation of an expertise 
from health care practitioners, during labeling and packaging design phase, 
might have prevented several errors. 
Whether for established drugs or new entities going through the approval process, 
the principles of safe practice in naming, labeling, and packaging are the same and 
must be very well controlled. Safety experts may differ about specifi c details, but 
there is little disagreement about the fundamental principles that should be incorporated 
into the drug approval process. 
Based on reports of errors associated with packaging and labeling, many recommendations 
have been proposed. Some of them are: 
1. Avoid storing medications with similar packaging in the same location or in 
close proximity. 
2. Follow the American Society of Health System Pharmacists (ASHP) guidelines 
or other legislation of a specifi c country for preventing hospital medication 
errors [40, 41] . The ASHP ’ s recommendations include the following: 
Fully document all medication prescription and deliveries and instruct staff 
that discrepancy or misunderstanding about prescription or patient 
information should be verifi ed with the prescribing physician. Staff 
members should be told that all caregivers (regardless of level) have the 
duty to question the prescribing physician (regardless of the physician ’ s 
relative position in the hospital hierarchy) if they have concerns about 
a drug, dose, or patient. 
Periodically train staffs in practices that will help avoid medication errors. 
Ensure that the medication storage and distribution to hospital locations 
outside the pharmacy are supervised by hospital pharmacy staff only. 
IMPORTANCE OF PROPER PACKAGING AND LABELING 183

184 PACKAGING AND LABELING 
Nonpharmacists should not be allowed to enter the pharmacy if it is 
closed. 
3. Perform failure mode and effects analysis. This is a technique used to identify 
all medication errors that could occur, determine how they occur, and estimate 
what their consequences would be. Steps then should be taken to prevent 
errors from occurring, when possible, and to minimize the effects of any errors 
that do occur. 
4. Report any information relating to medication errors to the Medication Errors 
Reporting Program operated by USP convention [10] and the Institute for 
Safe Medication Practice (ISMP) or other corresponding institutions in the 
different countries. The program shares information on medication errors with 
health care professionals to prevent similar errors from recurring. 
5. Hospitals should report incidents in which a device caused or helped cause a 
medication error. 
6. Urge suppliers to provide clear and unique labels and packages for their 
various individual medications. 
Some other considerations relating to standards for drug names, labeling standards, 
and packaging standards are as follows: 
1. Standard for Drug Names. The most critical issue in drug name selection is that 
one name should not be easily confused with another. This applies to both generic and 
brand names. A name must neither sound like that of another drug nor look like 
another drug name when it is written out by hand. From the industry ’ s standpoint, the 
challenge is to fi nd a name that is easy to recollect and appropriate for the connotation 
desired, do not lead astray (safe), and not already a trade name. 
Nowadays, increasing sophisticated and effective methods are available for determining 
the likelihood of confusion by sound or sight. 
2. Labeling Standards. To minimize the possibility of error, labels should be easy 
to read and avoid nonessential material. The name of the drug, and not the name 
of the manufacturer, should be the most prominent feature and should be in at least 
12 - point type. The use of color is very controversial; some believe that all colors 
should be prohibited to force personnel to read the labels. 
In the 1990s, a Washington State legislator proposed that every drug product 
entering the state must have a color - coded label. There was concern on the part of 
many that the state legislature would turn this idea into law. The prospect of having 
to color - code all the drugs entering a single state galvanized a response by industry, 
regulators, practitioners, and safe experts who agreed to revise pharmaceutical 
labeling. A Committee to Reduce Medication Errors was formed to study the 
problem. The effort eventually satisfi ed the color coders and the proposed legislation 
was dropped. 
The committee made several recommendations for standardizing and simplifying 
labels: 
1. Eliminate unnecessary words from the label, such as “ sterile, ” “ nonpyrogenic, ” 
and “ may be habit forming. ” 
2. Allow some abbreviations such as “ HCl ” and “ Inj. ” 
3. Make label information consistent. 

4. Require that vials containing medication that must be diluted bear the words 
“ Concentrated, must be diluted ” in a box on the label, that the vial have a 
black fl ip - top with those words on it, and that the ampules carry a black 
band. 
3. Packaging Standards. While there is no evidence that trademark colors and 
logos on boxes pose a problem, the use of color on bottle tops and labels creates 
many diffi culties. There are dozens of drugs whose names are quite different but 
whose packages look alike. This creates the potential for error when people “ see ” 
what they expect to see on the label. 
Standards need to be set for color on both caps and labels. Some believe that 
prohibiting all color would be safest — in effect, taking away a cue that could divert 
someone from reading the label. 
3.2.5 REGULATORY ASPECTS 
3.2.5.1 General Considerations 
Once the fi nished dosage form is made, the product should be packed into the 
primary container and labeled. Additional packaging and labeling are also included. 
Because of the many products and labeling materials, personnel in this area must 
be alert to prevent mix - ups. Controls and in - process checks should be carried out 
throughout the packaging/labeling operation to ensure proper labeling. 
Some examples of good manufacturing practices (GMP) requirements specifi c 
to packaging and labeling in different countries are as follows: 
In the United States the requirements should be written procedures designed to 
assure that correct labels, labeling, and packaging materials are used for drug 
products; such written procedures should be followed. These procedures 
should incorporate features such as prevention of mix - ups and cross - 
contamination by physical or spatial separation from operations on other drug 
products. 
In Canada, packaging operations are performed according to comprehensive and 
detailed written operating procedures or specifi cations, which include identi- 
fi cation of equipment and packaging lines used to package the drug, adequate 
separation, and, if necessary, the dedication of packaging lines packaging different 
drugs and disposal procedures for unused printed packaging materials. 
Packaging orders are individually numbered. 
In the European Union, the requirements should be formally authorized in the 
“ packaging instructions ” for each product containing pack size and type. They 
are normally included in process controls with instructions for sampling and 
acceptance limits [42] . 
3.2.5.2 Food, Drug and Cosmetic Act 
About 100 years after its foundation, the Congress of the United States recognized 
that subjects related to safety and public health could not exclusively be state 
dependent and measures should be taken to protect the population in vital areas. 
Therefore, the federal government became interested in regulating products for 
consumption. 
REGULATORY ASPECTS 185

186 PACKAGING AND LABELING 
In 1906, the Congress approved the Wiley Law to avoid the production, sale, or 
transport of food, medications, and alcoholic beverages that were inadequate or 
falsifi ed, poisonous, or harmful. It was the fi rst food and medication regulation 
adopted in interstate commerce. The Congress was given power to regulate commerce 
between foreign nations and several U.S. states. 
In 1912 a civil code law was enacted prohibiting any false affi rmation of curing 
or therapeutic effect on medication labels. The current law was enacted on June 27, 
1938, and regulates food, medications, medical devices, and diagnostic and cosmetic 
products. The law of 1938 stopped regulating the trade of alcoholic beverages. This 
law stated, among other recommendations, the following: 
1. The label of each medication had to give the name of each active component 
and the quantity of some specifi c substances, active or not. 
2. Cosmetics had to be inoffensive and be properly labeled and packaged. 
The 1938 law states that the label of a medication should contain adequate information 
regarding its use. However, in practice, it became evident that some pharmaceuticals 
and medications had to be administered by or under the orientation of 
a medical practitioner, due to the inability of a layman to diagnose a disease, choose 
an effective treatment, and recognize the cure or the symptoms. Several products 
were thus classifi ed, but “ the prescription concept of a medication ” was introduced 
only after Alteration in the Law of Durham - Humphrey ’ s in 1951. Since then, a label 
had to carry the warring “ Caution, the Federal Law prohibits dispensation without 
medical prescription. ” The use of these medications had to be restricted to prescription 
by a practitioner and the packing or printed material inside had to contain 
adequate information so that the practitioner could prescribe them safely. 
Alterations in 1962 of the 1938 Law constituted an attempt to establish rigid 
controls on the research, production, divulging, promotion, sale, and use of medications 
as well as to assure its quality, effi ciency, and effectiveness [43] . 
3.2.5.3 New Drugs 
Before starting clinical trials in humans, an authorization should be obtained from 
the FDA. This is known as a clinical trial authorization request for a new medication 
(AEM), on which it is necessary to establish the following: 
1. The name that best describes the medication, including the chemical name 
and the structure of any new molecule 
2. A complete list of medication components. 
3. A quantitative composition of the medication. 
4. The name and address of the vendor and an acquired description of the new 
drug 
5. The methods, facilities, and controls used for the production, processing, and 
packing of a new medication 
6. All available results available from preclinical and clinical trials 
7. Copies of medication labels and the informative material that will be supplied 
to the researchers 

8. A description of the scientifi c training and the appropriate experience considered 
by the proponent to qualify a researcher as an adequate expert to 
investigate the medication 
9. The names and “ curricula vitae ” of all researchers 
10. An investigation layout planned for test accomplishment in humans 
Solicitations for release of new medications are generally very extensive, 
sometimes thousands of pages. The information has to be enough to justify the 
affi rmations contained in the label of the proposed medication with respect to 
effectiveness, dosage, and safety. The exact composition of the content on the 
medication label is usually decided by consensus between the proponent and the 
FDA. 
The requisites for solicitation of new medications, whether by prescription or not, 
are identical. The instructions contained in the medication labels for use without 
prescription should demonstrate that the medication can be used safely without 
medical supervision. 
Once the medications are perfected, the publicity related to them has to be routinely 
presented to the FDA. 
The rules of 1985 also changed the requisites regarding addendums that are 
necessary when alterations are proposed in the medication or in its labeling, for 
example. 
In regulations promulgated by the FDA on February 12, 1972, a clinic should be 
called upon regarding the effectiveness of a medication. After that the information 
may be included in the label or in the drug informative leafl et with eligible sentence 
and defi ned by dark lines that contour it [43] . 
Other dispositions contained in the alterations to the 1962 law are as follows: 
1. Immediate registration with the FDA before starting the production, repacking, 
or relabeling of medications and later annual registration, with inspections 
to be made at least once every two years. 
2. Supportive inspections in the factory, particularly where prescription medications 
are produced. 
3. The procedures used by the manufacturers should be in conformity with the 
good manufacturing practices, which permits the government to better inspect 
of all the operations. 
4. The common name should be presented on the label. 
5. The publicity of a prescription medication should present a brief summary 
mentioning the secondary effects, the contraindications, and the medication 
effectiveness. 
6. All antibiotics are subject for certifi cation procedures. 
3.2.5.4 Labeling Requisites 
According to a 1962 law, the main requisites for labeling are as described below. 
The labeling of over - the - counter medications is regulated by the Food, Drug and 
Cosmetic Act, which states: 
REGULATORY ASPECTS 187

188 PACKAGING AND LABELING 
A medication should be considered falsifi ed unless the label contains: 1. Indications of 
adequate use and 2. Adequate warnings regarding the pathological indications in those 
it should not be used or not for children use, when its use can be dangerous for health, 
of dosages, methods or interval of administration, or unsafe application, of mode and 
in necessary form for patients ’ protection. 
“ Indications of use ” were defi ned in the regulations as information with which even 
a layman can use the medication safely and for the purpose to which it is 
designated. 
The label of an over - the - counter medication must refer to the active substances, 
but it is not necessary to indicate its relative quantity, except where the ingredient 
leads to habituation. In this case the warning “ Can lead to habituation ” should 
appear on the label. 
A drug can be considered falsifi ed if it does not provide, besides indications of 
adequate use, warnings against its use in some pathological conditions (or for children) 
in which the medication can constitute a health risk. Regulations have suggested 
warnings that can be used for most well - known dangerous substances. 
3.2.5.5 Prescription Drugs 
Specifi c requisites for labeling of ethical medications or of prescription medications 
are also found in the Food, Drug and Cosmetic Act. These need not to contain 
“ adequate indications of use ” ; however, they must contain indications for the 
practitioner, inside or outside the package in which the medication is going to be 
dispensed, with adequate information for its use. This information may in - 
clude indications, effects, dosages, route of administration, methods, frequency 
and duration of administration, important dangers, contraindications, secondary 
effects, and cautions “ according to which the practitioners can prescribe the medication 
assuredly and for the desirable effects, including those for which it is 
proclaimed. ” 
Regarding all medications, the act requires that the label present a precise affi rmation 
on the weight of the content, measure or counting, as well as the name and 
manufacturer ’ s address, packer or distributor. 
The label of a prescription medication destined for oral administration has to 
contain the quantity or proportion of each active substance. 
If the medication is for parenteral administration, the quantity or proportion of 
all the excipients have also to be mentioned on the label, except for those that are 
added to adjust pH or make it isotonic, in which case only the name and its effect 
are needed. However, if the vehicle for injection is water, this does not need to be 
mentioned. 
If the medication is not to be administered by any of the routes mentioned above, 
for example, a pomade or a suppository, all excipients must to be mentioned, except 
for perfuming agents. Perfumes can be designated as such without the need to 
mention the specifi c components. 
Coloring agents can be assigned without being specifi ed individually, unless this 
is required in a separate section for regulation of coloring agents, and inoffensive 
substances added exclusively for individual identifi cation of each product need not 
be mentioned. 

The only warning that is necessary, “ Attention: the Law prohibits the dispensing 
without prescription, ” should be on the label of a prescription medication or in its 
secondary packing if the label is too little to contain it. 
3.2.5.6 Drug Information Leafl et 
The inclusion of a drug information leafl et is not compulsory whatever the medication. 
However, all medications, whether prescription or of over the counter, have to 
contain a label with adequate indications for use. If the medication label does not have 
enough space to contain all the information, the drug information leafl et has to be 
included with necessary information. The drug information leafl et and labels containing 
indication information must include the date when the text was last revised. 
To satisfy the act, the drug information leafl et usually included in the prescription 
medication packaging should contain “ adequate information on usage, including 
indications, effects, dosages, methods, route, frequency and duration of administration. 
Any important dangers, contraindications, secondary effects and cautions, 
based on which the practitioner can prescribe the medication safely and for desirable 
effects, including those for which a clam is made. ” To present the information 
in a uniform manner, the FDA issued labeling policies describing its format and the 
order and headings for the drug information leafl et description, action, indication, 
contraindications, alerts, cautions, adverse reactions, dosage and administration, 
overdose (when applicable), and as it is supplied. 
The drug information leafl et can contain the following optional information: 
Animal pharmacology and toxicology 
Clinical studies 
References 
Other specifi c cautions on medication have to appear in a visible manner at the 
beginning of the drug information leafl et so that practitioners, pharmacists, and 
patients can easily see them. 
According to GMP, an inspector should be cautious with several aspects of drug 
production, including the following: 
1. Product containers and other components have to be tested and be considered 
adequate for their intended use only if they are not reactive, departure byproducts, 
or even have absorption capacity; so that they do not affect the safety, 
identity, potency, quality, or purity of the medication or its components. 
2. Packing and labeling operations should be adequately controlled to (1) guarantee 
that only those medications that own quality standards and attain established 
specifi cations in their production and control be distributed, (2) avoid 
mix - ups during the fi lling operations, packing, and labeling, (3) assure that the 
labels and labeling used are correct for the medication, and (4) identify the 
fi nished product with a batch or a control number that allows determination 
of the batch production and control history. 
Application of the federal law on food, drug, and cosmetics is the FDA ’ s responsibility, 
which is a subdivision of the Department of Health and Human Services. 
REGULATORY ASPECTS 189

190 PACKAGING AND LABELING 
The institution is managed by a Commissaries and is subdivided into several departments: 
Food safety and applied nutrition (CFSAN), Drug evaluation and research 
(CDER), Biologics evaluation and research (CBER), Devices and radiological 
health (CDRH), Veterinary medicine (CVM), Toxicological research (NCTR), 
Regulatory affairs (ORA) and the offi ce of the commissioner (OC) [4, 44] . 
3.2.5.7 Other Regulatory Federal Laws 
There are other federal laws with which a pharmacist should be familiar. Perhaps 
the most important are laws on packing and labeling, operations that are regulated 
by the FDA and the Federal Communications Commission (FCC). The law on 
packing and labeling is targeted mostly to protecting the consumer. In the case of 
liquid the ingredients should be on the visible part of the package. The law presents 
specifi c requisites concerning the location and size of the type. Violation of this law 
can lead to apprehension by the FDA or a withdrawal order from the FCC. 
Many times a pharmacist involved in developing a product is called upon in the 
publicity of the medication. For this, he or she must understand the politics of the 
regulatory agency involved. The FCC, according to the Federal Law of Commerce, 
has jurisdiction over the announcement and promotion of all consumables, including 
medications and cosmetics. 
This law extends to all publicity and has to do with practices of fraudulent publicity 
and with promotion that is understood to be false and fraudulent. In general, the 
FCC controls the publicity of nonprescription drugs and cosmetics with respect to 
false or fraudulent affi rmations, and the FDA is responsible for labeling of medications 
and for all publicity related to prescription medications. The principal objective 
of this is to avoid unnecessary duplication of procedures while enforcing the law. 
The agencies work closely together and the FCC relies strongly on the FDA due to 
its scientifi c knowledge. Any government has the right to approve laws for its citizens 
’ protection. This right constitutes the base on which laws regulate the drug 
substance, the drug product, and its production, distribution, and sale. It is common 
that these laws exist at a district level, state level, and national level and deal with 
falsifi cation and adulteration, fraudulent publicity, and maintenance of appropriated 
sanitary conditions. 
Most U.S. states specify the purity requisites, labeling, and applicable packaging 
of a medication that are generally defi ned in identical language in federal law. 
Almost all states, prohibit the commercialization of a new medication until an 
authorization request for commercialization of a new medication has been submitted 
to the FDA and has been approved. Medication labeling requisites in each 
state are established, just as the local laws are defi ned, taking into consideration 
arguments and information, such as name and place of activity (production), content 
quantity, drug name, name of ingredients, quantity or proportion of some ingredients, 
usage indications, warning regarding dependence, caution against deterioration 
(degradation), warning about situations in which the use can be dangerous, and 
special requisites for labeling of offi cial drugs [43] . 
3.2.5.8 Fair Packaging and Labeling Act [44] 
The FDA through Fair Packaging and Labeling Act regulates the labels on many 
consumer products, including health products. Title 15: Commerce and Trade 
Chapter 39: Fair Packaging and Labeling Program [44] 

Section 1451. Congressional Delegation of Policy Informed consumers are essential 
to the fair and effi cient functioning of a free market economy. Packages and 
their labels should enable consumers to obtain accurate information as to the quantity 
of the contents and should facilitate value comparisons. Therefore, it is hereby 
declared to be the policy of the Congress to assist consumers and manufacturers in 
reaching these goals in the marketing of consumer goods [44] . 
Section 1452. Unfair and Deceptive Packaging and Labeling: Scope of 
Prohibition 
(a) Nonconforming Labels It shall be unlawful for any person engaged in the 
packaging or labeling of any consumer commodity (as defi ned in this chapter) for 
distribution in commerce, or for any person (other than a common carrier for hire, 
a contract carrier for hire, or a freight forwarder for hire) engaged in the distribution 
in commerce of any packaged or labeled consumer commodity, to distribute or 
to cause to be distributed in commerce any such commodity if such commodity is 
contained in a package, or if there is affi xed to that commodity a label, which does 
not conform to the provisions of this chapter and of regulations promulgated under 
the authority of this chapter. 
(b) Exemptions The prohibition contained in subsection (a) of this section shall 
not apply to persons engaged in business as wholesale or retail distributors of consumer 
commodities except to the extent that such persons (1) are engaged in the 
packaging or labeling of such commodities, or (2) prescribe or specify by any means 
the manner in which such commodities are packaged or labeled. 
Section 1453. Requirements of Labeling; Placement, Form, and Contents of 
Statement of Quantity; Supplemental Statement of Quantity 
(a) Contents of Label No person subject to the prohibition contained in section 
1452 of this title shall distribute or cause to be distributed in commerce any packaged 
consumer commodity unless in conformity with regulations which shall be 
established by the promulgating authority pursuant to section 1455 of this title 
which shall provide that: 
• (1) The commodity shall bear a label specifying the identity of the commodity 
and the name and place of business of the manufacturer, packer, or 
distributor; 
• (2) The net quantity of contents (in terms of weight or mass, measure, or 
numerical count) shall be separately and accurately stated in a uniform location 
upon the principal display panel of that label, using the most appropriate units 
of both the customary inch/pound system of measure, as provided in paragraph 
(3) of this subsection, and, except as provided in paragraph (3)(A)(ii) or paragraph 
(6) of this subsection, the SI metric system; 
• (3) The separate label statement of net quantity of contents appearing upon or 
affi xed to any package: 
• (A) 
• (i) if on a package labeled in terms of weight, shall be expressed in pounds, 
with any remainder in terms of ounces or common or decimal fractions of 
REGULATORY ASPECTS 191

192 PACKAGING AND LABELING 
the pound; or in the case of liquid measure, in the largest whole unit (quart, 
quarts and pint, or pints, as appropriate) with any remainder in terms of 
fl uid ounces or common or decimal fractions of the pint or quart; 
• (ii) if on a random package, may be expressed in terms of pounds and 
decimal fractions of the pound carried out to not more than three decimal 
places and is not required to, but may, include a statement in terms of the 
SI metric system carried out to not more than three decimal places; 
• (iii) if on a package labeled in terms of linear measure, shall be expressed 
in terms of the largest whole unit (yards, yards and feet, or feet, as appropriate) 
with any remainder in terms of inches or common or decimal fractions 
of the foot or yard; 
• (iv) if on a package labeled in terms of measure of area, shall be expressed 
in terms of the largest whole square unit (square yards, square yards and 
square feet, or square feet, as appropriate) with any remainder in terms of 
square inches or common or decimal fractions of the square foot or square 
yard; 
• (B) shall appear in conspicuous and easily legible type in distinct contrast (by 
topography, layout, color, embossing, or molding) with other matter on the 
package; 
• (C) shall contain letters or numerals in a type size which shall be 
• (i) established in relationship to the area of the principal display panel of 
the package, and 
• (ii) uniform for all packages of substantially the same size; and 
• (D) shall be so placed that the lines of printed matter included in that statement 
are generally parallel to the base on which the package rests as it is 
designed to be displayed; and 
• (4) The label of any package of a consumer commodity which bears a representation 
as to the number of servings of such commodity contained in such 
package shall bear a statement of the net quantity (in terms of weight or mass, 
measure, or numerical count) of each such serving. 
• (5) For purposes of paragraph (3)(A)(ii) of this subsection the term “ random 
package ” means a package which is one of a lot, shipment, or delivery of packages 
of the same consumer commodity with varying weights or masses, that is, 
packages with no fi xed weight or mass pattern. 
• (6) The requirement of paragraph (2) that the statement of net quantity of 
contents include a statement in terms of the SI metric system shall not apply 
to foods that are packaged at the retail store level. 
(b) Supplemental Statements No person subject to the prohibition contained in 
section 1452 of this title shall distribute or cause to be distributed in commerce any 
packaged consumer commodity if any qualifying words or phrases appear in conjunction 
with the separate statement of the net quantity of contents required by 
subsection (a) of this section, but nothing in this subsection or in paragraph (2) of 
subsection (a) of this section shall prohibit supplemental statements, at other places 
on the package, describing in nondeceptive terms the net quantity of contents: Provided 
, That such supplemental statements of net quantity of contents shall not 

include any term qualifying a unit of weight or mass, measure, or count that tends 
to exaggerate the amount of the commodity contained in the package. 
Section 1454. Rules and Regulations 
(a) Promulgating Authority The authority to promulgate regulations under this 
chapter is vested in (A) the Secretary of Health and Human Services (referred to 
hereinafter as the “ Secretary ” ) with respect to any consumer commodity which is 
a food, drug, device, or cosmetic, as each such term is defi ned by section 321 of title 
21; and (B) the Federal Trade Commission (referred to hereinafter as the “ Commission 
” ) with respect to any other consumer commodity. 
(b) Exemption of Commodities from Regulations If the promulgating authority 
specifi ed in this section fi nds that, because of the nature, form, or quantity of a particular 
consumer commodity, or for other good and suffi cient reasons, full compliance 
with all the requirements otherwise applicable under section 1453 of this title 
is impracticable or is not necessary for the adequate protection of consumers, the 
Secretary or the Commission (whichever the case may be) shall promulgate regulations 
exempting such commodity from those requirements to the extent and under 
such conditions as the promulgating authority determines to be consistent with 
section 1451 of this title: 
(c) Scope of Additional Regulations Whenever the promulgating authority determines 
that regulations containing prohibitions or requirements other than those 
prescribed by section 1453 of this title are necessary to prevent the deception of 
consumers or to facilitate value comparisons as to any consumer commodity, such 
authority shall promulgate with respect to that commodity regulations effective 
to: 
• (1) establish and defi ne standards for characterization of the size of a package 
enclosing any consumer commodity, which may be used to supplement the label 
statement of net quantity of contents of packages containing such commodity, 
but this paragraph shall not be construed as authorizing any limitation on the 
size, shape, weight or mass, dimensions, or number of packages which may be 
used to enclose any commodity; 
• (2) regulate the placement upon any package containing any commodity, or 
upon any label affi xed to such commodity, of any printed matter stating or 
representing by implication that such commodity is offered for retail sale at a 
price lower than the ordinary and customary retail sale price or that a retail 
sale price advantage is accorded to purchasers thereof by reason of the size of 
that package or the quantity of its contents; 
• (3) require that the label on each package of a consumer commodity (other 
than one which is a food within the meaning of section 321(f) of title 21) bear 
(A) the common or usual name of such consumer commodity, if any, and 
(B) in case such consumer commodity consists of two or more ingredients, the 
common or usual name of each such ingredient listed in order of decreasing 
predominance, but nothing in this paragraph shall be deemed to require that 
any trade secret be divulged; or 
REGULATORY ASPECTS 193

194 PACKAGING AND LABELING 
• (4) prevent the nonfunctional - slack - fi ll of packages containing consumer commodities. 
For purposes of paragraph (4) of this subsection, a package shall be 
deemed to be nonfunctionally slack - fi lled if it is fi lled to substantially less than 
its capacity for reasons other than (A) protection of the contents of such 
package or (B) the requirements of machines used for enclosing the contents 
in such package. 
(d) Development by Manufacturers, Packers, and Distributors of Voluntary Product 
Standards Whenever the Secretary of Commerce determines that there is undue 
proliferation of the weights or masses, measures, or quantities in which any consumer 
commodity or reasonably comparable consumer commodities are being distributed 
in packages for sale at retail and such undue proliferation impairs the 
reasonable ability of consumers to make value comparisons with respect to such 
consumer commodity or commodities, he shall request manufacturers, packers, and 
distributors of the commodity or commodities to participate in the development of 
a voluntary product standard for such commodity or commodities under the procedures 
for the development of voluntary products standards established by the 
Secretary pursuant to section 272 of this title. Such procedures shall provide adequate 
manufacturer, packer, distributor, and consumer representation. 
(e) Report and Recommendations to Congress upon Industry Failure to Develop or 
Abide by Voluntary Product Standards If (1) after one year after the date on which 
the Secretary of Commerce fi rst makes the request of manufacturers, packers, and 
distributors to participate in the development of a voluntary product standard as 
provided in subsection (d) of this section, he determines that such a standard will 
not be published pursuant to the provisions of such subsection (d), or (2) if such a 
standard is published and the Secretary of Commerce determines that it has not 
been observed, he shall promptly report such determination to the Congress with 
a statement of the efforts that have been made under the voluntary standards 
program and his recommendation as to whether Congress should enact legislation 
providing regulatory authority to deal with the situation in question. 
Section 1455. Procedures for Promulgation of Regulations 
(a) Hearings by Secretary of Health and Human Services Regulations promulgated 
by the Secretary under section 1453 or 1454 of this title shall be promulgated, 
and shall be subject to judicial review, pursuant to the provisions of subsections (e), 
(f), and (g) of section 371 of title 21. Hearings authorized or required for the promulgation 
of any such regulations by the Secretary shall be conducted by the Secretary 
or by such offi cer or employees of the Department of Health and Human 
Services as he may designate for that purpose. 
(b) Judicial Review; Hearings by Federal Trade Commission Regulations promulgated 
by the Commission under section 1453 or 1454 of this title shall be promulgated, 
and shall be subject to judicial review, by proceedings taken in conformity 
with the provisions of subsections (e), (f), and (g) of section 371 of title 21 in the 
same manner, and with the same effect, as if such proceedings were taken by the 
Secretary pursuant to subsection (a) of this section. Hearings authorized or required 

for the promulgation of any such regulations by the Commission shall be conducted 
by the Commission or by such offi cer or employee of the Commission as the Commission 
may designate for that purpose. 
(c) Cooperation with Other Departments and Agencies In carrying into effect the 
provisions of this chapter, the Secretary and the Commission are authorized to 
cooperate with any department or agency of the United States, with any State, Commonwealth, 
or possession of the United States, and with any department, agency, or 
political subdivision of any such State, Commonwealth, or possession. 
(d) Returnable or Reusable Glass Containers for Beverages No regulation adopted 
under this chapter shall preclude the continued use of returnable or reusable glass 
containers for beverages in inventory or with the trade as of the effective date of 
this Act, nor shall any regulation under this chapter preclude the orderly disposal 
of packages in inventory or with the trade as of the effective date of such 
regulation. 
3.2.5.9 United States Pharmacopeia Center for the Advancement of Patient 
Safety [45] 
For nearly 33 years, the USP has been reporting programs for health care professionals 
to share experiences and observations about the quality and safe use of 
medications. This year, the USP Center for the Advancement of Patient Safety 
publishes its sixth annual report to the nation on medication errors reported to 
MEDMARX (Table 6 ). It was observed that drug product packaging/labeling is one 
of the main courses of medication errors in hospitals. 
3.2.5.10 National Agency of Sanitary Vigilance ( ANVISA , Brazil) 
ANVISA is a federal organization linked to Brazil ’ s Health Ministry, which has the 
incumbency of looking after medication quality and other health products aimed at 
patients ’ safety. Several documents regarding GMP and quality control are easily 
accessed. The agency is also responsible for establishing enforcing the rules and can 
take corrective measures and punish the offenders [46] . 
Product stability and compatibility with the conditioning material are distinct, 
separate, and complementary concepts which should be applied to the pharmaceutical 
product before being made available for health care. 
TABLE 6 Selected Causes of Error Related to Equipment, Product Packaging/Labeling, 
and Communication in ICUs 
Cause of Error N (Nonharmful + Harmful) Percent Harmful 
Label (the facility ’ s) design 1,236 6,9 
Similar packaging/labeling 
Packaging/container design 
Label (manufacturer ’ s) design 
Brand/generic names look - alike 
Source : MEDMARX Data Report: A Chartbook of 2000 – 2004 Findings from Intensive Care Units 
(ICUs) and Radiological Services. 
REGULATORY ASPECTS 195

196 PACKAGING AND LABELING 
In the compatibility test between formulation and the conditioning material, 
several options of conditioning materials are evaluated to determine the most adequate 
for the product. 
The environmental conditions and periodicity analyses can be the same as those 
mentioned for the stability studies for the formulation. In this phase, the possible 
interactions between the product and the conditioning material which is in direct 
contact with the medication are verifi ed. Phenomena such as absorption, migration, 
corrosion, and others that compromise integrity can be observed. Considering that 
these types of tests are generally destructive, it is necessary to defi ne the number of 
samples to be tested. 
In ANVISA ’ s documents, different types of tests are established that should be 
carried out with different types of available materials and employed for conditioning 
medications and cosmetics (cellulose packagings, metallic, plastic, pressurized, 
etc.) [46] . 
3.2.5.11 International Committee on Harmonization ( ICH ) 
In the document “ Good Manufacturing Practice Guide for Active Pharmaceutical 
Ingredients (APIs) ” of the ICH Harmonized Tripartite Guideline, the following 
instructions are given for packaging and identifi cation labeling of APIs and intermediates 
[47] . 
General 
• There should be written procedures describing the receipt, identifi cation, quarantine, 
sampling, examination and/or testing and release, and handling of packaging 
and labeling materials. 
• Packaging and labeling materials should conform to established specifi cations. 
Those that do not comply with such specifi cations should be rejected to prevent 
their use in operations for which they are unsuitable. 
• Records should be maintained for each shipment of labels and packaging 
materials showing receipt, examination, or testing, and whether accepted or 
reject. 
Packaging Materials 
• Containers should provide adequate protection against deterioration or contamination 
of the intermediate or API that may occur during transportation 
and recommended storage. 
• Containers should be clean and, where indicated by the nature of the intermediate 
or API, sanitized to ensure that they are suitable for their intended use. 
These containers should not be reactive, addictive, or absorptive so that the 
quality of the intermediate or API complies with the specifi cations. 
• If containers are reused, they should be cleaned in accordance with documented 
procedures and all previous labels should be removed or defaced. 

Label Issuance and Control 
• Access to the label storage areas should be limited to authorized personnel. 
• Procedures should be used to reconcile the quantities of labels issued, used, 
and returned and to evaluate discrepancies found between the number of containers 
labeled and the number of labels issued. Such discrepancies should be 
investigated and the investigation should be approved by the quality unit(s). 
• All excess labels bearing batch numbers or other batch - related printing should 
be destroyed. Returned labels should be maintained and stored in a manner 
that prevents mix - ups and provides proper identifi cation. 
• Obsolete and outdated labels should be destroyed. 
• Printing devices used to print labels for packaging operations should be controlled 
to ensure that all imprinting conforms to the print specifi ed in the batch 
production record. 
• Printed labels issued for a batch should be carefully examined for proper identity 
and conformity to specifi cations in the master production record. The 
results of this examination should be documented. 
• A printed label representative of those used should be included in the batch 
production record. 
Packaging and Labeling Operations 
• There should be documented procedures designed to ensure that correct packaging 
materials and labels are used. 
• Labeling operations should be designed to prevent mix - ups. There should be 
physical or spatial separation from operations involving other intermediates or 
APIs. 
• Labels used on containers of intermediates or APIs should indicate the name 
or identifying code, the batch number of the product, and storage conditions, 
when such information is critical to assure the quality of intermediate API. 
• If the intermediate or API is intended to be transferred outside the control of 
the manufacturer ’ s material management system, the name and address of the 
manufacturer, quantity of contents and special transport conditions, and any 
special legal requirements should also be included on the label. For intermediates 
or APIs with an expiry date, the expiry date should be indicated on the 
label and certifi cate of analysis. For intermediates or APIs with a retest date, 
the retest date should be indicated on the label and/or certifi cate of analysis. 
• Packaging and labeling facilities should be inspected immediately before use 
to ensure that all materials not needed for the next packaging operation have 
been removed. This examination should be documented in the batch production 
records, the facility log, or other documentation system. 
• Packaged and labeled intermediates or APIs should be examined to ensure that 
containers and packages in the batch have the correct label. This examination 
should be part of the packaging operation. Results of these examinations should 
be recorded in the batch production or control records. 
REGULATORY ASPECTS 197

198 PACKAGING AND LABELING 
• Intermediate or API containers that are transported outside of the manufacturer 
’ s control should be sealed in a manner such that, if the seal is breached 
or missing, the recipient will be alerted to the possibility that the contents may 
have been altered. 
3.2.5.12 European Union Regulatory Bodies 
European regulatory requirements say little to date about container closure integrity 
of parenteral or sterile pharmaceutical products. Regulations provide for 
package integrity verifi cation of parenteral vials to be supported by the performance 
of sterility tests as part of the stability program. More specifi c information is 
described in the European Union (EU) 1998 “ Rules Governing Medical Products 
in the European Union, Pharmaceutical Legislation. ” These GMP regulations 
require that the sealing or closure process be validated. Packages sealed by fusion 
(e.g., ampules) should be 100% integrity tested. Other packages should be sampled 
and checked appropriately. Packages sealed under vacuum should be checked for 
the presence of vacuum. While not as detailed as the FDA guidances, it is evident 
that the EU rules also require the verifi cation of parenteral product package seal 
integrity. It is important to note that the EU rules specifi cally require 100% product 
testing for fusion - sealed packages, sampling and testing of all other packages, and 
vacuum verifi cation for packages sealed under partial pressure [42] . 
The vacuum/pressure decay test is performed by placing the package in a tightly 
closed test chamber, a pressure or vacuum is applied inside the chamber, and then 
the rate of pressure/vacuum change in the chamber over time is monitored. The rate 
or extent of change is compared to that previously exhibited by a control, nonleaking 
package. Signifi cantly greater change for a test package is indicative of a leak. 
REFERENCES 
1. Griffi n , J. P. Ed. ( 2002 ), The Textbook of Pharmaceutical Medicine , 4th ed., BMJ Publishing 
, London . 
2. Harburn , K. ( 1990 ), Quality Control of Packaging Materials in the Pharmaceutical Industry 
, Marcel Dekker , New York . 
3. O ’ Brien , J. D. ( 1990 ), Medical Device Packaging Handbook , Marcel Dekker , New York . 
4. U.S. Food and Drug Administration (FDA) ( 1999 , May) Guidance on container closure 
systems for packaging human drugs and biologics, U.S. Department of Health and Human 
Services, FDA, Washington, DC. 
5. Yoshioka , S. ( 2000 ), Stability of Drugs and Dosage Forms , Kluwer Academic Publishers : 
New York, NY, USA , p 272 . 
6. Banker , G. S. , and Rhodes , C. T. ( 2002 ), Modern Pharmaceutics , 4th ed., rev. and expanded, 
Marcel Dekker , New York . 
7. Connor , J. , Rafter , N. , and Rodgers , A. ( 2004 ), Do fi xed - dose combination pills or unit - 
of - use packaging improve adherence ? A systematic review. Br. World Health Org. , 82 , 
935 – 939 . 
8. Bloomfi eld , S. F. ( 1990 ), Microbial contamination: Spoilage and hazard , in Denyer , S. , and 
Baird , R. , Eds., Guide to Microbiological Control in Pharmaceuticals , Ellis Horwood, 
Chichester , England , pp 29 – 52 . 

9. Aspinall , J. A. , Duffy , T. D. , Saunders , M. B. , and Taylor , C. G. ( 1980 ), The effect of low 
density polyethylene containers on some hospital - manufactured eye drop formulations. 
1. Sorption of phenyl mercuric acetate , J. Clin. Hosp. Pharm. , 5 , 21 – 29 . 
10. United States Pharmacopeia ( 2006 ), 29th ed., United States Pharmacopeial Convention, 
Rockville, MD. 
11. Parker , W. A. , and MacCara , M. E. ( 1980 ), Compatibility of diazepam with intravenous 
fl uid containers and administration sets , Am. J. Hosp. Pharm. , 37 , 496 – 500 . 
12. Mizutani , T. , Wagi , K. , and Terai , Y. ( 1981 ), Estimation of diazepam adsorbed on glass 
surfaces and silicone - coated surfaces as models of surfaces of containers , Chem. Pharm. 
Bull. , 29 , 1182 – 1183 . 
13. Yahya , A. M. , McElnay , J. C. , and D ’ Arcy , P. F. ( 1985 ), Binding of chloroquine to glass , 
Int. J. Pharm. , 25 , 217 – 223 . 
14. Vromans , H. , and Van Laarhoven , J. A. H. ( 1992 ), A study on water permeation through 
rubber closures of injection vials , Int. J. Pharm. , 79 , 301 – 308 . 
15. Matsuura , I. , and Kawamata , M. ( 1978 ), Studies on the prediction of shelf life. III. Moisture 
sorption of pharmaceutical preparation under the shelf condition , Yakugaku Zusshi , 
98 , 986 – 996 . 
16. Nakabayashi , K. , Tuchida , T. , and Mima , H. ( 1980 ), Stability of packaged solid dosage 
forms. I. Shelf - life prediction of packaged tablets liable to moisture damage , Chem. 
Pharm. Bull. , 28 , 1090 – 1098 . 
17. Nakabayashi , K. , Shimamoto , T. , and Mima , H. ( 1980 ), Stability of packaged solid dosage 
forms. II. Shelf - life prediction for packaged sugar - coated tablets liable to moisture and 
heat damage , Chem. Pharm. Bull. , 28 , 1099 – 1106 . 
18. Nakabayashi , K. , Shimamoto , T. , and Mima , H. ( 1980 ), Stability of packaged solid dosage 
forms. III. Kinetic studies by differential analysis on the deterioration of sugar - coated 
tablets under the infl uence of moisture and heat , Chem. Pharm. Bull. , 28 , 1107 – 1111 . 
19. Tonnesen , H. H. ( 1996 ), Photostability of Drugs and Drug Formulations , CRC Press , 
London . 
20. Kontny , M. J. , Koppenol , S. , and Graham , E. T. ( 1992 ), Use of the sorption – desorption 
moisture transfer model to assess the utility of a desiccant in a solid product , Int. J. Pharm. , 
84 , 261 – 271 . 
21. Pikal , M. J. , and Lang , J. E. ( 1978 ), Rubber closures as a source of haze in freeze dried 
parenterals: Test methodology for closure evaluation , J. Parenteral drug Assoc. , 32 , 
162 – 173 . 
22. Jaehnke , R. W. O. , Kreuter , J. , and Ross , G. ( 1990 ), Interaction of rubber closures with 
powders for parenteral administration , J. Parenteral sci. Tech. , 44 , 282 – 288 . 
23. Jaehnke , R. W. O. , Kreuter , J. , and Ross , G. ( 1991 ), Content/container interactions: The 
phenomenon of haze formation on reconstitution of solids for parenteral use , Int. J. 
Pharm. , 77 , 4755 . 
24. Moorhatch , P. , and Chiou , W. L. ( 1974 ), Interactions between drugs and plastic intravenous 
fl uid bags. II: Leaching of chemicals from bags containing various solvent media , 
Am. J. Hosp. Pharm. , 31 , 149 – 152 . 
25. Venkataramanan , R. , Burckart , G. J. , Ptachcinski , R. J. , Blaha , R. , Logue , L. W. , Bahnson , 
A. C. , and Brady , G. J. E. ( 1986 ), Leaching of diethylhexyl phthalate from polyvinyl 
chloride bags into intravenous cyclosporine solution , Am. J. Hosp. Pharm. , 43 , 2800 – 
2802 . 
26. Boruchoff , S. A. ( 1987 ), Hypotension and cardiac arrest in rats after infusion of mono 
(2ethylhexyl) phthalate (MEHP), a contaminant of stored blood , N. Engl. J. Med. , 316 , 
1218 – 1219 . 
REFERENCES 199

200 PACKAGING AND LABELING 
27. U.S. Food and Drug Administration , Code of Federal Regulations (CFR) — Title 21, Food 
and drugs, Chapters 174 – 186, available: http://www.access.gpo.gov/nara/cfr/index.html , 
accessed Mar. 11, 2005. 
28. Kowaluk , E. A. , Roberts , M. S. , Blackburn , H. D. , and Polack , A. E. ( 1981 ), Interactions 
between drugs and polyvinyl chloride infusion bags , Am. J. Hosp. Pharm. , 38 , 1308 – 1314 . 
29. Illum , L. , and Bundgaard , H. ( 1982 ), Sorption of drugs by plastic infusion bags , Int. J. 
Pharm. , 10 , 339 – 351 . 
30. Illum , L. , Bundgaard , H. , and Davis , S. S. ( 1983 ), A constant partition model for examining 
the sorption of drugs by plastic infusion bags , Int. J. Pharm. , 17 , 183 – 192 . 
31. Atkinson , H. C. , and Duffull , S. B. ( 1990 ), Prediction of drug loss from PVC infusion bags , 
J. Pharm. Pharmacol. , 43 , 374 – 376 . 
32. Richardson , N. E. , and Meakin , B. J. ( 1974 ), The sorption of benzocaine from aqueous 
solution by nylon 6 powder , J. Pharm. Phamacol. , 26 , 166 – 174 . 
33. Santoro , M. I. R. M. , Kedor - Hackmann , E. R. M. , and Moudatsos , K. M. ( 1993 ), Estabilidade 
de sais de reidrata c a o oral em diferentes tipos de embalagem . Bol. Sanit. Panam. , 
115 , 310 – 315 . 
34. World Health Organization (WHO) ( 2003 ), The International Pharmacopoeia, Tests and 
General Requirements for Dosage Forms: Quality Specifi cations for Pharmaceutical Substances 
and Tablets , 3rd ed., Vol. 5, WHO , Geneva. 
35. Santoro , M. I. R. M. , Oliveira , D. A. G. C. , Kedor - Hackmann , E. R. M. , and Singh , A. K. 
( 2004 ), Quantifying benzophenone - 3 and octyl methoxycinnamate in sunscreen emulsions 
, Cosm. & Toil. , 119 , 77 – 82 . 
36. Santoro , M. I. R. M. , Oliveira , D. A. G. C. , Kedor - Hackmann , E. R. , and Singh , A. K. ( 2005 ), 
The effect of packaging materials on the stability of sunscreen emulsions , Int. J. Pharm. , 
13 , 197 – 203 . 
37. Thoma , K. , and Kerker , R. ( 1992 ), Photoinstability of drugs. 6. Investigations on the photosansibility 
of molsidomine , Pharm. Ind. , 54 , 630 – 638 . 
38. British Pharmacopoeia ( 2002 ), Her Majesty ’ s Stationary Offi ce, London, pp A144, 135 – 
136, 196, 671 – 673, 778 – 780, 976 – 978, 1145 – 1146. 
39. Albert , D. E. ( 2004 ), Evaluating pharmaceutical container closure systems , Pharm. & 
Med. Packaging News , 3 , 76 – 78 . 
40. ASHP Council on Professional Affairs ( 1993 ), ASHP Guidelines on preventing medication 
errors in hospital , Am. J. Hosp. Pharm. , 50 , 305 – 314 . 
41. ASHP Council on Professional Affairs ( 2001 ), ASHP guidelines on preventing medication 
errors in hospital , Am. J. Hosp. Pharm. , 58 , 3033 – 3041 . 
42. European Pharmacopoeia ( 2001 ), 4th ed., Council of Europe, Strasbourg. 
43. Lachman , L. , Lieberman , H. A. , and Kanig , J. L. ( 2001 ), Teoria e pr a tica na ind u stria 
farmac e utica , Funda c a o Calouste Gulbenkian , Lisboa . 
44. U.S. Food and Drug Administration, Fair Packaging and Labeling Act . Title 15 — 
Commerce and Trade, Chapter 39 — Fair Packaging and Labeling Program, available: 
http://www.fda.gov/opacom/laws/fplact.htm accessed Mar. 11, 2005. 
45. Santell , J. P. , Hicks , R. W. , and Cousins , D. D. ( 2005 ), MEDMARX Data Report: A Chartbook 
of 2000 – 2004 Findings from Intensive Care Units and Radiological Services , USP 
Center for Advancement of Patient Safety , Rockville, MD . 
46. Ag e ncia Nacional de Vigil a ncia Sanit a ria (ANVISA) ( 2004 ), Guia de Estabilidade de 
Produtos Cosm e ticos , ANVISA , Bras i lia . 
47. International Organization on Harmonisation (2000), ICH harmonized tripartite guideline: 
Good manufacturing practice guide for active pharmaceutical ingredients, available: 
http://www.ICH.org , accessed June 23, 2005. 
48. Sarbach , C. , Yagoubi , N. , Sauzieres , J. , Renaux , C. , Ferrier , D. , and Postaire , E. ( 1996 ), 
Migration of impurities from a multi-layer plastics container into a parenteral infusion 
solution , Int. J. Pharm. , 140 , 169 – 174 . 

201 
3.3 
CLEAN - FACILITY DESIGN, 
CONSTRUCTION, AND 
MAINTENANCE ISSUES 
Raymond K. Schneider 
Clemson University, Clemson, South Carolina 
Contents 
3.3.1 Introduction 
3.3.2 Planning for Project Success 
3.3.2.1 Needs Assessment 
3.3.2.2 Front - End Planning 
3.3.2.3 Preliminary Design 
3.3.2.4 Procurement 
3.3.2.5 Construction 
3.3.2.6 Start - Up and Validation 
3.3.2.7 Summary 
3.3.3 Design Options 
3.3.3.1 Clean - Facility Scope 
3.3.3.2 Design Parameters 
3.3.3.3 Architectural Design Issues 
3.3.3.4 Materials of Construction 
3.3.3.5 HVAC System 
3.3.3.6 Clean - Room Testing 
3.3.3.7 Utilities 
3.3.4 Construction Phase: Clean Build Protocol 
3.3.4.1 General 
3.3.4.2 Level I Clean Construction 
3.3.4.3 Level II Clean Construction 
3.3.5 Maintenance 
Appendix A: Guidelines for Construction Personnel and Work Tools in a Clean 
Room 
Appendix B: Cleaning the Clean Room 
Bibliography 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

202 CLEAN-FACILITY DESIGN, CONSTRUCTION, AND MAINTENANCE ISSUES 
3.3.1 INTRODUCTION 
While there are discrete steps in the design and construction of a pharmaceutical 
manufacturing plant project, those projects deemed successful incorporate certain 
practices that promote fl ow of the construction process toward completion on time 
and within budget. Proper front - end planning is not completed until it results in 
appropriate values for design parameters, “ buy - in ” at all levels of management, and 
clear direction for the design phase. Engineering the clean room in accordance with 
recognized industry practice would produce construction documents that facilitate 
clear procurement and construction planning as well as a focused, effi cient, construction 
effort. A full return on the energy expended through the construction phase 
cannot be realized without a well - executed start - up and validation process that 
provides baseline data for effective ongoing operation and maintenance. 
The steps in the clean - room construction project include: 
Needs assessment 
Front - end planning 
Preliminary design 
Construction document development 
Procurement 
Construction 
Start - up and validation 
One of the truisms of the construction industry is that the greatest impact on the 
cost of a facility can be made at the earliest stages of the process. The construction 
process can be likened to a snowball rolling down a snow - covered hill. It grows and 
gains momentum, seemingly taking on a life of its own, until it can only be brought 
under control with a major effort. So too with manufacturing plant projects. Careful 
work during the fi rst three stages will ensure that the project begins on a well - 
directed course and moves to a successful conclusion. 
Sometimes the special nature of pharmaceutical manufacturing plant projects 
clouds the fact that building such a plant is in fact a construction project. The facility 
engineering team of a small to medium company may be tempted to turn away from 
such projects due to the projects ’ perceived uniqueness and leave the key decision 
making to others. In fact, it is the construction experience of that team that is most 
required to keep the project costs under control. The way to accomplish this is for 
the team to be involved in the process from its earliest stages. 
Let us review the steps in such a project and identify what should occur at each 
step and the potential for trouble. 
3.3.2 PLANNING FOR PROJECT SUCCESS 
3.3.2.1 Needs Assessment 
It is during this early stage that a requirement for a clean manufacturing facility is 
perceived. The need for the facility may be precipitated by a new product, an 

improved product, an improved manufacturing methodology, new or more stringent 
regulation requirements, or perhaps a change in marketing strategy. 
At this point a study should be undertaken to determine the benefi ts to be realized 
by the new facility as well as the costs to be incurred. Costs arise from not only 
construction but also ongoing operation and maintenance. These costs are affected 
by the plant location and the availability of a trained or trainable workforce. Does 
the day - to - day operation of the facility generally require that special attire be worn? 
Are special procedures, possibly more time consuming than those presently used, 
required? It is important that this study is complete and accurate in order to prevent 
any unrealistic expectations on the part of management and plant operations and 
to permit advanced planning for revised procedures once the facility is in use. 
The study should describe the goals of the project, its impact on present operations, 
budget restraints, tentative schedule, and path forward. It will serve as the 
basis for front - end planning and will provide the standard against which the success 
of the program is measured. 
3.3.2.2 Front - End Planning 
While the needs assessment study may be conducted by a limited number of people, 
the front - end planning process should be open to all. Plant facilities people will be 
bearing the brunt of the responsibility for bringing the facility online, on schedule, 
and within budget. Process people are responsible for ensuring that the facility will 
adequately house the process equipment and that the facility incorporates suffi cient 
space, utilities, process fl ow considerations, and provision for fl ow of people and 
material to support the goals of the building program. Human resources people 
have to staff the facility, either out of the present employee pool or from the general 
local labor market. They must know the requirements of potential employees as 
well as the conditions under which they will be working. 
Procurement people will be purchasing furnishings and process equipment for 
the plant as well as overseeing the contracts let to the design and construction professionals. 
Operations people should have input regarding design parameters such 
as temperature, humidity, lighting, vibration, cleanliness class, and energy needs. 
Materials handling people should participate in order to understand the requirements 
for storing and transporting raw materials as well as retrieving, storing, and 
shipping fi nished goods from the plant. 
An integral part of the front - end planning team should be the design professionals 
charged with developing the plant design based on client input in such a way as 
to satisfy as many requirements developed in needs assessment as possible. This 
team may be assembled internally but frequently is drawn from specialty builders, 
architectural and engineering (A & E) fi rms, and design/build fi rms active in the 
pharmaceutical industry. The team of design professionals should have pharmaceutical 
experience on facilities comparable in size and complexity to that being planned 
as well as extensive experience in construction projects of all types. The design team 
may offer design only, design/build, procurement, construction management, or 
combinations of these services. This design team should be considered a resource 
during the front - end planning phase. It is the wise client who takes advantage of 
the experience of the design team, permitting them a large role as facilitators of the 
planning sessions. 
PLANNING FOR PROJECT SUCCESS 203

204 CLEAN-FACILITY DESIGN, CONSTRUCTION, AND MAINTENANCE ISSUES 
An appropriate design team will demonstrate expertise in contamination control 
philosophies, space planning, code compliance, and mechanical and electrical design 
and will be familiar with materials of construction currently being used in pharmaceutical 
projects. It is frequently helpful to include a member of the construction 
team in the front - end planning effort to advise on constructibility of the facility 
being planned. Unrealistic construction schedules will be avoided and fi eld rework 
will be minimized if appropriate attention is paid to the construction phase early in 
the planning process. 
3.3.2.3 Preliminary Design 
Front - end planning typically utilizes the expertise of client process people to convey 
the requirements of the pharmaceutical facility to the design team. With this information 
in hand the design team begins the facility design incorporating process 
needs, code requirements, safety issues, material and personnel fl ow, work - in - process 
storage, utility needs, and so on, into a fi rst - cut approach. 
Client representatives have an opportunity to review the effort and begin fi ne 
tuning the design to incorporate late - breaking process changes. The preliminary 
design is a target that helps both the design team and the client solidify design goals. 
Change is inexpensive, and therefore encouraged, at this stage and buy - in by all 
concerned is a major objective of this phase of the design effort. 
A budget based on the agreed - upon preliminary design should be developed to 
make sure that the overall project is on course. This will minimize surprises further 
along in the design/build process. Ideally the design will be “ cast in stone ” at the 
end of the preliminary phase. This permits the production work on the design documents 
to proceed unhindered. The more unknowns left at the end of the preliminary 
phase, the more diffi cult it will be to complete design documents in a timely 
fashion. 
Construction Document Development The construction documents should convey 
the intent of the design team and client to the construction team. A good set of 
construction documents should result in a tight spread of construction bids as there 
should be little room for varying interpretation on the part of the potential construction 
contractors. The drawings should have suffi cient notes to convey the design 
intent without creating a cluttered appearance. The written specifi cations should be 
as brief as possible consistent with clarity. 
Complicated documents create the impression that a project may be more 
involved, and therefore more costly, than it should be. Cautious contractors may 
unnecessarily infl ate their bid to cover perceived contingencies. Specifi cations that 
are too wordy may be diffi cult to follow and similarly result in higher prices as 
bidders make sure all bases are covered. No one likes surprises. 
The development of construction documents should be a straightforward process 
with little involvement by the client except to monitor the process and ensure that 
the original design intent is followed. While changes will always occur during this 
phase ( “ cast in stone ” is a euphemism for “ let ’ s keep the changes under control ” ), 
they are certainly less costly at this point than during the construction phase. It is 
desirable to minimize such changes. A continuous sequence of changes suggests that 
the preliminary design phase was not entered into seriously. It demonstrates a lack 

of preparedness on the part of the client and a lack of ability to communicate and 
draw out the client ’ s needs on the part of the design team. A sense of clarity of 
purpose slips away with ongoing change and the possibility for errors in construction 
documents, which eventually surface as costly construction changes, increases. 
3.3.2.4 Procurement 
A detailed scope of work describing the materials and services required is a vital 
part of the procurement process. There is no purpose to keeping the project bidders 
in the dark regarding what is required of them. The role of the procurement function 
is to obtain maximum value, that is, the best quality and schedule at the lowest 
price. The clearer the scope of work and construction documents, the better will be 
the chance of this happening. A low price is not a good value if the schedule slips 
by several months as a result. A marginal plant that does not maintain design conditions 
or meet production goals is a poor value even if it was delivered within 
schedule. 
The procurement process should qualify potential bidders by ensuring that similar 
pharmaceutical projects have been delivered on time, within budget, and on schedule. 
References should be checked. It is expected that references offered by a 
potential bidder would have good things to say about that bidder, but this is not a 
certainty and pointed questioning about personnel, schedule, quality, change orders, 
follow - up, and so on, can help develop a warm feeling or an uncertain feeling about 
potential bidders. If bids are in fact quite close, it is the quality of references that 
might suggest a particular bidder be given preference. 
There are a number of ways in which the project can be procured. Use of in - house 
engineering and construction expertise may work in special situations or on smaller 
projects. Typically problems arise when facilities departments, stretched to their 
limit with ongoing plant requirements, must lower the priority of the new facility to 
meet other commitments. Schedules may stretch out unacceptably. 
A number of specialty contractors have proven over the years to be adept at 
installing small turnkey facilities of limited complexity in a timely and economical 
fashion. If extensive engineering is required, if local code compliance becomes an 
issue, if complex process requirements must be met, or if the client requirements 
exceed the experience of the supplier there could be cause for concern. 
Design/build is a popular approach in that it suggests a single source of responsibility 
for all phases of the project. Frequently fi rms billing themselves as “ design/ 
build ” are strong in either design or build, but not both. The strong design fi rm can 
put the essentials on paper but the fi nal price and schedule may suffer. The strong 
construction fi rm may lack the expertise to create an appropriate manufacturing 
environment, particularly where clean - room expertise is required. The project may 
be outstanding in all respects except performance. A good review of references is 
essential before selecting a design/build fi rm. 
Construction management has been increasingly used on larger projects. A good 
construction management fi rm will work closely with the client - selected design 
company to review constructability and adequacy of construction documents. It will 
assist to qualify bidders, maintain schedule, track costs, administer and oversee, and 
generally ensure that a team incorporating the strongest skills is assembled to complete 
the project. Pharmaceutical experience is essential. 
PLANNING FOR PROJECT SUCCESS 205

206 CLEAN-FACILITY DESIGN, CONSTRUCTION, AND MAINTENANCE ISSUES 
3.3.2.5 Construction 
The construction process should proceed smoothly if the remarks presented above 
are followed. Cost can increase during this phase if changes must be implemented. 
While change is inevitable, a construction change procedure negotiated during the 
bidding phase and in place during construction will keep such change from getting 
out of control. 
The requirement for “ building clean ” has arisen in recent years as more stringent 
clean rooms have become more popular. Imposing a clean construction protocol on 
contractors can lengthen the schedule and increase cost. The protocol should be 
developed during the construction document phase and be an integral part of the 
bid documents. Once the decision is made to work clean, protocols developed 
should be followed by everyone on the jobsite associated with the clean areas. A 
poorly conceived and enforced protocol will be a costly and futile exercise. The 
tendency to build clean on every new or retrofi t project should be carefully evaluated 
and a practical protocol should be developed consistent with the needs of the 
project. 
Client end users should be encouraged to observe construction as it progresses. 
They will be more intelligent about how their facility was built and therefore more 
attuned to maintaining the facility once it is completed and in operation. While 
suggestions should be welcomed as construction progresses, it is important that a 
chain of command be enforced. Any questions or suggestions or concerns should 
not be expressed to workers on the site but rather through project management 
channels. In this way good ideas can be implemented and bad ideas shelved without 
impacting the construction effort in a negative manner. Note the one exception to 
this practice is in regard to safety. Everyone on the site has safety responsibility. 
Any unsafe acts should be questioned and supervisors consulted immediately. 
3.3.2.6 Start - Up and Validation 
Subcontractors on the jobsite should be responsible for start - up as well as installation 
of equipment. Equipment manufacturers typically have personnel available to 
ensure appropriate start - up procedures are followed. If several trades are involved 
in the installation of a particular piece of equipment, then one trade should be 
assigned, by contract, as having coordinating responsibility for that piece of equipment. 
This will minimize “ fi nger pointing ” when equipment does not start or operate 
properly. This can be a sensitive issue and a construction manager can set the tone 
for cooperation in this area. 
An independent contractor responsible to the construction manager or owner 
should do testing and balancing (TAB) of mechanical systems. All start - up should 
be complete and initial valve or damper settings made (and recorded) by the subcontractor 
before testing and balancing begins. The TAB contractor should not have 
to repair equipment or troubleshoot inoperative equipment but rather only adjust 
and verify performance of equipment. 
A separate contractor should certify clean - room areas. This might be the TAB 
contractor if that fi rm is suitably qualifi ed. There should be no question of equipment 
being operative at this stage of the project since start - up and testing and 
balancing are complete. Certifi cation is the verifi cation of facility compliance 

DESIGN OPTIONS 207 
with clean - room specifi cations. If the facility design is well conceived and the construction 
team has installed a quality project, any certifi cation test failure will most 
likely be corrected through fairly minor adjustments. Failure of the clean room to 
pass certifi cation tests might require redesign but more frequently requires some 
equipment adjustment or perhaps a fi lter repair and then a retest. It is important 
that a clear understanding of responsibility be communicated before problems are 
encountered. Failure to plan for potential problems could result in extending the 
schedule and incurring unforeseen costs at a crucial point in the project. 
3.3.2.7 Summary 
Recognizing the step - by - step process involved in even the smallest pharmaceutical 
project can help focus attention in a manner that will result in a successful project. 
The formal schedule of a well - conceived project will include needs assessment, 
front - end planning, and preliminary design. It is important that project progress is 
measured against such a schedule and not just by the visual impact caused by bricks 
and mortar being installed. 
3.3.3 DESIGN OPTIONS 
3.3.3.1 Clean - Facility Scope 
The purpose of this section is to identify design and construction options for those 
parts of a pharmaceutical facility intended to house process equipment. These suggestions 
are intended to assure that the facilities, when used as designed, will meet 
the requirements of current good manufacturing practices (cGMPs). Air cleanliness 
within the facility may range from International Organization for Standardization 
(ISO) 5 (Class 100) through ISO 8 (Class 100,000). In addition, areas may be considered 
clean or labeled as “ controlled environment ” without having a cleanliness 
class assigned to the space. Note that throughout this chapter cleanliness class will 
be described using the designation presented in the new ISO 14644 (e.g., ISO 5, ISO 
8) and parenthetically as presented in the currently obsolete (but widely understood 
and quoted) U.S. Federal Standard 209 (e.g., Class 100, Class 100,000). 
A cleanliness classifi cation in accordance with the latest revision of ISO 14644 is 
generally inadequate by itself to describe a facility used for pharmaceutical processes. 
The presence of viable particles (living organisms) within the particle count 
achieved by applying methods described in the standard may affect the product 
within the facility. A measure of both viable and nonviable particles is required to 
provide suffi cient information upon which to base a decision regarding the suitability 
of the clean room for its intended purpose. 
The options presented herein are intended to provide facilities that will effectively 
restrict both viable and nonviable particles from entering the clean areas, 
minimize contamination introduced by the facility itself, and continuously remove 
contaminants generated during normal operations. 
Measurement of total particle count in the clean room is described in ISO 14644. 
This count may be composed of viable, nonviable, or nonviable host particles with 
a viable traveler. There is no generally accepted relationship between total particle 

208 CLEAN-FACILITY DESIGN, CONSTRUCTION, AND MAINTENANCE ISSUES 
count and viable particle count. While maintaining appropriate particle counts is 
important in clean - room design and operation, a protocol designed to identify viable 
particles should be inherent in the certifi cation/validation testing of a pharmaceutical 
clean room. 
No facility design can compensate for excessive contamination generated within 
it. In addition to effective facility design, the user must also institute a routine maintenance 
program as well as maintain personnel and operational disciplines that limit 
particles both entering and being generated within the facility. 
While this section identifi es options for contamination control in facility design, 
any such options must be implemented in accordance with all appropriate government 
and regulatory building and safety codes. The design guideline is nonspecifi c 
as regards biological or chemical materials that may be used within the facility but 
generally addresses bulk pharmaceutical chemical plants (BPCs), secondary manufacturing 
chemical plants, bulk biopharmaceutical plants, and plants used for fi ll and 
fi nish operations. Good practice as well as any regulations governing biological and 
pharmaceutical processes conducted within the facility must be adhered to as 
required and could modify some of the suggestions contained herein. 
3.3.3.2 Design Parameters 
The design of the facility is based upon specifi cation of certain design parameters. 
These in turn are used to calculate building system equipment capacities and aid in 
the selection of the appropriate types of equipment that are required. Design 
parameters that may be critical are discussed below. 
Cleanliness Classifi cation The classifi cation of the clean areas is determined by 
the using organization consistent with the level of nonviable and viable particulate 
contamination acceptable to the process conducted within the facility. This may be 
governed by regulatory agencies, client organizations, or company protocols. Target 
goals are set for nonviable particle count in accordance with the ISO. Viable particle 
target goals should be stated in colony - forming units (CFU) per square centimeter. 
In accordance with ISO 14644, particle goals will typically be identifi ed for “ at rest ” 
and “ operational ” modes. 
In the absence of other guidance governing the cleanliness classifi cation and 
acceptable levels of microbial contamination of the clean room, the values presented 
in Table 1 may be used. The room grades presented are from most critical 
(A) to least critical (E). The defi nition of criticality is left to the clean - room user 
organization. 
Other Design Parameters Facility design parameters that support the process 
within the clean room should be established by the user organization. Parameters 
such as temperature, humidity, lighting requirements, sound level, and/or vibration 
may be process driven or comfort driven and therefore are selected to accommodate 
specifi c process or comfort requirements as determined by the end user. 
Local Control Under some circumstances, cleanliness requirements can be 
achieved through the use of localized controls such as clean tents, glove boxes, 
minienvironments, or isolators. These provide unidirectional fi ltered airfl ow within 

DESIGN OPTIONS 209 
a limited area. They may be located within a facility that provides the necessary 
temperature and humidity conditions or they may be provided with integral environmental 
control equipment designed to maintain necessary conditions. 
Air Change Rate The airfl ow pattern and air change rate in a clean room largely 
determines the class of cleanliness that can be maintained during a given operation. 
Non - unidirectional fl ow clean rooms rely on air dilution as well as a general ceiling - 
to - fl oor airfl ow pattern to continuously remove contaminants generated within the 
room. Unidirectional fl ow is more effective in continuously sweeping particles from 
the air due to the piston effect created by the uniform air velocity. The desired air 
change rate is determined based on the cleanliness class of the room and the density 
of operations expected in the room. An air change rate of 10 – 25 per hour is common 
for a large, low - density ISO 8 (Class 100,000) clean room. ISO 7 (Class 10,000) clean 
rooms typically require 40 – 60 air changes per hour. In unidirectional fl ow clean 
rooms, the air change rate is generally not used as the measure of airfl ow but rather 
the average clean - room air velocity is the specifi ed criterion. The average velocity 
in a typical ISO 5 (Class 100) clean room will be 70 – 90 ft/min. A tolerance of plus 
or minus 20% of design airfl ow is usually acceptable in the clean room. The foregoing 
values have been found to be appropriate in many facilities. Generally air change 
rate or air velocity is not a part of regulations. It is left to the user to demonstrate 
that the selected design parameter is appropriate for the products being manufactured. 
An exception to this may be in the case of fi lling operations where a unidirectional 
fl ow velocity of 90 ± 20 ft/min may be required. 
Pressurization A pressure differential should be maintained between adjacent 
areas, with the cleaner area having the higher pressure. This will minimize infi ltration 
of external contamination through leaks and during the opening and closing of 
personnel doors. A minimum overpressure between clean areas of 5 Pa [0.02 in. of 
TABLE 1 An Example of Cleanliness Classifi cation Goals 
Room 
Grade 
Cleanliness 
Class a 
Particle Counts e Microbial Contamination 
At Rest Operational 
Air 
Sample 
Settle 
Plates 
Contact 
Plates 
Glove 
Print 
(cfu/m 3 ) (cfu/4 h) b (cfu/plate) c (cfu/glove) d 
A f M3.5 (100) 3,500 3,500 < 1 < 1 < 1 < 1 
B g M3.5 (100) 3,500 35,000 10 5 5 5 
C M5.5 (10000) 350,000 3,500,000 100 50 25 — 
D M6.5 (100000) 3,500,000 N/A 200 100 50 — 
E Uncontrolled N/A N/A N/A N/A N/A N/A 
a In accordance with U.S. Federal Standard 209E. 
b 90 - mm - diameter settling plate. These are average values and individual plates may have < 4 h of exposure. 
c 55 - mm contact plates. 
d Five - fi ngered glove. 
e Maximum particle counts per cubic meter > 0.5 . m. 
f Unidirectional airfl ow at 90 ft/min. 
g Non - unidirectional airfl ow. 

210 CLEAN-FACILITY DESIGN, CONSTRUCTION, AND MAINTENANCE ISSUES 
water column (in. WC)] is recommended. The pressure between a clean area and 
an adjacent unclean area should be 12 – 14 Pa (0.05 in. WC). Where several clean 
rooms of varying levels of cleanliness are joined as one complex, a positive - pressure 
hierarchy of cleanliness levels should be maintained, including air locks and gowning 
rooms. Note that for certain processes and products it may be desirable to have a 
negative pressure relative to the surrounding ambient in one or more rooms when 
containment is a major concern. A “ room within a room ” may have to be designed 
to achieve this negative pressure yet still meet the needs of clean operation. 
Temperature Control Where occupant comfort is the main concern, a temperature 
of 68 – 70 ° F ± 2 ° F will usually provide a comfortable environment for people wearing 
a typical lab coat. Where a full “ bunny suit ” or protective attire is to be worn, room 
temperature as low as 66 ° F may be required. If the temperature is to be controlled 
in response to process concerns, the value and tolerance should be specifi ed early 
in the design phase to ensure that system selection is appropriate and that budgeting 
is accurate. Note that a tight tolerance (e.g., ± 1 ° F or less) will typically be more 
costly to maintain than a less stringent tolerance. 
Humidity Control The humidity requirement for comfort is in the range of 30 – 
60% relative humidity (RH). If process concerns suggest another value, it should 
be specifi ed as soon as possible in the design process. Biopharmaceutical materials 
sensitive to humidity variations or excessively high or low values may require stringent 
controls. 
3.3.3.3 Architectural Design Issues 
Facility Layout The facility layout should support the process contained within 
the clean room. While a rectangular shape is easiest to accommodate, other shapes 
may be incorporated into the facility as long as appropriate attention is paid to 
airfl ow patterns. The facility should be able to accommodate movement of equipment, 
material, and personnel into and out of the clean room. The layout of the 
clean suite should facilitate maintaining cleanliness class, pressure differentials, and 
temperature/humidity conditions by isolating critical spaces and by excluding nonclean 
operations. See Figure 1 . The potential for cross - contamination is addressed 
as both an architectural and a mechanical issue. Generally, in a facility where multiple 
products are to be processed, each product has a dedicated space, isolated 
physically from adjacent spaces, and each has its own air conditioning system, independent 
of adjacent systems. 
Air Locks or Anteroom This is a room between the clean room and an unrated 
or less clean area surrounding the clean room or between two rooms of differing 
cleanliness class. The purpose of the room is to maintain pressurization differentials 
between spaces of different cleanliness class while still permitting movement 
between the spaces. An air lock can serve as a gowning area. Certain air locks may 
be designated as an equipment or material air lock and provide a space to remove 
packaging material and/or to clean equipment or materials before they are introduced 
into the clean room. Interlocks are recommended for air lock door sets to 
prevent opening of both doors simultaneously. The air lock is intended to separate 
the clean from the unclean areas. 

DESIGN OPTIONS 211 
Prior to equipment or raw materials being introduced into the clean room, they 
should be prepared. This may mean removing an outer package wrap or perhaps 
surface cleaning of the object. Material handling equipment used within the clean 
room should be dedicated to the clean room. Physical barriers may be integrated 
into the material air lock design to prevent material handling equipment from 
leaving the clean room or outside equipment from passing into the clean room. 
Windows Windows are recommended in interior clean - room walls to facilitate 
supervision and for safety, unless prohibited by the facility protocol for visual security 
reasons. Windows in exterior building walls adjacent to a clean space are problematic. 
Windows can be a source of leakage and can result in contaminants entering 
the space. Windows should be placed to permit viewing of operations in order to 
minimize the need for non - clean - room personnel to enter the clean room. Windows 
should be impact - resistant glass or acrylic, fully glazed, installed in a manner that 
eliminates or minimizes a ledge within the clean space. Double glazing is frequently 
used to provide a fl ush surface on both sides of the wall containing the window. 
Windows may be included if there is a public relations requirement for visitors to 
view the operations. Speaking diaphragms or fl ush, wall - mounted, intercom systems 
are recommended near all windows to facilitate communication with occupants of 
the clean room. 
Pass - Through A pass - through air lock should be provided for the transfer of 
product or materials from uncontrolled areas into the clean room or between areas 
of different cleanliness class (Figure 2 ). The pass - through may include a speaking 
diaphragm, intercom, or telephone for communication when items are transferred 
and interlocks to prevent both doors from being opened at the same time. A cart - 
size pass - through installed at fl oor level can be used to simplify the movement 
of carts between clean areas. Stainless steel is typically the material of choice 
(Figure 3 ). 
FIGURE 1 Sample clean - room lay - out. 
Bench 
Emergency exit 
Main clean room 
Material 
air lock 
Window wall 
(Eg. 4' wide x 3' high x no. of windows) 
Gowning 
room 
Personal 
Locker 
Area 
Air lock 
Clean-room 
entrance 
Clean-room 
exit 
Pass-thru 
window 
Clean 
Garment 
Storage 
Soiled 
Garment 
Disposal 
Pass-thru 
window

212 CLEAN-FACILITY DESIGN, CONSTRUCTION, AND MAINTENANCE ISSUES 
Gowning Room Gowning rooms should be designed to support the garment protocol 
established for the facility. A typical gowning room may have a wall - or fl oor - 
mounted coat rack for clean garment storage (Figure 4 ); a bench specifi cally designed 
for clean - room use (Figure 5 ); a full - length mirror installed near the door for 
gowning self - inspection; storage for new packaged garments; and bins for disposal 
of soiled garments. 
Personal lockers and coat racks for the storage of notebooks, coats, and personal 
items should be located outside the gowning room or in an anteroom separate from 
FIGURE 2 Stainless steel pass - through with interlock designed to permit safe passage of 
small items between spaces of differing cleanliness. ( Courtesy of Terra Universal. ) 
FIGURE 3 Cart pass - through enabling larger amounts and sizes of items to be transported. 
Note that the cart shown is not to be taken from the clean room. Typically a physical barrier 
is incorporated into the cart pass - through design. ( Courtesy of Terra Universal. )

DESIGN OPTIONS 213 
the clean gowning area. Restroom facilities may also be located outside the gowning 
room or in an anteroom adjacent to the clean gowning area. A common gowning 
room design has two areas divided by a bench. The “ unclean ” area is used to remove 
and store outer garments. Stepping over the bench as the clean - room footwear is 
being put on ensures that the “ clean ” side of the gowning room will remain that 
way. Final donning of the clean - room garb is then accomplished. 
FIGURE 4 Furnishings in the gowning room are typically of a nonshedding material such 
as the stainless steel designs shown. The gown rack will generally have a ceiling - mounted 
HEPA fi lter above it to continually bathe the garments in clean air. ( Courtesy of Terra 
Universal. ) 
FIGURE 5 The stainless steel clean benches have has a perforated seat to permit airfl ow 
from ceiling to fl oor essentially unobstructed. ( Courtesy of Terra Universal. )

214 CLEAN-FACILITY DESIGN, CONSTRUCTION, AND MAINTENANCE ISSUES 
Male and female gowning rooms may be required depending on the make - up of 
the work force and the type of garments being used. 
Siting A clean room that serves as an element of a larger process line should be 
integrated into the line to permit movement of personnel and materials in and out 
of the room. A free - standing clean room may be located in any convenient site; 
however, certain conditions adjacent to the facility may degrade its performance. 
Vibration sources inside or near a clean room will encourage particle release within 
the room and under severe conditions may cause leaks in fi lters and ductwork. 
Heavy equipment, including the heating, ventilation, and air conditioning (HVAC) 
system components, pumps, house and vacuum system, ought to be vibration isolated. 
Location of a clean room directly adjacent to heavy equipment or loading 
docks that see heavy truck traffi c and other sources of vibration, shock, and noise 
may be problematic. The outdoor air intake for the clean - room makeup air must be 
carefully located to prevent overloading of fi lters or entrance of contaminating gases 
that the fi lter will not remove. Clean - room air intakes should not be located near 
loading docks, traffi c lanes, or other areas where vehicles may drive through or idle. 
These intakes should not be located near the exhaust locations of other processing 
facilities. Use of gas - phase fi ltration may be required if the quality of make - up air 
is not acceptable. 
3.3.3.4 Materials of Construction 
Walls Generally wall material selection should be based on the operations and 
material handling equipment to be used within the space. The walls should be 
strong enough to withstand repeated impact of carts or other equipment without 
deterioration. The materials should also be selected with the sanitizing protocol 
in mind. Chemicals, high - pressure wash, and steam can cause reduced wall life if 
proper materials are not selected. Seamless walls, to the extent possible, are 
desirable. 
Basic steel stud construction with gypsum board paneling can be used in biopharmaceutical 
clean rooms when appropriately coated with a nonshedding fi nish. 
Modular wall systems utilizing coated steel or aluminum panel construction are 
growing in popularity due to the ability to easily retrofi t a lab or production space 
at a later date with minimal disruption and construction debris. Stainless steel may 
be appropriate but costly. Modular systems have been developed that address the 
concerns of the biopharmaceutical clean - room user relative to surface fi nish integrity 
and smooth surfaces. The joint between adjacent modular panels is commonly 
treated with a gunnable sealant to provide a smooth, cleanable joint that will not 
hold contaminants. 
Concrete masonry unit (CMU) construction is widely used (Figure 6 ). It can 
prevent buildup of contaminants when fi nished with an epoxy or other smooth, 
chemical - resistant coating. Where retrofi t is not a regular practice, the strength of 
concrete block and its long life recommend it. 
Rounded, easy - to - clean corners and smooth transitions between architectural 
features such as windows and walls (Figure 7 ) should be featured in all wall system 
designs, whether modular or “ stick built. ” 

DESIGN OPTIONS 215 
Wall Finishes Inexpensive latex wall paints will deteriorate over time and are unacceptable 
in clean rooms. Acceptable wall fi nishes include epoxy paint, polyurethane, 
or baked enamel of a semigloss or gloss type. These may be applied in the factory to 
metal wall system panels. Field application of epoxy to gypsum board or CMU should 
be done to ensure a smooth, nonporous, monolithic surface that will not provide a 
breeding site for organisms. Exposed outside corners in high traffi c areas as well as 
on lower wall surfaces may have stainless steel facings or guards to prevent impact 
FIGURE 6 A CMU wall treated with block fi ller and epoxy fi nish to provide a smooth, 
cleanable wall surface. ( Courtesy of Niagara Walls. ) 
FIGURE 7 A window detail that provides a smooth, easy - to - clean surface on both wall 
faces. ( Courtesy of Portafab. ) 

216 CLEAN-FACILITY DESIGN, CONSTRUCTION, AND MAINTENANCE ISSUES 
damage to the wall. This is particularly true when gypsum board construction is used. 
Corner and wall guards should extend from the fl oor to at least the 4 - ft height. Traditionally 
the clean room has been white throughout as an indication of the clean nature 
of the facility and to identify it as a special work space. Other colors may be used in 
the clean room to provide an interesting environment as long as the materials of construction 
do not contribute particles to the air stream and will withstand the sanitizing 
agents and procedures used in the facility (Figure 8 ). 
Doors Entry should be through air locks to maintain clean - room pressure differentials. 
Emergency exit doors should incorporate a panic - bar mechanism (or a 
similar emergency opening device) with alarms for exit only. Emergency exit doors 
must be secured in a manner that prevents entry from the outside yet permits exiting 
from within. All doors should include essentially air - tight seals. Neoprene seals are 
generally acceptable. Brush - type door seals are not recommended. Foam rubber 
door seals are not recommended as these have been found to quickly deteriorate 
and shed particles. All personnel doors and swinging equipment doors should 
include self - closing mechanisms. Manual and automatic sliding doors may be useful 
when space is an issue or to facilitate movement between spaces of similar cleanliness 
class for personnel whose hands are otherwise engaged. As the mechanism of 
such doors can generate particles, a design specifi cally intended for clean - room 
application should be selected. 
Ceilings The ceiling fi nish should be similar to that used on the walls. The requirements 
for sanitizing typically address the ceiling as well as the walls and ceiling 
material and fi nish selection should refl ect this. Suspended ceilings using an inverted - 
T grid and lay - in panels may have a place in that part of the clean - room suite not 
subjected to the rigors of frequent sanitizing and where the possibility of trapped 
FIGURE 8 A modular wall system has been installed in a manner that provides a smooth 
surface for cleaning. The fi t of the components and the method of sealing are important when 
a modular wall is selected. ( Courtesy of Portafab. ) 

DESIGN OPTIONS 217 
spaces to support organism growth is not considered an issue (Figure 9 ). When suspended 
panel ceilings are used, the panels must be securely clipped or sealed in 
place to prevent movement due to air pressure changes. 
Modular wall systems designed for biopharmaceutical applications frequently 
have a “ walk - on ” ceiling designed using materials and fi nish similar to the wall. A 
rounded, easy - to - clean intersection between ceiling and walls should be a feature 
of the clean - room ceiling design, whether modular or stick built. Monolithic (seamless) 
ceilings can be installed using inverted - T grid supports and gypsum panels 
(Figure 10 ). This design permits incorporation of fi ltration and lighting into what is 
essentially a monolithic ceiling. 
FIGURE 9 A suspended ceiling utilizing lay - in panels and lay - in lighting troffers. A variety 
of cleanable materials can be used for the panels. The lay - in lights should be of 
a design that will provide appropriate service based on the cleaning protocol to be used. 
( Courtesy of CleanTek. ) 
FIGURE 10 An area of HEPA fi lters is installed above a process machining. Tear drop 
lighting is used to permit maximum fi lter coverage. A monolithic ceiling construction of 
gypsum panels suspended from a framework. The panels are fi nished with an epoxy coating 
compatible with cleaning/sterilization procedures. ( Courtesy of CleanTek. ) 

218 CLEAN-FACILITY DESIGN, CONSTRUCTION, AND MAINTENANCE ISSUES 
Floors Commonly used fl oor fi nishes for biopharmaceutical clean rooms include 
sheet vinyl installed using heat - welded or chemically fused seams to provide a seamless 
surface. Troweled epoxy and epoxy paint (Figure 11 ) have also found wide use. 
Compatibility of the fl oor material with solvents, chemicals, and cleaning agents to 
be used in the room must be considered. A minimum 4 - in. cove at the junction of 
fl oor and walls is recommended to facilitate cleaning. Some modular wall systems 
have a recess or offset that permits sheet vinyl to be installed in a manner that 
creates a seamless junction between fl oor and wall. When a stick - built approach is 
used, care should be taken to design cleanable intersections of walls and fl oors 
(Figure 12 ). 
3.3.3.5 HVAC System 
Air Side The clean - room HVAC system must be designed to maintain the required 
particulate cleanliness, temperature, humidity, and positive pressure at the expected 
outside environmental extremes and during the expected worst - case use operations. 
Rapid recovery from upset conditions such as door openings and contaminant - 
generating events is also a consideration. The high cost of conditioning outside air 
suggests that as much air as possible be recirculated. Recirculated air should be 
high - effi ciency particulate air (HEPA) fi ltered in those spaces requiring a cleanliness 
classifi cation in accordance with ISO 14644. Air that may be hazardous to 
health, even after HEPA fi ltration, should be exhausted after appropriate treatment. 
The required quantity of make - up air is calculated based on process exhaust plus 
air leakage from the clean room. A rate of two air changes per hour for clean room 
pressurization may be used in the absence of a more detailed calculation of air 
FIGURE 11 The process area is subjected to substantial chemical action due to the sterilizing 
protocol. It has a troweled epoxy, easy - to - clean fi nish. ( Courtesy of Dex - O - Tex. ) 

DESIGN OPTIONS 219 
leakage. Make - up air should be drawn from the outdoors, conditioned, and fi ltered 
as necessary before being introduced into the clean - room recirculation air stream. 
Care should be taken to ensure that make - up air intakes are not drawing in contaminated 
air. 
The potential for cross - contamination is an issue that should be addressed. A 
fl exible manufacturing facility is one in which a variety of products can be manufactured 
simultaneously. If the facility has a single air - handling system, the likelihood 
of materials from one space intruding into an adjacent space is high. For this 
reason each fi lling or compounding operation, or operation where noncompatible 
product can be expected to be picked up by the air stream, should be served by its 
own air - handling system (Figure 13 ). Isolated systems will minimize the possibility 
of cross - contamination. This can be a costly option and should not be undertaken 
FIGURE 12 The wall system used in the facility incorporates a monolithic sheet vinyl fl ooring 
junction between fl oor and wall face. Note the coving run up the wall around the edges 
to provide a smooth surface for cleaning. ( Courtesy of Portafab. ) 
FIGURE 13 The air handler has several stages of fi ltration combined with heating, cooling, 
humidifi cation, and dehumidifi cation capability. ( Courtesy of Air Enterprises. ) 

220 CLEAN-FACILITY DESIGN, CONSTRUCTION, AND MAINTENANCE ISSUES 
lightly. The current use of the plant and the anticipated future use should be assessed 
before a blanket decision that may lead to costly duplicated systems is made. 
Filtration The fi ltration system for a biopharmaceutical clean room typically consists 
of several stages of fi lters. Prefi lters are selected, sized, and installed to maximize 
the life of the fi nal HEPA fi lters. With proper selection of prefi lters, the fi nal 
HEPA fi lters should not require replacement within the life of the fi lter media and 
seal materials, a period of several years (perhaps as long as 10 – 15 years). Make - up 
air is commonly fi ltered by a low - effi ciency [30% as set by the American Society of 
Heating, Refrigerating, and Air Conditioning Engineers (ASHRAE)] prefi lter followed 
by an intermediate - (60% ASHRAE) or high - effi ciency (95% ASHRAE) 
fi nal fi lter (Figure 14 ). A screen should be included at the make - up air inlet to keep 
out pests and large debris. The make - up air is then directed to the recirculating air 
handler which also may have a low - effi ciency prefi lter, although prefi ltration of 
recirculated clean - room air is often omitted because of its high cleanliness level 
even after having passed through the clean room. The air is then directed through 
HEPA fi lters into the clean room. HEPA fi lters must be a minimum of 99.97% effi - 
cient on 0.3 - . m particles in accordance with military standard Mil - F - 51068 or the 
Institute of Environmental Science and Technology IEST - RP - CC001. Note that the 
fi ltration system for an unrated “ controlled area ” is the same, except that the HEPA 
fi lter stage may be omitted. Refer to Figure 15 . 
Filter Location HEPA fi lters may be installed in a facility either within an air 
handler or at the inlet to a plenum above the clean room or in the clean room ceiling. 
High - velocity HEPA fi lters, that is, fi lters with a face velocity up to 500 ft/min, are 
frequently installed in air handlers serving Class 100,000 clean rooms and are also 
used in make - up air handlers. Where hazardous materials may be trapped by the 
fi lters a “ bag - in – bag - out ” fi lter arrangement, such as that depicted in Figure 16 , may 
FIGURE 14 Non - unidirectional clean - room with lay - in HEPA fi lter modules. 
Make-up air unit 
Outside 
air 
95% Prefilter 
Cooling coil 
Reheat coil 
Humidifier 
Fan 
Prefilter 
Cooling coil 
Fan 
Air handler 
30% Prefilter 
Clean room 
HEPA filter modules 
Preheat coil 
Air handler 
Prefilter 
Cooling coil 
Fan

DESIGN OPTIONS 221 
be employed. Figure 17 shows a schematic arrangement with HEPA fi lters installed 
in the air handler. During the design phase care should be taken to provide access 
to both the upstream and downstream face of these fi lters to permit periodic challenging 
and leak testing. 
To provide HEPA fi ltered air over a limited area within a larger controlled space, 
a ceiling - mounted pressure plenum may be used. This plenum has an air distribution 
means at its lower face that permits air to be introduced in a unidirectional manner 
over the critical process area. Refer to Figure 18 . 
HEPA fi lters are installed at the upper face of the pressure plenum and the 
plenum is pressurized with fi ltered air. The ceiling - mounted HEPA fi lters have a 
face velocity up to 100 – 120 ft/min. This is somewhat higher than the HEPA fi lters 
serving the rest of the clean room. The fi lters are commonly supplied with air by a 
FIGURE 15 Several panel - type fi lters commonly used as prefi lters in air handlers. Second 
from left is a high - dust - loading fi lter available in ASHRAE effi ciency as high as 95% frequently 
used in make - up air handlers. If HEPA fi ltration of the make - up air is required, the 
high - velocity duct - mounted HEPA fi lter third from the left is appropriate. It can tolerate face 
velocities up to 500 fpm, compared to the standard HEPA, which is usually designed for face 
velocity on the order of 90 – 100 fpm. A standard HEPA designed for bio - pharma facility 
ceiling installation is shown at right. ( Courtesy of of CamFil. ) 
FIGURE 16 The “ bag - in – bag - out ” fi lter unit contains a HEPA fi lter and permits personnel 
to change the fi lter without coming into contact with possibly hazardous materials that may 
have been fi ltered from the air. ( Courtesy of Flanders Filters Inc. ) 

222 CLEAN-FACILITY DESIGN, CONSTRUCTION, AND MAINTENANCE ISSUES 
duct distribution network consisting of rectangular or round trunk ducts and fl exible 
or rigid round branch ducts. Full coverage, typical for ISO 5 (Class 100) clean rooms, 
or partial coverage, for higher class (less stringent) clean rooms, can be accomplished 
using 2 . 4 - ft lay - in HEPA fi lter modules installed in the ceiling. 
3.3.3.6 Clean - Room Testing 
ISO 14644 describes methodology and instrumentation for particle counting in the 
clean room. The tests described there are the basis for assigning a cleanliness rating 
FIGURE 17 Non - unidirectional clean - room with air handler mounted HEPA fi lters. 
Make-up air unit 
Outside 
air 
95% Prefilter 
Cooling coil 
Reheat coil 
Humidifier 
Fan 
Prefilter 
Cooling coil 
Fan 
Air handler 
30% Prefilter 
Clean room 
Preheat coil 
Air handler 
Prefilter 
Cooling coil 
Fan 
Ceiling diffuser 
HEPA filter 
HEPA filter 
FIGURE 18 Non - unidirectional clean - room with critical area unidirectional fl ow 
plenum. 
Make-up air unit 
Outside 
air 
95% prefilter 
Cooling coil 
Reheat coil 
Humidifier 
Fan 
Prefilter
Cooling coil 
Fan 
Air handler 
30% Prefilter Preheat coil 
Air handler 
Prefilter 
Cooling coil 
Fan 
Unidirectional airflow over 
critical process machine/surface 
Standard velocity 
HEPA filters 
Pressure plenum 
Air distributor 
Clean room 
High-velocity HEPA filters

to the facility. IEST - RP - CC006 similarly provides a procedure for particle counting 
but goes beyond that to a full series of tests that can be conducted to determine the 
effectiveness of the clean - room design and operability. The determination of which 
tests should be run is up to the clean - room end user. As a minimum, particle counting, 
room pressurization, and fi lter leakage tests should be run. Other tests dealing 
with airfl ow patterns, temperature, humidity, lighting, and sound levels are available. 
The array of tests selected is determined by the owner based on the effect the 
various design parameters will have on the product. The data obtained in acceptance 
tests become baseline data against which future testing is compared to determine 
if clean - room performance is changing over time. Ongoing periodic monitoring of 
the facility will ensure that clean - room performance degradation is identifi ed as it 
occurs. Pass – fail criteria are not part of the ISO standards but are to be developed 
on a case - by - case basis by the end user of the facility. These standards become part 
of the operational protocol of the facility. 
The clean - room testing described here is part of the commissioning or validation 
process wherein all equipment in the facility is run, tested, and observed to ensure 
it is working as designed. 
3.3.3.7 Utilities 
Biopharmaceutical clean - rooms typically house process equipment requiring utilities 
such as pure water, electricity, vacuum, and clean compressed air. The source of 
these utilities is usually outside the clean room. During the design phase a utility 
matrix is developed, in conjunction with end users and equipment manufacturers, 
identifying all equipment and the utilities required. This is the basis for determining 
the capacity of the utility systems as well as the point - of - use location of specifi c 
utilities. 
When bringing the utilities to the point of use, care should be taken to ensure 
that the clean room is not compromised. A clean construction protocol should be 
implemented and wall, ceiling, and fl oor penetrations, if needed, should be fl ashed 
and sealed in such a manner as to prevent contaminants from entering the clean 
room. Such entry points should also be smoothly sealed to ensure that there are no 
crevices to harbor organisms. Drains should be avoided in the clean room wherever 
possible. When this is not possible, the drains should be covered when not in use 
with a means specifi cally designed for biopharmaceutical clean - room application. 
Such means are tight, smooth, cleanable, and corrosion resistant. 
In small facilities an individual pipeline may be run from outside the facility to 
the point of use. In large facilities a utility chase (Figure 19 ) that enables major 
utility lines to be brought to the vicinity of process tools may be provided. Final 
hook - up between the chase and point of use then becomes a relatively simple, 
minimally intrusive procedure. The utility chase concept is also benefi cial in facilities 
that undergo frequent retrofi t or upgrade. 
3.3.4 CONSTRUCTION PHASE: CLEAN BUILD PROTOCOL 
Ongoing experience has demonstrated that an aggressive clean construction protocol 
program is generally not required for biopharmaceutical facilities that do not 
CONSTRUCTION PHASE: CLEAN BUILD PROTOCOL 223

224 CLEAN-FACILITY DESIGN, CONSTRUCTION, AND MAINTENANCE ISSUES 
carry a cleanliness rating. Where cleanliness classifi cations less stringent than Class 
10,000 are used, standard construction techniques followed by careful cleanup and 
wipe - down within the clean space have proven quite acceptable. Cleanliness levels 
of Class 1000 or Class 10,000 are achievable shortly after startup and maintainable 
thereafter. For cleanliness levels of Class 100 a somewhat more restrictive protocol 
is required. Once a facility is up and running, any intrusion into clean areas for retrofi 
t work should be done in conjunction with some level of clean build protocol 
in place, dependent on the rating of the facility and the degree of disruption 
encountered during the retrofi t project. 
The levels of clean construction described herein can provide a practical means 
of meeting operational cleanliness goals in a cost - effective fashion. Each project, 
whether new construction or retrofi t of an existing process, should have as part of 
it an evaluation of the required elements of the build clean protocol to be employed. 
The information provided below is broad and can act as a template for the protocol 
put in place for a specifi c project. 
A key to successful clean construction is the appointment of an individual as a 
clean - room monitor who is well versed in the clean - room construction protocol. 
That person is charged with maintaining a clean environment and monitoring the 
activities of all personnel within the clean area during the construction phase and 
is concerned with maintaining budget and schedule goals. The clean - room monitor 
should have the confi dence to make “ real - world ” decisions supporting the “ spirit ” 
of the protocol as well as the “ letter. ” 
FIGURE 19 The utility chase is located between two clean rooms. Major utility lines are 
installed within the chase and hook - up lines for local pieces of process equipment are connected 
through the clean - room walls. A major benefi t of this arrangement is that installers 
need not be fully garbed in clean - room attire to access the utility lines. ( Courtesy of 
CleanTek. )

3.3.4.1 General 
All clean - facility construction, while employing standard construction techniques, 
should be accomplished in a manner that does not create excessive particulate 
contamination. A temporary lay - down area within the building adjacent to the clean 
area should be set aside for storage of clean construction components. All tools used 
for clean construction should be in an “ as - new ” condition and be cleaned and 
inspected prior to use. The pass – fail criteria for tool and material inspection is “ no 
visible dirt. ” 
Cleanup within the clean area at the end of each shift should consist of broom 
cleaning and vacuum cleaning the fl oor with a clean vacuum, that is, a vacuum with 
a HEPA fi lter (99.97% effi cient on 0.3 - . m particles). 
Clean - facility construction materials should be left in an outer shipping wrap 
until moved to the temporary lay - down area, where they should then be unwrapped 
and wiped down before being moved into the clean space. 
Adherence to these guidelines will make fi nal clean - up faster and acceptable 
start - up and certifi cation/validation more certain. While a goal of clean construction 
is rapid start - up and certifi cation/validation, a long - range goal is the maintenance 
of the facility cleanliness without intrusion, over an extended period of time, of 
contaminants deposited during construction due to a poor protocol or improper 
implementation of the protocol. 
Appendix A and Appendix B offer a template for working in a clean environment 
as well as clean - room cleaning procedures. Procedures should be modifi ed 
with caution to suit a particular project. 
3.3.4.2 Level I Clean Construction 
Level I clean construction is used for all areas with a cleanliness rating of Class 1000 
(ISO 6) or higher (less stringent), including those spaces within which clean processes 
are conducted in minienvironments/isolators and those unrated areas identi- 
fi ed as being “ controlled environments. ” 
Standard construction techniques are used until the clean - room envelope is 
completed, HEPA fi lters with protective fi lm in place are installed and air handlers 
are ready to start. The clean envelope consists of clean - room walls, ceiling, and fl oor. 
Prior to starting the air handlers, a thorough clean - up of the space within the clean 
envelope is accomplished as described in Appendix B, 2A – 2J. Following clean - up 
and start - up of the clean - space air - handling system, particle counts should quickly 
drop to well within operational requirements. 
Once the clean room is operational, as described above, additional construction 
related to process equipment installation and facility modifi cation within the clean 
room can be done in compliance with the Guidelines in Appendix A. 
3.3.4.3 Level II Clean Construction 
This is used for construction of those areas rated at Class 100 (ISO 5) employing a 
100% HEPA fi lter ceiling. Generally standard construction techniques should be 
used. The clean - room envelope includes walls, ceiling, fl oor, return ductwork, supply 
fans, and supply ductwork. 
CONSTRUCTION PHASE: CLEAN BUILD PROTOCOL 225

226 CLEAN-FACILITY DESIGN, CONSTRUCTION, AND MAINTENANCE ISSUES 
All ductwork sections should be cleaned and sealed with plastic wrap at the time 
of fabrication until just prior to installation or start - up to prevent contaminants from 
accumulating inside air - handling passageways. The sections of ductwork should be 
unsealed only as required for installation. Open ends of ducts and fans should 
remain sealed until connecting duct is about to be installed. A fi nal isopropyl alcohol 
(IPA) wipe - down of all interior duct sections and fan surfaces should be done 
immediately prior to installation. 
When general construction of the clean room is completed, steps 2A – 2J of 
Appendix B describing coarse cleaning can be implemented. Following coarse 
cleaning the protective fi lm can be removed from the HEPA fi lters and the air - 
handling system started. 
Successful completion of the cleaning process described above will indicate that 
installation of process equipment may begin. Note that procedures described in 
Appendix A should be followed. After installation of all process equipment or when 
the clean room is to be prepared for certifi cation, steps 2K – 2O of Appendix B for 
fi nal wipe - down can be followed. 
A black - and - white felt rub - down test is performed to demonstrate adequate 
cleanliness of the interior clean - envelope surfaces. This test consists of both black - 
and - white felt being wiped over any surface for 1 m linear distance with a fi rm hand 
pressure. No residue should be visible on the cloth. Each cloth should be 60 cm 
square black or white static - free natural fi ber felt folded with cut edges inside to a 
25 - cm square. The cut edges should be sealed with an approved latex sealant. 
3.3.5 MAINTENANCE 
To maximize the life and effectiveness of the facility, it must be maintainable. The 
facility should be designed to permit ongoing day - to - day preventive maintenance 
of the mechanical systems and, should a failure occur, permit needed repairs to be 
made in an expeditious manner. Perhaps of equal importance is the janitorial maintenance 
required to keep the facility suitable for pharmaceutical manufacturing. 
Proper janitorial maintenance begins with the design of the facility and evolves into 
an operational protocol, personnel training, and effective implementation. 
In the design phase it is important to provide suffi cient access to mechanical and 
process equipment to enable preventive maintenance procedures to be carried out 
with minimum effort. Typically manufacturer ’ s installation instructions offer guidelines 
as to how much space should be left open around equipment to permit removal 
of critical components. One driver of construction cost is fl oor space. Making a space 
as small as possible to house an operation presumably will result in fi rst - cost savings. 
If the space does not provide suffi cient access for lubrication, fi lter changes, belt 
adjustments, and the like, there is a strong possibility that this preventive maintenance 
will be ignored. A predictable result is shortened equipment life and the disappearance 
of any fi rst - cost savings that may have been realized. If there is a major 
equipment failure that requires replacement of an inaccessible component, the cost 
associated with knocking down a wall to gain equipment access will very likely 
negate fi rst - cost savings. 
Storage of maintenance items should be identifi ed early in the design process. 
Spare - parts storage, janitorial supply storage, janitors ’ sink closets, repair work shops, 

and storage space for consumable maintenance items (e.g. air fi lters) will require fl oor 
space in the facility design. Frequently tools are dedicated to the clean facility or are 
required specifi cally for unique process equipment and must also have a storage area. 
Accommodation of these items is an important part of the planning process. 
A requirement of a clean facility is that the cleaning materials should be specifi - 
cally intended for use in a “ clean ” operation, should be kept in good ( “ like new ” ) 
repair, and should not be used in other, nonclean, areas of the facility. Using general 
cleaning materials manned by the “ house ” janitorial staff will invariable introduce 
more contamination into the clean portions of the facility than it removes. 
A central housekeeping vacuum is very useful in keeping contamination under 
control. While “ wet - and - dry ” versions of the central vacuum are available, the 
manner in which each is to be used should be carefully reviewed to ensure that it 
is in keeping with the sanitary requirements of the facility. A common housekeeping 
procedure addresses spills with local clean - up and uses a dry - type central vacuum 
for dry particulate contaminants. 
APPENDIX A: GUIDELINES FOR CONSTRUCTION PERSONNEL AND 
WORK TOOLS IN A CLEAN ROOM 
1.0 General requirements 
A. Makeup will not be allowed inside the clean room. 
B. Smoking will not be allowed in or around the clean room. 
C. Tobacco chewing will not be allowed in the clean room. 
D. Paper or paper by - products will not be allowed in the clean room except 
clean - room approved paper and pens. 
E. Prints or papers will be allowed only if totally laminated in plastic and 
cleaned with isopropyl alcohol prior to entry. 
F. Lead pencils will not be allowed in the clean room. Ball point pens only. 
G. Clean - room garments, to include shoe covers, coveralls, and head cover, 
will be worn within the clean room. 
H. Clean - room garments will not be unfastened or unzipped while inside the 
clean room. 
I. No writing will be allowed on the clean - room garments. 
J. Food and drink will not be allowed in the clean room. 
K. Combing of hair will not be allowed in the clean room or gowning area. 
L. Stepping on chairs, work benches, test equipment or process equipment is 
not allowed. 
M. Damaged garments (rips, worn booties, torn gloves) will be replaced immediately. 
Do not wait for a convenient time. DO IT NOW! 
N. Tool pouches are not allowed in a clean room. 
O. All work areas and adjacent areas will be vacuumed after completion of 
work and prior to leaving the clean room. 
P. ALWAYS wash hands before entering the clean room to remove residues 
from food, smoke, and/or other sources. 
2.0 Personnel 
A. All personnel working inside a clean room will be required to follow all 
dress codes associated with the particular clean space. 
APPENDIX A 227

228 CLEAN-FACILITY DESIGN, CONSTRUCTION, AND MAINTENANCE ISSUES 
B. Street clothes or company uniforms will be allowed as standard undergarments 
provided they are well maintained and clean. No such garments will 
be allowed that are soiled with grease, dirt, or any detectable stains. 
C. Any garments producing excessive fi bers (such as fuzzy sweaters) will not 
be allowed as an undergarment. 
D. Standard safety shoes (or other specifi ed footware) will be required. Shoe 
covers must be worn. 
E. Bare feet, socks, and stockings are not allowed inside booties. 
F. Coats, lunches, and private items will not be allowed inside the clean 
room. 
3.0 Gowning procedure 
A. Each individual is responsible for knowing and using the correct method 
of gowning prior to entering the clean room. (See Figure 20 .) 
1. Clean shoes prior to entering the gowning room. 
2. The order of dress should be as follows: 
a. Shoe covers 
b. Hairnet/beard cover (required after fi nal cleaning) 
c. Hood 
d. Coveralls 
e. Face cover (required after fi nal cleaning) 
f. Gloves (required after fi nal cleaning) 
B. Ensure that hoods are tucked inside neck opening of coveralls and pants 
legs are tucked and snapped inside booties. Garments are to be snapped 
FIGURE 20 Clean - room garments are intended to keep contaminants from entering the 
clean room. In a critical environment the “ bunny suit ” shown may be required. In a less critical 
environment a lab coat may suffi ce. The clean - room construction protocol should identify 
the type of garment that will be required at various stages of construction and for process 
equipment installation. ( Courtesy of Terra Universal. )

closed at the neck, wrist and ankle opening and sleeves tucked inside 
gloves. 
C. All head hair must be covered at all times. 
D. Do not allow garments to touch the fl oor while dressing or undressing. 
E. Avoid leaning on walls, lockers, or other personnel at all times. DO NOT 
place feet on benches. 
F. The order of undress should be as follows: 
1. Gloves 
2. Coveralls 
3. Face cover and hood 
4. Shoe covers 
G. If you will be reentering the clean room, unsoiled garments may be hung 
for reuse; gloves are not to be reused. 
4.0 Work tools, parts, and equipment 
A. All tools and equipment used in a clean room should be in like - new 
condition. 
B. All parts will be removed from their shipping container prior to cleaning 
and introduction into the clean room. NO PAPER PRODUCTS will be 
allowed inside the clean room. 
C. All tools, parts, and equipment will be properly cleaned prior to entering 
the clean room. Minimum cleaning should be a total wipe - down with isopropyl 
alcohol, using certifi ed clean - room wipes, to assure that the last wipe 
does not leave visible residue on the wipe. Parts should be blown off 
outside the clean room using fi ltered nitrogen when available. 
D. All parts and equipment should be sent through the equipment wipe - down 
area (material air lock) and not carried through the gowning area. 
5.0 Working in a Clean Room 
A. A major concern when working in a clean room is the generation of particles 
of the size that cannot be seen and spreading these particles throughout 
the clean room. Every possible precaution must be taken to contain 
these contaminants and protect the clean - room environment. Everything 
that is done as a standard operation must be analyzed to determine if it 
will adversely affect the cleanroom. If you have any concerns, ask the clean - 
room monitor before you damage the environment and incur unnecessary 
clean - up cost. 
B. All procedures must be reviewed with the clean - room monitor to ensure 
compliance with clean - room operation practices. All procedures that can 
generate particles should be done outside the clean - room whenever possible. 
In the listing below all prohibited procedures are subject to review 
by the clean - room monitor. The intent is to get the job done; however, some 
preplanning with the clean - room monitor can result in a positive result and 
a clean facility. 
1. Drilling: 
a. All power drills will be wrapped to encapsulate any contamination 
generated during operation. 
b. Drills may be operated in sealed enclosures equipped with an exhaust 
vacuum. 
APPENDIX A 229

230 CLEAN-FACILITY DESIGN, CONSTRUCTION, AND MAINTENANCE ISSUES 
c. Surface to be drilled will be tented or vacuumed to prevent the 
spread of contamination. 
2. Grinding: NO GRINDING will be allowed in the clean room. 
3. Welding: NO WELDING will be allowed inside the clean room. 
4. Soldering: May be allowed after total review. 
5. Painting: NO PAINTING will be allowed after the start - up of the clean 
room. 
6. Sanding/fi ling: Will be allowed only within a properly tented space. 
7. Cutting: Will be allowed with clean - room approved vacuums removing 
particles created. 
APPENDIX B: CLEANING THE CLEANROOM 
1.0 Final cleaning: During this cleaning phase, the clean - room proper should be 
prepared for certifi cation to the appropriate cleanliness level. Extreme care 
must be exercised by all those involved in this procedure to minimize the 
potential for contamination. 
2.0 Procedure 
A. Secure the entrance to the clean space “ envelope, ” the gowning area, and 
the entrance from the gowning room to the clean room with locks or 
limited access via card keys. Post a restriction notice: YOU ARE ENTERING 
A CONTROLLED, PARTICULATE - FREE ENVIRONMENT, 
CONTACT ____________ FOR PERMISSION TO ENTER. 
B. Perform two coarse cleanings of the clean room. Each cleaning should 
include the following: 
1. Wipe - down of the HEPA fi lter grid, all lights, walls, fl oors, windows, and 
all exposed interior surfaces. This will include any outlet boxes or fl oor/ 
wall recesses. 
2. Wipe - down should be by clean potable water and mild nonphosphate 
detergent using clean, lint - free cloths approved by the clean - room 
monitor. 
3. A second wash - down should commence using clean potable water in 
the same manner. 
4. Floors should be scrubbed and polished using a fl oor - polishing machine. 
No wax is to be used. 
C. Partitions and fl oors should be washed to maintain a dust - free condition. 
D. Access into the clean room should be restricted to discourage infi ltration 
of outside particulates. 
E. Tacky mats 3 ft by 6 ft should be installed inside the entrance of the gowning 
area as well as at the entrance to the clean room. 
F. Foot covers should be worn by all personnel entering the clean room after 
the second coarse cleaning is complete. 
G. Caulking crew should be assigned and commence work after the second 
coarse cleaning is completed. They should complete all caulking as required 
by specifi cation. 
H. Simultaneously with the caulking procedure, air handlers and associated 
ducts and plenums should be checked for cleanliness. 

I. The HEPA fi lter protective fi lm should be removed. Air - handling equipment 
should be activated and the clean space pressurized to maintain a 
positive static pressure. 
J. From this point forward, clean - room garments and head covers should be 
worn by all personnel entering the clean space. 
K. Commence with the fi rst of two fi nal wipe - downs. Nonshedding clean - 
room wipes (saturated with isopropyl alcohol) or tacky wipes should be 
used. All exposed surfaces should be wiped. 
L. A particle counter should be installed in the clean space and samples taken 
at several control points over the next 48 hours. A steady decrease in the 
particle count over time should be achieved. 
M. If particle counts stabilize at a level above that desired, a search for fi lter 
leakage will be required. 
N. If the search for fi lter leakage fails to fi nd a leak, the entire area should be 
recleaned as described for the fi nal wipe - down. 
O. Once the air standards are achieved, fi nal air balance can begin followed 
by clean - room certifi cation testing. 
BIBLIOGRAPHY 
U . S . Food and Drug Administration, Washington, DC 
21 CFR Part 210, Current good manufacturing practice in manufacturing, processing, packing, 
or holding of drugs. 
21 CFR Part 211, Current good manufacturing practice for fi nished pharmaceuticals. 
Institute of Environmental Sciences and Technology, Rolling Meadows, IL 
IEST - RP - CC001.4: HEPA and ULPA Filters , Nov. 7, 2005 . 
IEST - RP - CC002.2: Unidirectional Flow Clean - Air Devices , Jan. 19, 1999 . 
IEST - RP - CC003.3: Garments Systems Considerations for Cleanrooms and Other Controlled 
Environments , Aug. 11, 2003 . 
IEST - RP - CC004.3: Evaluating Wiping Materials Used in Cleanrooms and Other Controlled 
Environments , Aug. 23, 2004 . 
IEST - RP - CC005.3: Gloves and Finger Cots Used in Cleanrooms and Other Controlled 
Environments , May 1, 2003 . 
IEST - RP - CC006.3: Testing Cleanrooms , Aug. 30, 2004 . 
IEST - RP - CC008 - 84: Gas - Phase Adsorber Cells , Nov. 1, 1984 . 
IEST - RP - CC012.1: Considerations in Cleanroom Design , Mar. 1, 1998 . 
IEST - RP - CC013 - 86 - T: Equipment Calibration or Validation Procedures , Nov. 1, 1986 . 
IEST - RP - CC016.2: The Rate of Deposition of Nonvolatile Residue in Cleanrooms , Nov. 15, 
2002 . 
IEST - RP - CC018.3: Cleanroom Housekeeping: Operating and Monitoring Procedures , Jan. 1, 
2002 . 
IEST - RP - CC019.1: Qualifi cations for Organizations Engaged in the Testing and Certifi cation 
of Cleanrooms and Clean - Air Devices , Jan. 23, 2006 . 
IEST - RP - CC023.2: Microorganisms in Cleanrooms , Jan. 31, 2006 . 
BIBLIOGRAPHY 231

232 CLEAN-FACILITY DESIGN, CONSTRUCTION, AND MAINTENANCE ISSUES 
IEST - RP - CC026.2: Cleanroom Operations , July 21, 2004 . 
IEST - RP - CC027.1: Personnel Practices and Procedures in Cleanrooms and Controlled Environments 
, Apr. 1, 1999 . 
IEST - RP - CC028.1: Minienvironments , Sept. 1, 2002 . 
IEST - RP - CC034.2: Hepa and ULPA Filter Leak Tests , June 23, 2005 . 
IEST - STD - CC1246D: Product Cleanliness Levels and Contamination Control Program , 
Jan. 1, 2002 . 
International Organization for Standardization ( ISO ) Standards 
ISO 14644 - 1: Classifi cation of air cleanliness. 
ISO 14644 - 2: Specifi cations for testing and monitoring to prove continued compliance. 
ISO 14644-3: Test methods. 
ISO 14644 - 4: Design, construction and start - up. 
ISO 14644 - 5: Operations. 
ISO 14644-6: Terms and defi nitions. 
ISO 14644 - 7: Separative devices (clean air hoods, gloveboxes, isolators, and 
minienvironments). 
ISO 14644 - 8: Classifi cation of airborne molecular contamination. 
ISO 14698 - 1: Bicontamination control — General principles. 
ISO 14698 - 2: Biocontamination control — Evaluation and interpretation of biocontamination 
data. 
ISO 14698 - 3: Biocontamination control — Methodology for measuring the effi ciency of processes 
of cleaning and/or disinfection of inert surfaces bearing biocontaminated wet soiling 
or biofi lms. 

NORMAL DOSAGE FORMS 
SECTION 4


235 
4.1 
SOLID DOSAGE FORMS 
Barbara R. Conway 
Aston University, Birmingham, United Kingdom 
Contents 
4.1.1 Biopharmaceutics Classifi cation System 
4.1.2 Systematic Formulation Development 
4.1.3 Standard and Compressed Tablets 
4.1.4 Excipients in Solid Does Formulations 
4.1.4.1 Diluents 
4.1.4.2 Binders 
4.1.4.3 Lubricants 
4.1.4.4 Glidants and antiadherents 
4.1.4.5 Disintegrants 
4.1.4.6 Superdisintegrants 
4.1.4.7 Added Functionality Excipients 
4.1.4.8 Colorants 
4.1.4.9 Interactions and Safety of Excipients 
4.1.5 Coated Tablets 
4.1.5.1 Sugar - Coated Tablets 
4.1.5.2 Compression Coating and Layered Tablets 
4.1.5.3 Film - Coated Tablets 
4.1.5.4 Tablet Wrapping or Enrobing 
4.1.6 Hard and soft gelatin capsules 
4.1.6.1 Hard - Shell Gelatin Capsules 
4.1.6.2 Manufacture of Hard Gelatin Shells 
4.1.6.3 Hard Gelatin Capsule Filling 
4.1.6.4 Soft Gelatin Capsules 
4.1.6.5 Manufacture of Soft Gelatin Capsules 
4.1.6.6 Dissolution Testing of Capsules 
4.1.7 Effervescent Tablets 
4.1.7.1 Manufacture of Effervescent Tablets 
4.1.8 Lozenges 
4.1.8.1 Chewable Lozenges 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

236 SOLID DOSAGE FORMS 
4.1.9 Chewable Tablets 
4.1.9.1 Testing of Chewable Tablets 
4.1.10 Chewing Gums 
4.1.10.1 Composition of Chewing Gum 
4.1.10.2 Manufacture of Chewing Gum 
4.1.10.3 Drug Release from Chewing Gums 
4.1.10.4 Applications for Chewing Gums 
4.1.11 Orally Disintegrating Tablets 
4.1.11.1 Dissolution Testing of ODTs 
4.1.12 Solid Dosage Forms for Nonoral Routes 
References 
Drug substances are most frequently administered as solid dosage formulations, 
mainly by the oral route. The drug substance ’ s physicochemical characteristics, as 
well the excipients added to the formulations, all contribute to ensuring the desired 
therapeutic activity. Tablets and capsules are the most frequently used solid dosage 
forms, have been in existence since the nineteenth century, and are unit dosage 
forms, comprising a mixture of ingredients presented in a single rigid entity, usually 
containing an accurate dose of a drug. There are other types of solid dosage forms 
designed to fulfi ll specifi c delivery requirements, but they are generally intended for 
oral administration and for systemic delivery. The major solid oral dosage form is 
the tablet, and these can range from relatively simple, single, immediate - release 
dosage forms to complex modifi ed - release systems. Tablets offer advantages for 
both patients and manufacturers (Table 1 ). Most tablets are intended to be swallowed 
whole and to rapidly disintegrate and release drug in the gastrointestinal 
tract. Tablets are classifi ed by their route of administration or their function, form, 
or manufacturing process. For example, some tablets are designed to be placed in 
the oral cavity and to dissolve there or to be chewed before swallowing, and 
there are many kinds of formulation designed for sustained or controlled release 
(Table 2 ). 
Solid dose formulations, including tablets, must have the desired release properties 
coupled with manufacturability and aesthetics and must involve rational formulation 
design. The dose of the drug and its solubility are important considerations 
TABLE 1 Advantages of Tablets as a Dosage Form 
Easy to handle 
Variety of manufacturing methods 
Can be mass produced at low cost 
Consistent quality and dosing precision 
Can be self - administered 
Enhanced mechanical, chemical, and microbiological stability compared to liquid dosage 
forms 
Tamperproof 
Lend themselves to adaptation for other profi les, e.g., coating for sustained release 

in the design of the formulation as are the type of dosage form and its method of 
preparation. 
4.1.1 BIOPHARMACEUTICS CLASSIFICATION SYSTEM 
Dissolution of the drug must occur before or on reaching the absorption site before 
absorption can occur, and generally water - soluble drugs do not exhibit formulation 
diffi culties. For poorly water - soluble drugs, the absorption rate may be dictated by 
the dissolution rate, and, if dissolution is slow, bioavailability may be compromised. 
The solubility of a drug should, therefore, be considered along with its dose when 
designing formulations, and unsuitable biopharmaceutical properties is the major 
reason for the failure of new drugs. 
In 1995, the Biopharmaceutics Classifi cation System (BCS) was devised to classify 
drugs based on their aqueous solubility and intestinal permeability [1] . According 
to the BCS, drug substances are classifi ed as follows [2] : 
Class I: high permeability, high solubility 
Class II: high permeability, low solubility 
Class III: low permeability, high solubility 
Class IV: low permeability, low solubility 
TABLE 2 Types of Solid Dosage Form 
Formulation type Description 
Immediate - release tablet/capsule Intended to release the drug immediately after 
administration 
Delayed - release tablet/capsule Drug is not released until a physical event has 
occurred, e.g., change in pH 
Sustained - release tablet/capsule Drug is released slowly over extended time 
Soluble tablets Tablet is dissolved in water prior to administration 
Dispersible tablet Tablet is added to water to form a suspension prior to 
administration 
Effervescent tablet Tablet is added to water, releasing carbon dioxide to 
form a effervescent solution 
Chewable tablet Tablet is chewed and swallowed 
Chewable gum Formulation is chewed and removed from the mouth 
after a directed time 
Buccal and sublingual tablets Tablet is placed in the oral cavity for local or systemic 
action 
Orally disintegrating tablet Tablet dissolves or disintegrates in the mouth without 
the need for water 
Lozenge Slowly dissolving tablet designed to be sucked 
Pastille Tablet comprising gelatin and glycerine designed to 
dissolve slowly in the mouth 
Hard gelatin capsule Two - piece capsule shell that can be fi lled with powder, 
granulate, semisolid or liquid 
Soft gelatin capsule (softgel) One - piece capsule containing a liquid or semisolid fi ll 
BIOPHARMACEUTICS CLASSIFICATION SYSTEM 237

238 SOLID DOSAGE FORMS 
A dose solubility volume can be defi ned for all drugs (i.e., the volume required 
to dissolve the dose). A drug substance is considered highly soluble when the highest 
dose strength is soluble in . 250 mL water over a pH range of 1 – 7.5. A drug substance 
is considered highly permeable when the extent of absorption in humans is determined 
to be . 90% of an administered dose, based on mass balance or in comparison 
to an intravenous reference dose. A drug product is considered to be rapidly dissolving 
when . 85% of the labeled amount of drug substance dissolves within 30 min 
using U.S. Pharmacopeia (USP) apparatus I or II in a volume of . 900 mL buffer 
solutions. 
It was recognized that dissolution rate has a negligible impact on bioavailability 
of highly soluble and highly permeable (BCS class I) drugs when dissolution of their 
formulation is suffi ciently rapid. As a result, various regulatory agencies including 
the U.S. Food and Drug Administration (FDA) now allow bioequivalence of formulations 
of BCS class I drugs to be demonstrated by in vitro dissolution (often 
called a biowaiver) [3] . Therefore, one of the goals of the BCS is to recommend a 
class of immediate - release (IR) solid oral dosage forms for which bioequivalence 
may be assessed based on in vitro dissolution tests. 
4.1.2 SYSTEMATIC FORMULATION DEVELOPMENT 
Systematic development approaches are needed to gather a full and detailed understanding 
of marketable formulations in order to satisfy the requirements of regulatory 
bodies and to provide a research database. Effi cient experimental design using 
in - house or commercial software packages can ensure quality while avoiding expensive 
mistakes and lost time. Information from various categories such as the properties 
of the drug substance and excipients, interactions between materials, unit 
operations, and equipment are required [4] . Design of experiments (DOE) and 
statistical analysis have been applied widely to formulation development. Using 
DOE facilitates evaluation of all formulation factors in a systematic and timely 
manner to optimize the formulation and manufacturing process. Abbreviated excipient 
evaluation techniques such as Plackett – Burman design can be applied to minimize 
the number of experiments and identify critical components or processes. 
Optimization processes can then be applied. When the formulation and manufacturing 
processes of a pharmaceutical product are optimized by a systematic approach, 
the scale - up and processes validation can be very effi cient because of the robustness 
of the formulation and manufacturing process. 
Innovations in statistical tools such as multivariate analysis, artifi cial intelligence, 
and response surface methodology have enabled rational development of formulations, 
and such methods allow formulators to identify critical variables without 
having to test each combination. 
4.1.3 STANDARD AND COMPRESSED TABLETS 
The simplest tablet formulations are uncoated products that are made by direct 
compression or compression following wet or dry granulation. They are a versatile 
drug delivery system and can be intended for local action in the gastrointestinal 

(GI) tract or for systemic effects. General design criteria for tablets are accuracy 
and uniformity of drug content, stability of the drug candidate and the formulation, 
optimal dissolution and availability for absorption (whether immediate or extended 
release), and patient acceptability in terms of organoleptic properties and appearance. 
Flocculant, low - density drugs can be diffi cult to compress and formulate into 
tablets. This is a particular issue with drugs of low potency. Also some poorly water - 
soluble, poorly permeable drugs or highly metabolized drugs cannot be given this 
way. Additionally, local irritant effects can be harmful to the mucosa of the GI 
tract. 
Tablets are a popular dosage form due to their simplicity and economy of manufacture, 
relative stability, and convenience in packaging, shipping, and storage. For 
the patient, uniformity of dose, blandness of taste, and ease of administration ensure 
their popularity. Thus, the purpose of the formulation and the identifi cation of suitable 
excipients are of primary importance in the development of a successful formulation. 
A well - designed formulation should contain, within limits, the stated 
quantity of active ingredient, and it should be capable of releasing that amount of 
drug at the intended rate and site. Tablets need to be strong enough to withstand 
the rigors of manufacture, transport, and handling, and they need to be of acceptable 
size, taste, and appearance. A typical manufacturing process for a tablet product 
includes weighing, milling, granulation and drying, blending and lubrication, compression, 
and coating. Each processing step involves several process parameters. For 
a given formulation, all processing steps should be thoroughly evaluated so that a 
robust manufacturing process can be defi ned, and DOE can be applied effectively 
to optimize this process. 
Direct compression is a simple process being more economical and less stressful 
to ingredients in terms of heat and moisture, However, there are limitations governed 
by the physical properties of the ingredients, and raw materials must be carefully 
controlled. It is diffi cult to form directly compressed tablets containing 
high - dose and poorly compactible drugs. Granulation can be employed to improve 
the compaction characteristics of the powder. Granulation can also improve fl ow 
properties and reduce the tendency for segregation of the mix due to a more even 
particle size and bulk density. Granules can be produced by either wet or dry 
methods based on the stability of the drug and excipients. 
Although the basic mechanical process of producing tablets by compression has 
not changed, there has been much work on improving tableting technology [5] . 
Understanding of the physical and mechanical properties of powders and the compaction 
process has improved and will continue to improve product design while 
increases in the speed and uniformity of action of tableting presses improve the 
process. 
4.1.4 EXCIPIENTS IN SOLID DOSE FORMULATIONS 
In addition to the active ingredients, solid oral dosage forms will also contain a range 
of substances called excipients. The role of excipients is essential in ensuring that 
the manufacturing process is successful and that the quality of the resultant formulation 
can be guaranteed. The appropriate selection of excipients and their relative 
concentrations in the formulation is critical in development of a successful product. 
EXCIPIENTS IN SOLID DOSE FORMULATIONS 239

240 SOLID DOSAGE FORMS 
Although they are often categorized as inert, preformulation studies can determine 
the infl uence of excipients on stability, bioavailability, and processability. Excipients 
are categorized into groups according to their main function, although some may 
be multifunctional, and examples of common excipients used in the manufacture of 
tablets and capsule are detailed in Table 3 . 
4.1.4.1 Diluents 
An inert substance is frequently added to increase the bulk of a tablet for processing 
and handling. The lower weight limit for formulation of a tablet is usually 50 mg. 
Ideally, diluents should be chemically inert, nonhygroscopic, and hydrophilic. Having 
an acceptable taste is important for oral formulations, and cost is always a signifi cant 
factor in excipient selection. 
Lactose is a common diluent in both tablets and capsules, and it fulfi ls most of 
these criteria but is unsuitable for those who are lactose intolerant. Various lactose 
grades are commercially available which have different physical properties such as 
particle size distribution and fl ow characteristics. This permits the selection of the 
most suitable material for a particular application. Usually, fi ne grades of lactose 
are used for preparation of tablets by wet granulation or when milling during processing 
is carried out, since the fi ne size permits better mixing with other formulation 
ingredients and facilitates more effective action of the binder [6] . 
Diluents for direct compression formulations are often subject to prior processing 
to improve fl owability and compression, for example, amorphous lactose, but 
this can contribute to reduced stability especially under high - humidity conditions 
when reversion to the crystalline form is more likely [6] . 
Microcrystalline cellulose (Avicel) is purifi ed partially depolymerized cellulose, 
prepared by treating . - cellulose with mineral acids. In addition to being used as a 
fi ller, it is also used as dry binder and disintegrant in tablet formulations. Depending 
on the preparation conditions, it can be produced with a variety of technical speci- 
fi cations depending on particle size and crystallinity. It is often used as an excipient 
in direct compression formulations but can also be incorporated as a diluent for 
tablets prepared by wet granulation, as a fi ller for capsules and for the production 
of spheres. 
TABLE 3 Excipients Used in Solid Dose Formulations 
Excipient Category Examples 
Fillers/diluents Lactose, sucrose, glucose, microcrystalline cellulose 
Binders Polyvinyl pyrrolidone, starch, gelatin, cellulose derivatives 
Lubricants Magnesium stearate, stearic acid, polyethylene glycol, sodium 
chloride 
Glidants Fine silica, talc, magnesium stearate 
Antiadherents Talc, cornstarch, sodium dodecylsulfate 
Disintegrants and 
superdisintegrants 
Starch, sodium starch glycollate, cross - linked polyvinyl pyrrolidone 
Colorants Iron oxide, natural pigments 
Flavor modifi ers Mannitol, aspartame 

Diluents, although commonly presumed inert, do have the ability to infl uence the 
stability or bioavailability of the dosage form. For example, dibasic calcium phosphate 
(both anhydrous and dihydrate forms) is the most common inorganic salt 
used as a fi ller – binder for direct compression. It is particularly useful in vitamin 
products as a source of both calcium and phosphorous. Milled material is typically 
used in wet - granulated or roller - compacted formulations. The coarse - grade material 
is typically used in direct compression formulations. It is insoluble in water, but its 
surface is alkaline and it is therefore incompatible with drugs sensitive to alkaline 
pH. Additionally, it may interfere with the absorption of tetracyclines [7] . 
4.1.4.2 Binders 
Binders (or adhesives) are added to formulations to promote cohesiveness within 
powders, thereby ensuring that the tablet remains intact after compression as well 
as improving the fl ow by forming granules. A binder should impart adequate cohesion 
without retarding disintegration or dissolution. Binders can be added either as 
a solution or as a dry powder. Binders added as dry powders are mixed with other 
powders prior to agglomeration, dissolving in water or solvent added during granulation, 
or added prior to compaction. Solution binders can be sprayed, poured, or 
mixed with the powder blend for agglomeration and are generally more effective, 
but further dry binder can be added prior to tableting. Starch, gelatin, and sugars 
are used along with gums, such as acacia and sodium alginate, and are used at concentrations 
between 2 and 10% w/w. Celluloses and polyvinyl pyrrolidone (PVP) 
are also utilized, often as dry binders. 
4.1.4.3 Lubricants 
Lubricants can reduce friction between the tablet and the die wall during compression 
and ejection by interposing an intermediate fi lm of low shear strength at the 
interface between the tablet and the die wall. The best lubricants are those with low 
shear strength but strong cohesive tendencies perpendicular to the line of shear [8] . 
The hydrophobic stearic acid and stearic acid salts, primarily magnesium stearate, 
are the most widely used and are included at concentrations less than 1% w/w in 
order to minimize any deleterious effects on disintegration or dissolution. They 
should be added after the disintegrant to avoid coating it and preferably at the fi nal 
stage prior to compression to ensure mixing time is kept to a minimum. Hydrophilic 
lubricants such as polyethylene glycols (PEGs) and lauryl sulfates can be used to 
redress the issues with dissolution but may not be as effi cient as their hydrophobic 
counterparts. 
4.1.4.4 Glidants and Antiadherents 
Like lubricants, glidants are fi ne powders and may be required for tablet compression 
at high production speeds to improve the fl ow properties of the material 
into the die or during initial compression stages. They are added in the dry state 
immediately prior to compression and, by virtue of their low adhesive potential, 
reduce the friction between particles. Colloidal silica is popular, as are starches 
and talc. 
EXCIPIENTS IN SOLID DOSE FORMULATIONS 241

242 SOLID DOSAGE FORMS 
Antiadherents can also be added to a formulation that is especially prone to 
sticking to the die surface (or picking). Water - insoluble lubricants such as magnesium 
stearate can be used as antiadherents, as can talc and starch. 
4.1.4.5 Disintegrants 
Disintegrants are added to a formulation to overcome the cohesive strength imparted 
during compression, thus facilitating break up of the formulation in the body and 
increasing the surface area for dissolution. They can be either intragranular, extragranular, 
or both, and there is still a lack of understanding concerning their precise 
mechanism of action. On contact, disintegrants can draw water into the tablet, swelling 
and forcing the tablet apart. Starch, a traditional and still widely used disintegrant, 
will swell when wet, although it has been reported that its disintegrant action 
could be due to capillary action [6] . Levels can be increased beyond the normal 5% 
w/w to 15 – 20% w/w if a rapid disintegration is required. Surfactants can also act as 
disintegrants promoting wetting of the formulation, and sodium lauryl sulfate can 
be combined with starch to increase effectiveness. 
Tablet disruption following production of carbon dioxide is another mechanism 
used to enhance disintegration. This uses a mixture of sodium bicarbonate and a 
weak acid such as citric acid or tartaric acid and is exploited for effervescent 
formulations. 
4.1.4.6 Superdisintegrants 
Compared to the more traditional starch, newer disintegrants are effective at much 
lower levels and comprise three groups: modifi ed starches, modifi ed cellulose, and 
cross - linked povidone. Their likely mechanism of action is a combination of proposed 
theories including water wicking, swelling, deformation recovery, repulsion, 
and heat of wetting [9] . Superdisintegrants are so called because of the relatively 
low levels required (2 – 4% w/w). Sodium starch glycollate (Primojel, Explotab) is 
made by cross - linking potato starch and can swell up to 12 - fold in less than 30 s. 
Crospovidone is completely insoluble in water, although it rapidly disperses and 
swells in water, but does not gel even after prolonged exposure. It rapidly exhibits 
high capillary activity and pronounced hydration capacity with little tendency to 
form gels and has a greater surface area – volume ratio compared to other disintegrants. 
Micronized versions are available to improve uniformity of mix. Croscarmellose 
sodium, a cross - linked polymer of carboxymethyl cellulose sodium is also 
insoluble in water, although it rapidly swells to 4 – 8 times its original volume on 
contact with water [6] . 
4.1.4.7 Added Functionality Excipients 
Adverse physiochemical and mechanical properties of new chemical entities prove 
challenging for formulation development. There is an increasing demand for faster 
and more effi cient production processes. Also, biotechnological developments and 
various emerging protein - based therapies are broadening the defi nition for excipient 
products. Although the description of excipients from inactive ingredients is shifting 
toward functionally active materials and will continue to grow in this area, the intro

duction of improved versions of long - existing excipients is probably the more successful 
development. New single - component and coprocessed products have been 
introduced, for example, fi ller – binders. In addition, there have been advances in the 
understanding of how such substances act and hence how they can be optimally 
designed. Excipients for use in direct compression product forms or physically or 
chemically modifi ed excipients used in relatively new drug delivery systems, such as 
patches or inhalation systems, are examples of these developments. 
4.1.4.8 Colorants 
Colorants are frequently used in uncoated tablets, coated tablets, and hard and soft 
gelatin capsules. They can mask color changes in the formulation and are used to 
provide uniqueness and identity to a commercial product. Concerns over the safety 
of coloring agents in formulations generally arise from adverse effects in food substances. 
Colorants are therefore subject to regulations not associated with other 
pharmaceutical excipients. Legislation specifi es which colorants may be used in 
medicinal products and also provides for purity specifi cations. The number of permitted 
colors has decreased in recent years, and a list of approved colorants allowed 
by regulatory bodies can vary from country to country. 
Colorants can be divided into water - soluble dyes and water - insoluble pigments. 
Some of the insoluble colors or pigments can also provide opacity to tablet coatings 
or gelatin shells, which can promote stability of light - sensitive active materials. Pigments 
such as the iron oxides, titanium dioxide, and some of the aluminum lakes 
are especially useful for this purpose. 
Water - soluble dyes are usually incorporated within the granulation process to 
ensure even distribution throughout the formulation, but there can be an uneven 
distribution due to migration of the dye during drying processes. Therefore, water - 
soluble dyes can also be adsorbed into a carrier such as starch or lactose and dry 
blended prior to the fi nal mix. Water - insoluble pigments are more popular in direct 
compression and are dry blended with the other ingredients. 
Lakes are largely water - insoluble forms of common synthetic water - soluble dyes 
and are prepared by adsorbing the sodium or potassium salt of a dye onto a very 
fi ne substrate of hydrated alumina, followed by treatment with a further soluble 
aluminum salt. The lake is then purifi ed and dried. Lakes are frequently used in 
coloring tablet coatings since they are more stable and have greater opacity than a 
water - soluble dye [6] . 
4.1.4.9 Interactions and Safety of Excipients 
Because there is such a wide selection available, rational choice of the necessary 
excipients and their concentration is required. Consideration must also be given to 
cost, reliability, availability, and international acceptability. Although generally considered 
inert, formulation incompatibility of excipients is also necessary. Lactose, 
for example, can react with primary and secondary amines via its aldehyde group 
by Maillaird condensation reaction [6] , and calcium carbonate is incompatible with 
acids due to acid – base chemical reaction and with tetracyclines due to complexation. 
Additionally, excipients can contribute to the instability of the active substance 
through moisture distribution. 
EXCIPIENTS IN SOLID DOSE FORMULATIONS 243

244 SOLID DOSAGE FORMS 
Despite the importance of drug – excipient compatibility testing, there is no generally 
accepted method available for this purpose. After identifi cation of any major 
known incompatibilities, a compatibility screen needs to be proposed. Issues such 
as sample preparation, storage conditions, and methods of analysis should be 
addressed and factorial design applied to reduce the number if tests required. 
Drug – excipient compatibility studies can be performed with minimal amounts of 
materials. Usually, small amounts of each material are weighed into a glass vial, in 
a ratio representative of the expected ratio in the formulation. The vials can be 
sealed as is or with additional water, either in an air environment or oxygen - free 
(nitrogen head space) environment, and stored in the presence or absence of ambient 
light, at various temperatures. Factorial or partial factorial design experiments can 
be set up to determine important binary and multiple component interaction factors. 
This information helps determine which excipients should be avoided and whether 
oxidation or light instability in the formulation is a consideration. Controls consisting 
of the active pharmaceutical ingredient (API) alone in the various conditions 
also should be run to determine whether the API is susceptible alone or must have 
the mediating excipient or water additives for instability. 
4.1.5 COATED TABLETS 
Tablets are often coated to protect the drug from the external environment, to mask 
bitter tastes, add mechanical strength, or to enhance ease of swallowing. A coating 
can also be used for aesthetic or commercial purposes, improving product appearance 
and identity. 
4.1.5.1 Sugar - Coated Tablets 
Sugar coating can be benefi cial in masking taste, odors, and colors. It is useful in 
protecting against oxidation, and sugar coating was once very common due to its 
aesthetic results and cheapness of materials. Use has declined in recent years due 
to the complexity of the process and skills required, but advances in technology 
have led to a resurgence in popularity. Typical excipients used are sucrose (although 
this can be substituted with low - calorie alternatives), fi llers, fl avors, fi lm formers, 
colorants, and surfactants. It is usually carried out in tumbling coating pans and 
comprises several stages. 
The fi rst sealing stage uses shellac or cellulose acetate phthalate, for example, to 
prevent moisture from reaching the tablet core. This has to be kept to a minimum 
to prevent impairment of drug release. The subcoating is an adhesive coat of gum 
(such as acacia or gelatin) and sucrose used to round off the edges, and the tablets 
can be dusted with substances such as kaolin or calcium carbonate to harden the 
coating. A smoothing coat is built up in layers using 70% v/v sucrose syrup and often 
opacifi ers such as titanium dioxide, and the tablets are dried between each application. 
A colorant is added to the fi nal few layers and followed with a fi nal polishing 
step which can make further embossing diffi cult. The coating is relatively brittle, 
prone to chipping or cracking, and there is a substantial increase in weight, up to 
50%, and size of the product. 

HARD AND SOFT GELATIN CAPSULES 245 
4.1.5.2 Compression Coating and Layered Tablets 
A coating can be applied by compression using specially designed tablet presses. 
The same process can be used to produce layered tablets which can comprise two 
or even three layers if complete separation of the ingredients is required. This 
process is used when physical separation of ingredients is desired due to incompatibility 
or to produce a repeat - action product. The formulation can also be designed 
to provide an immediate and a slow - release component. Release rates can be 
controlled by modifi cation of the geometry, the composition of the core, and the 
inclusion of a membrane layer. 
The technique involves using a preliminary compression step to produce a relatively 
soft tablet core which is then placed in a large die containing coating material. 
Further coating material is added and the content compressed. A similar light compression 
is used for the production of layers and a fi nal main compression step used 
to bind the layers together. 
4.1.5.3 Film - Coated Tablets 
Film coating, although most often applied to tablets, can also be used to coat other 
formulations including capsules. Film coating imparts the same general characteristics 
as sugar coating but weight gain is signifi cantly less (typically up to 5%), it is 
easier to automate, and it has capacity to include organic solvents if required. The 
main methods involved are modifi ed conventional coating pans, side - vented pans, 
and fl uid - bed coating. Celluloses are often used as fi lm - forming polymers, as detailed 
in Table 4 , and usually require addition of a compatible plasticizer as glass transition 
temperatures are higher than the temperatures used in the process. Polyethylene 
glycol, propylene glycol, and glycerol are commonly used, and colorants and opaci- 
fi ers can also be added to the coating solution. Specialist coatings such as Opadry 
fx and Opaglos 2 can be used to give a high gloss fi nish to improve brand identity 
and consumer recognition. 
4.1.5.4 Tablet Wrapping or Enrobing 1 
Recent innovations in tablet coating include the use of gelatin and non - animal - 
derived coatings for tablets that require formulation of a pre - formed fi lm that is 
then used to encapsulate the product (e.g., Banner ’ s Sofl et Gelcaps or Bioprogress ’ 
Nrobe technology). The coated formulations are tamper evident and can be designed 
with different colors for branding purposes. They are reported to be preferred by 
patients due to their ease of swallowing and superior taste - and odor - masking properties. 
An alternative is the Press - fi t Geltabs system, which uses a high - gloss gelatin 
capsule shell to encapsulate a denser caplet formulation. 
4.1.6 HARD AND SOFT GELATIN CAPSULES 
Capsules are solid oral dosage forms in which the drug is enclosed within a hard or 
soft shell. The shell is normally made from gelatin and results in a simple, easy - to - 
swallow formulation with no requirement for a further coating step. They can be 
1 See http://www.banpharm.com/technologiesSofl etGelcap.cfm and http://www.fmcmagenta.com/NRobe/ 
tabid/145/Default.aspx . 

246 SOLID DOSAGE FORMS 
TABLE 4 Polymers Commonly Used in Film Coating of Tablets 
Polymer Soluble in Description 
Methylcellulose (MC) Cold water, GI fl uids, 
and organic solvents 
Low - viscosity grades best for aqueous 
fi lms 
Ethylcellulose (EC) Organic solvents and 
GI fl uids (insoluble 
in water) 
Used in combination with water - 
soluble agents for immediate 
release 
Hydroxyethyl cellulose 
(HEC) 
Water and GI fl uids Similar to MC with clear solutions 
Hydroxypropyl 
cellulose (HPC) 
Cold water, GI fl uids, 
and polar solvents 
Results in a tacky coat and used in 
combination to promote adhesion 
Hydroxypropylmethyl 
cellulose (HPMC) 
Cold water, GI fl uids, 
and alcohols 
Excellent fi lm former, low - viscosity 
grades best 
Sodium carboxymethyl 
cellulose 
Water and polar 
solvents 
Cannot be used if presence of 
moisture is a problem 
Methylhydroxyethyl 
cellulose (MHEC) 
Water and GI fl uids Similar to HPMC but less soluble in 
organic solvents 
Povidone (PVP) Water, GI fl uids, 
alcohol, and 
isoproplyl alcohol 
(IPA) 
Can lead to tackiness during drying, 
often brittle and hygroscopic 
PEGs Water, GI fl uids, some 
organic solvents 
High molecular weights best for fi lm 
forming and low molecular weights 
used as plasticizer; can be waxy 
Enteric coatings such as 
poly(methacrylates) 
or cellulose acetate 
phthalate 
Soluble at elevated 
pHs 
Used for delayed - release formulations 
Source : Adapted from refs. 5 and 10 . 
either hard or soft depending on the nature of the capsule shell, with soft capsules 
possessing a fl exible, plasticized gelatin fi lm. Hard gelatin capsules are usually rigid 
two - piece capsules that are manufactured in one procedure and packed in another 
totally separate operation, whereas the formulation of soft gelatin capsules is more 
complex but all steps are integrated. 
There is a growing interest in using non - animal - derived products for formulation 
of the capsule shells to address cultural, religious, and dietary requirements. HPMC 
(e.g., V - caps, Quali - VC, Vegicaps) and pullulan shells (NPCaps) and starch are 
alternatives. 
4.1.6.1 Hard - Shell Gelatin Capsules 
Although the challenges of powder blending, homogeneity, and lubcricity exist for 
capsules as for tablets, they are generally perceived to be a more fl exible formulation 
as there is no requirement for the powders to form a robust compact. This 
means that they may also be more suitable for delivery of granular and beadlike 
formulations, fragile formulations that could be crushed by the normal compaction 
step. They are commonly employed in clinical trials due to the relative ease of blinding 
and are useful for taste masking. 

HARD AND SOFT GELATIN CAPSULES 247 
Capsules are usually more expensive dosage forms than an equivalent tablet 
formulation due to the increased cost of the shells and the slower production rates. 
Even with modern fi lling equipment, the fi lling speeds of capsule machines are much 
slower than tablet presses. However, increased costs can be offset by avoiding a 
granulation step. Capsules, although smoother and easy to swallow, also tend to be 
larger than corresponding tablet formulations, potentially leading to retention in 
the esophagus. Humidity needs to be considered during manufacture and storage, 
with moisture leading to stickiness and desiccation causing brittleness. Cross - linking 
of gelatin in the formulation can also lead to dissolution and bioavailability 
concerns. 
Capsule excipients are similar to those required for formulation of tablets and 
include diluents, binders, disintegrants, surfactants, glidants, lubricants, and dyes or 
colorants. The development of a capsule formulation follows the same principles as 
tablet development, and consideration should be given to the same BCS issues. The 
powder for encapsulation can comprise simple blends of excipients or granules 
prepared by dry granulation or wet granulation. There is a reduced requirement for 
compressibility, and often the fl ow properties are not as critical as in an equivalent 
tablet formulation. The degree of compressibility required is the major difference, 
and capsules can therefore be employed when the active ingredient does not possess 
suitable compression characteristics. 
4.1.6.2 Manufacture of Hard Gelatin Shells 
Gelatin is a generic term for a mixture of purifi ed protein fractions obtained either 
by partial acid hydrolysis (type A gelatin) or by partial alkaline hydrolysis (type B 
gelatin) of animal collagen. Type A normally originates from porcine skin while B 
is usually derived from animal bones, and they have different isoelectric points 
(7.0 – 9.0 and 4.8 – 5.0, respectively) [6] . The protein fractions consist almost entirely 
of amino acids joined together by amide linkages to form linear polymers, varying 
in molecular weight from 15,000 to 250,000. Gelatin can comprise a mixture of both 
types in order to optimize desired characteristics, with bone gelatin imparting fi rmness 
while porcine skin gelatin provides plasticity. Gelatin Bloom strength is measured 
in a Bloom gelometer, which determines the weight in grams required to 
depress a standard plunger in a 6.67% w/w gel under standard conditions. Bloom 
strength and viscosity are the major properties of interest for formulation of capsules, 
and Bloom strength of 215 – 280 is used in capsule manufacture. 
Gelatin is commonly used in foods and has global regulatory acceptability, is a 
good fi lm former, is water soluble, and generally dissolves rapidly within the body 
without imparting any lag effect on dissolution. Gelatin capsules are strong and 
robust enough to withstand the mechanical stresses involved in the automated fi lling 
and packaging procedures. 
In addition to gelatin, the shells may contain colorants, opacifi ers, and preservatives 
(often parabens esters). There are eight standard capsule sizes, and the largest 
capsule size considered suitable for oral use is size 0 (Table 5 ). 
To manufacture the shells, pairs of molds, for the body and the cap, are dipped 
into an aqueous gelatin solution (25 – 30% w/w), which is maintained at about 50 ° C 
in a jacketed heating pan. As the pins are withdrawn, they are rotated to distribute 
the gelatin evenly and blasted with cool air to set the fi lm. Drying is carried out by 

248 SOLID DOSAGE FORMS 
passing dry air over the shell as heating temperatures are limited due to the low 
melting point of gelatin. The two parts are removed from the pins, trimmed, and 
joined using a prelock mechanism. The external diameter of the body is usually 
wider at the open end than the internal diameter of the cap to ensure a tight fi t. 
They can be made self - locking by forming indentations or grooves on the inside of 
both parts so that when they are engaged, a positive interlock is formed (e.g., 
Posilok, Conicap, Loxit). 
Alternatively, they may be hermetically sealed using a band of gelatin around 
the seam between the body and the cap (Qualicaps). This can be applied without 
the application of heat and provide a tamper - evident seal. LEMS (liquid encapsulation 
microspray sealing) used in Licaps is a more elegant seal in which sealing fl uid 
(water and ethanol) is sprayed onto the joint between the cap and body of the 
capsule. This lowers the melting point of gelatin in the wetted area. Gentle heat is 
then applied which fuses the cap to the body of the Licaps capsule. The moisture 
content of manufactured shells is 15 – 18% w/w and levels below 13% will result in 
problems with the capsule fi lling machinery. Therefore, capsules are stored and fi lled 
in areas where relative humidity is controlled to between 30 and 50%. 
4.1.6.3 Hard - Gelatin Capsule Filling 
The fi lling material must be compatible with the gelatin shell and, therefore, deliquescent 
or hygroscopic materials cannot be used. Conversely, due the moisture 
content in the capsule shells, they cannot be used for moisture - sensitive drugs. All 
ingredients need to be free of even trace amounts of formaldehyde to minimize 
cross - linking of gelatin. 
Powders and granules are the most common fi lling materials for hard - shell gelatin 
capsules, although pellets, tablets, pastes, oily liquids, and nonaqueous solutions and 
suspensions have been used. Filling machines are differentiated by the way they 
measure the dose of material and range in capacity from bench - top to high - output, 
industrial, fully automated machines. Those that rely on the volume of the shell are 
known as capsule dependent, whereas capsule - independent forms measure the 
quantity to be fi lled in a separate operation. The simplest dependent method of 
fi lling is leveling where powder is transferred directly from a hopper to the capsule 
TABLE 5 Capsule Size and Corresponding Volume or 
Weight of Fill 
Size Volume (mL) Fill weight a (g) 
000 1.37 1.096 
00 0.95 0.760 
0 0.68 0.544 
1 0.50 0.400 
2 0.37 0.296 
3 0.30 0.240 
4 0.21 0.168 
5 0.13 0.104 
Source : Adapted from http://capsugel.onlinemore.info/download/ 
BAS192 - 2002.pdf . 
a Assumes a powder density of 0.8 g/cm 3 . 

HARD AND SOFT GELATIN CAPSULES 249 
body, aided by a revolving auger or vibration. Additional powder can be added to 
fi ll the space arising, and the fi ll weight depends on the bulk density of the powder 
and the degree of tamping applied. 
Most automated machinery is of the independent type and compresses a controlled 
amount of powder using a low compression force (typically 50 – 200 N ) to 
form a plug. Most are piston - tamp fi llers and are dosator or dosing disk machines. 
The powder is passed over a dosing plate containing cavities slightly smaller than 
the capsule diameter, and powder that falls into the holes is tamped by a pin to form 
a plug. This can be repeated until the cavity is full and the plugs (or slugs) are ejected 
into the capsule shells. The minimum force required to form a plug should be used 
to reduce slowing of subsequent dissolution. 
In the dosator method, the plug is formed within a tube with a movable piston 
that controls the dosing volume and applies the force to form the plug. The dose is 
controlled by the dimensions of the dosator, the position of the dosator in the 
powder bed, and the height of the powder bed. Fundamental powder properties to 
ensure even fi lling are good powder fl ow, lubricity, and compressibility. The auger 
or screw method, now largely surpassed, uses a revolving archimedian screw to feed 
powder into the capsule shell. 
A liquid fi ll can be useful when manufacturing small batches if limited quantities 
of API are available. Liquid fi lls also offer improved content uniformity for potent, 
low - dose compounds and can reduce dust - related problems arising with toxic compounds. 
Two types of liquid can be fi lled into hard gelatin capsules: nonaqueous 
solutions and suspensions or formulations that become liquid on application of heat 
or shear stress. These require hoppers with heating or stirring systems. For those 
formulations that are liquid at room temperature, the capsule shells need to be 
sealed after fi lling to prevent leakage of the contents and sticking of the shells. It is 
essential to ensure the liquid is compatible with the shell (Table 6 ). 
4.1.6.4 Soft - Gelatin Capsules 
Soft gelatin capsules are hermetically sealed one - piece capsules containing a liquid 
or a semisolid fi ll. Like liquid - fi lled hard capsules, although the drug is presented in 
a liquid formulation, it is enclosed within a solid, thus combining the attributes 
TABLE 6 Liquid Excipients Compatible with Hard 
Gelatin Capsules 
Peanut oil Paraffi n oil 
Hydrogenated peanut oil Cetyl alchohol 
Castor oil Cetostearyl alcohol 
Hydrogenated castor oil Stearyl alcohol 
Fractionated coconut oil Stearic acid 
Corn oil Beeswax 
Olive oil Silica dioxide 
Hydrogenated vegetable oil Polyethylene glycols 
Silicone oil Macrogol glycerides 
Soya oil Poloxamers 
Source : Adapted from http://www.capsugel.com/products/licaps_ 
oil_chart.php . 

250 SOLID DOSAGE FORMS 
of both. Soft gelatin capsules (softgels) offer a number of advantages including 
improved bioavailability, as the drug is presented in a solubilized form, and enhanced 
drug stability. Consumer preference regarding ease of swallowing, convenience, and 
taste can improve compliance, and they offer opportunities for product differentiation 
via color, shape, and size and product line extension. The softgels can be enteric 
coated for delayed release. They are popular for pharmaceuticals, cosmetics, and 
nutritional products, but highly water - soluble drugs and aldehydes are not suitable 
for encapsulation in softgels. Formulations are tamper evident and can be used for 
highly potent or toxic drugs. However, they do require specialist manufacture and 
incur high production costs. 
4.1.6.5 Manufacture of Soft Gelatin Capsules 
The shell is primarily composed of gelatin, plasticizer, and water (30 – 40% wet gel), 
and the fi ll can be a solution or suspension, liquid, or semisolid. The size of a softgel 
represents its nominal capacity in minims, for example, a 30 oval softgel can accommodate 
30 minims (or 1.848 cm 3 ). Glycerol is the major plasticizer used, although 
sorbitol and propylene glycol can also be used. Other excipients are dyes, pigments, 
preservatives, and fl avors. Up to 5% sugar can be added to give a chewable quality. 
Most softgels are manufactured by the process developed by Scherer [11] . The 
glycerol – gelatin solution is heated and pumped onto two chilled drums to form two 
separate ribbons (usually 0.02 – 0.04 in. thick) which form each half of the softgel. 
The ribbons are lubricated and fed into the fi lling machine, forcing the gelatin to 
adopt the contours of the die. The fi ll is manufactured in a separate process and 
pumped in, and the softgels are sealed by the application of heat and pressure. Once 
cut from the ribbon, they are tumble - dried and conditioned at 20% relative 
humidity. 
Fill solvents are selected based on a balance between adequate solubility of the 
drug and physical stability. Water - miscible solvents such as low - molecular - weight 
PEGs, polysorbates, and small amounts of propylene glycol, ethanol, and glycerin 
can be used. Water - immiscible solvents include vegetable and aromatic oils, aliphatic, 
aromatic, and chlorinated hydrocarbons, ethers, esters, and some alcohols. 
Emulsions, liquids with extremes of pH ( < 2.5 and > 7.5), and volatile components 
can cause problems with stability, and drugs that do not have adequate stability in 
the solvents can be formulated as suspensions. In these instances, the particle size 
needs to be carefully controlled and surfactants can be added to promote wetting. 
Vegicaps soft capsules from Cardinal Health are an alternative to traditional 
softgels, containing carageenan and hydroxyproyl starch. As with traditional soft 
gelatin capsules, the most important packaging and storage criterion is for adequate 
protection against extremes of relative humidity. The extent of protection required 
also depends on the fi ll formulation and on the anticipated storage conditions. 
4.1.6.6 Dissolution Testing of Capsules 
In general, capsule dosage forms tend to fl oat during dissolution testing with the 
paddle method. In such cases, it is recommended that a few turns of a wire helix 
around the capsule be used [12] . Inclusion of enzymes in the dissolution media must 
be considered on case - by - case basis. A Gelatin Capsule Working Group (including 

participants from the FDA, industry, and the USP) was formed to assess the noncompliance 
of certain gelatin capsule products with the required dissolution speci- 
fi cations and the potential implications on bioavailability [13] . The working group 
recommended the addition of a second tier to the standard USP and new drug and 
abbreviated new drug applications (NDA/ANDA) dissolution tests: the incorporation 
of enzyme (pepsin with simulated gastric fl uid and pancreatin with simulated 
intestinal fl uid) into the dissolution medium. If the drug product fails the fi rst tier 
but passes the second tier, the product ’ s performance is acceptable. The two - tier 
dissolution test is appropriate for all gelatin capsule and gelatin - coated tablets and 
the phenomenon may have little signifi cance in vivo. 
4.1.7 EFFERVESCENT TABLETS 
Effervescence is the reaction in water of acids and bases to produce carbon dioxide, 
and effervescent tablets are dissolved or dispersed in water before administration. 
Advantages of effervescent formulations over conventional formulations are that 
the drug is usually already in solution prior to ingestion and can therefore have a 
faster onset of action. Although the solution may become diluted in the GI tract, 
any precipitation should be as fi ne particles that can be readily redissolved. Variability 
in absorption can also be reduced. Formulations can be made more palatable 
and there can be improved tolerance after ingestion. Thus, the types of drugs suited 
to this formulation method are those that are diffi cult to digest or are irritant to 
mucosa. Analgesics such as paracetamol and aspirin and vitamins are common 
effervescent formulations. The inclusion of buffering agents can aid stability of pH - 
sensitive drugs. There is also the opportunity to extend market share and to deliver 
large doses of medication. 
Effervescents comprise a soluble organic acid and an alkali metal carbonate salt. 
Citric acid is most commonly used for its fl avor - enhancing properties. Malic acid 
imparts a smoother after taste and fumaric, ascorbic, adipic, and tartaric acids are 
less commonly used [14] . Sodium bicarbonate is the most common alkali, but potassium 
bicarbonate can be used if sodium levels are a potential issue with the formulation. 
Both sodium and potassium carbonate can also be employed. Other excipients 
include water - soluble binders such as dextrose or lactose, and binder levels are kept 
to a minimum to avoid retardation of disintegration. All ingredients must be anhydrous 
to prevent the components within the formulation reacting with each other 
during storage. 
Lubricants such as magnesium stearate are not used as their aqueous insolubility 
leads to cloudy solutions and extended disintegration times. Spray - dried leucine and 
PEG are water - soluble alternatives [15, 16] . Both artifi cial and natural sweeteners 
are used and an additional water - soluble fl avoring agent may also be required. If a 
surfactant is added to enhance wetting and dissolution, the addition of an antifoaming 
agent may also be considered [17] . 
4.1.7.1 Manufacture of Effervescent Tablets 
Essentially, effervescent formulations are produced in the same way as conventional 
tablets, although due to the hygroscopicity and potential onset of the effervescence 
EFFERVESCENT TABLETS 251

252 SOLID DOSAGE FORMS 
reaction in the presence of water, environmental control of relative humidity and 
water levels is of major importance during manufacture. A maximum of 25% relative 
humidity (RH) at 25 ° C is required. Closed material - handling systems can be 
used or open systems with minimum moisture content in the ventilating air. 
A dry method of granulation is preferred as no liquid is involved but may not 
always be possible. Wet granulation can be carried out under carefully controlled 
conditions using two separate granulators for the alkaline and acid components. 
Water can be added at 0.1 – 1.0% w/w, and it initiates a preeffervescent reaction. The 
cycle is stopped by drying, usually by transfer into a preheated fl uidized - bed dryer. 
Fluid - bed spray granulation is a process wherein granulation and drying are simultaneous 
and can be useful for effervescent formulations. Water (or a binder solution) is 
sprayed onto the mixture, which is suspended in a stream of hot, dry air. Organic solvents 
can also be employed for granulation avoiding the need for water and are useful 
for heat - labile formulations, although complex handling equipment is required. 
Effervescent formulations must contain less than 0.3% w/w water and are often 
quite large. Sticking due to insuffi cient lubrication can be overcome by adaptation 
of punches for external lubrication or using fl at - faced punches with disks of poly 
(tetrafl uoroethylene) (PTFE). Poor lubrication can also be the cause of poor fl ow 
characteristics, and this can be addressed by using a constant level powder feed 
system. The tablets should be stored in tightly closed containers or moisture - proof 
packs. In tube arrangements, dry air is added prior to sealing and desiccants to 
reduce enclosed moisture levels once the pack has been opened. Foil packaging 
should be heavy gauge to minimize risk of holes, and the surrounding pocket should 
be large enough to hold the tablets but minimize inclusion of air. 
In - process quality control is of major importance for these formulations as are 
stability testing and stress testing of packaging. Tablet disintegration and dissolution 
are of prime importance, and disintegration should be carried out using representative 
conditions. Hardness and friability are also important as these large tablets tend 
to chip easily. Common areas for problems are that the packaging permits entry of 
water, the seal is compromised or that the excipients can react with each other. 
4.1.8 LOZENGES 
Lozenges are tablets that dissolve or disintegrate slowly in the mouth to release 
drug into the saliva. They are easy to administer to pediatric and geriatric patients 
and are useful for extending drug form retention within the oral cavity. They usually 
contain one or more ingredient in a sweetened fl avored base. Drug delivery can be 
either for local administration in the mouth, such as anaesthetics, antiseptics, and 
antimicrobials or for systemic effects if the drug is well absorbed through the buccal 
lining or is swallowed. More traditional drugs used in this dosage form include 
phenol, sodium phenolate, benzocaine, and cetylpyridinium chloride. Decongestants 
and antitussives are in many over - the - counter (OTC) lozenge formulations, and 
there are also lozenges that contain nicotine (as bitartrate salt or as nicotine polacrilex 
resin), fl urbiprofen (Strefen), or mucin for treatment of dry mouth (A.S Saliva 
Orthana). 
Lozenges can be made by molding or by compression at high pressures, often 
following wet granulation, resulting in a mechanically strong tablet that can dissolve 

in the mouth. Compressed lozenges (or troches) differ from conventional tablets in 
that they are nonporous and do not contain disintegrant. As the formulation is 
designed to release drug slowly in the mouth, it must have a pleasant taste, smoothness, 
and mouth feel. The choice of binder, fi ller, color, and fl avor is therefore most 
important. The binder is particularly important in ensuring retardation of dissolution 
and pleasant mouth feel. Suitable binders include gelatin, guar gum, and acacia 
gum. Sugars such as sucrose, dextrose, and mannitol are preferred to lactose, and 
xylitol is often included in sugar - free formulations. In order to ensure adequate 
sweetness and taste masking, artifi cial sweeteners including aspartame, saccharin, 
and sucralose are also included subject to regulatory guidelines. 
Other variations include hard - candy - type and soft or chewable lozenges. Most 
hard - candy - type lozenges contain sugar, corn syrup, acidulant, colorant, and fl avors. 
They are made by heating sugars and other ingredients together and then pouring 
the mixture into a mold. Corn syrup combined with sucrose and dextrose can form 
an amorphous glass suitable for such formulations [18] . Colorants can be added to 
enhance product appearance or to mask products of degradation. Stability and 
compatibility with the drug must be established along with the other excipients. 
Flavors tend to be complex entities, and stability or compatibility can pose major 
formulation challenges. Acidulants such as citric and tartaric acids are often added 
to enhance fl avors, thus lowering pH of the formulation as low as 2.5 – 3.0. Addition 
of bases such as calcium carbonate, sodium bicarbonate, and magnesium trisilicate 
is common to increase pH and enhance drug stability. For example, in vivo and in 
vitro studies confi rmed that the pH of the dissolved lozenge solution was the single 
most infl uential, readily adjustable formulation parameter infl uencing the activity 
of cetylpyridinium chloride activity in candy - based lozenges [19] . The dosage form 
needs a low moisture content (0.5 – 1.5% w/w), so water is evaporated off by boiling 
the sugar mixture during the compounding process, thus limiting the process to 
nonlabile drugs, and the manufacture requires specialized candy processing facilities. 
Packaging also needs to protect the formulated product from moisture and 
ranges from individual bunch wrapping to foil wraps. 
4.1.8.1 Chewable Lozenges 
Chewable lozenges are popular with the pediatric population since they are “ gummy - 
type ” lozenges. Most formulations are based on a modifi ed suppository formula consisting 
of glycerin, gelatin, and water. These lozenges are often highly fruit - 
fl avored and may have a slightly acidic taste to cover the acrid taste associated with 
glycerin. Soft lozenges typically comprise ingredients such as PEG 1000 or 1450, or a 
sugar – acacia base. Silica gel can be added to prevent sedimentation, and again this 
dosage form requires fl avors and sweeteners to aid compliance. Soft lozenges tend to 
dissolve faster than gelatin bases and can be used if taste masking is not effective. 
4.1.9 CHEWABLE TABLETS 
Chewable tablets are designed to be mechanically disintegrated in the mouth. 
Potential advantages of chewable tablets are mainly concerning patient convenience 
and acceptance, although enhanced bioavailability is also claimed. This can be due 
CHEWABLE TABLETS 253

254 SOLID DOSAGE FORMS 
to a rapid onset of action as disintegrate is more rapid and complete compared to 
standard formulations that must disintegrate in the GI tract. The dosage form is an 
appealing alternative for pediatric and geriatric consumers. Chewable tablets also 
offer convenience for consumers, avoiding the necessity of coadministration with 
water, and creation of palatable formulations can increase compliance. Antacids and 
pediatric vitamins are often formulated as chewable tablets, but other formulations 
include antihistamines (Zyrtec), antimotility agents (Imodium Plus) and antiepileptic 
agents (Epanutin Infatabs), antibiotics (Augmentin Chewable), asthma treatments 
(Singulair), and analgesics (Motrin). 
Constraints with these systems are that many pharmaceutical actives have an 
unpleasant bitter taste that can actually reduce compliance among patients. Iron 
salts, for example, can impart a rusty taste, and some antihistamines such as promethazine 
HCl can have a bitter aftertaste. As such, active formulations require 
very effective taste - masking strategies to provide acceptable patient tolerance and 
to ensure patient adherence to their pharmaceutical regimen. 
Formulation factors governing design are similar to standard formulations (e.g., 
compactability, fl ow, etc.), but disintegrants are not included. Organoleptic properties 
are a major concern, especially in the design of products for children, and usage 
has been limited as formulators have encountered diffi culties in achieving satisfactory 
sensory characteristics. 
Certain diluents are benefi cial in the formulation of chewable tablets by compression 
such as mannitol, lactose, sucrose, and sorbitol. They can aid disintegration upon 
chewing and can help with acceptable taste and mouth feel. Mannitol, for example, 
can impart a cooling or soothing sensation. Specialist excipients with improved 
sensory components such as mouth feel and lack of grittiness have been developed 
for formulation of chewable tablets. For example, Avicel CE - 15 [a combination of 
microcrystalline cellulose (MCC) and guar gum] can reduce grittiness, leading to a 
creamier mouth feel and improved overall compatibility. 
Citric acid, grape, raspberry, lemon, and cherry fl avors are often used in chewable 
tablets and lozenges (Table 7 ). Flavoring agents are commonly volatile oils, and they 
can be dissolved in alcohol and then sprayed onto another excipient or granules. 
They are usually added immediately prior to compression to avoid loss due to their 
volatile nature. Dry fl avors have advantages in terms of stability and ease of handling 
and are formed by emulsifi cation of the fl avor into an aqueous solution of a 
carrier followed by drying, encapsulating the fl avor within the carrier. This is useful 
if the agent is prone to oxidation. Common carrier substances are acacia gum, starch, 
and maltodextrin. Sweeteners such as aspartame can also be added. Low - calorie 
and non - sugar - based excipients may present a marketing advantage. 
Issues of taste masking for chewable formulations may be addressed by coating 
in wet granulation. The granulating/coating agent should form a fl exible rather than 
TABLE 7 Flavor Groups for Taste Types 
Sweet Vanilla, grape, maple, honey 
Sour Citrus, raspberry, anise 
Salty Mixed fruit, mixed citrus, butterscotch, maple 
Bitter Licorice, coffee, mint, cherry, grapefruit 
Metallic Grape, lemon, lime 
Source : From ref. 18 . 

brittle fi lm, have no unpleasant taste of its own, not interfere with dissolution, and 
be insoluble in saliva. Microencapsulation for taste masking can be achieved by 
phase separation or coacervation and may also impart stability. The same taste - 
masking technologies may be used to encapsulate drugs for formulation into 
chewable, softchew, and fast dissolving dosage forms. Coating materials include 
carboxymethylcellulose, polyvinyl alcohol (PVA), and ethylcellulose. Xylitol is the 
sweetest sugar alcohol, and it has a high negative heat of solution, making it a good 
candidate as an excipient for chewable tablets. There are many types of compressible 
sugars today, and most of them are composed of sucrose granulated with small 
amounts of modifi ed dextrins in order to make the sucrose more compressible [20] . 
Modifi cations to sugar - based excipients such as spray - dried crystalline maltose and 
directly compressible sucrose (95% sucrose and 5% sorbitol) to facilitate direct 
compression are also aiding development in this area [21] . 
4.1.9.1 Testing of Chewable Tablets 
Dissolution testing for chewable tablets should be the same as that used for regular 
tablets [22] . This is because patients could swallow the dosage form without adequate 
chewing, in which case the drug would still need to be released to ensure 
the desired pharmacological action. However, as outlined, chewable tablets will 
typically have different excipients than standard formulations, including agents to 
either mask or add fl avor, and may undergo a different manufacturing process. 
Where applicable, test conditions would preferably be the same as used for nonchewable 
tablets of the same active pharmaceutical ingredient, but because of the 
nondisintegrating nature of the dosage form, it may be necessary to alter test 
conditions (e.g., increase the agitation rate) and specifi cations (e.g., increase the 
test duration). The reciprocating cylinder (USP apparatus 3) with the addition of 
glass beads may also provide more intensive agitation for in vitro dissolution 
testing of chewable tablets. As another option, mechanical breaking of chewable 
tablets prior to exposing the specimen to dissolution testing could be considered. 
Chewable tablets should also be evaluated for in vivo bioavailability and/or 
bioequivalence. 
Additional concerns in the testing of chewable tablets are organoleptic, chemical, 
and physical stability. As it is a critical factor in the design of such formulations, 
taste masking should be incorporated into excipient testing during preformulation 
studies. Technologies like the “ electronic tongue ” can be used to match desirable 
taste characteristics [23, 24] . 
4.1.10 CHEWING GUMS 
4.1.10.1 Composition of Chewing Gum 
Medicated chewing gums are gums made with a tasteless masticatory gum base that 
consists of natural or synthetic elastomers [25] . They include excipients such as 
fi llers, softeners, and sweetening and fl avoring agents. Natural gum bases include 
chicle and smoked natural rubber and are permitted in formulations by the FDA, 
but modern gum bases are mostly synthetic in origin and approved bases include 
CHEWING GUMS 255

256 SOLID DOSAGE FORMS 
styrene – butadiene rubber, polyethylene, and polyvinylacetate. Gum base usually 
forms about 40% of the gum, but can comprise up to 65%, and is a complex mixture, 
insoluble in saliva, comprising mainly of elastomer, plasticizers, waxes, lipids, and 
emulsifi ers (see Table 8 ). It will also contain an adjuvant such as talc to modify the 
texture of the gum and low quantities of additional excipients including colorants 
and antioxidants such as butylated hydroxyanisole. Elastomers control the gummy 
texture while the plasticizers and texture agents regulate the cohesiveness of the 
product. The lipid and waxes melt in the mouth to provide a cooling, lubricating 
feeling while the juicy feel of the gum texture is from the emulsifi ers. The choice 
and formulation of gum base will affect the release of active ingredient, and the 
texture, stability, and method of manufacture of the product. 
The remaining ingredients in the chewing gum itself include drug, sweeteners, 
softeners, and fl avoring and coloring agents. A typical chewing gum formulation is 
shown in Table 9 . The sugar is for sweetening the product while the corn syrup keeps 
the gum fresh and fl exible. Softeners or fi llers are included to help blend the ingredients 
and retain moisture. Sugar - free gum has sorbitol, mannitol, aspartame, or 
saccharin instead of sugar. Optimized chewing gum formulations will require tailoring 
for each individual product. For example, nicotine - containing gums are formulated 
with the nicotine within an ion exchange resin and pH - modifying carbonates 
and/or bicarbonates to increase the percentage of the drug in its free base form 
in saliva. 
TABLE 8 Typical Formulation of Gum Base 
Ingredient Weight (%) Example 
Elastomer 10 Styrene – butadiene rubber 
Plasticizer 30 Rosin esters 
Texture agent/fi ller 35 Calcium carbonate 
Wax 15 Paraffi n wax 
Lipid 7 Soya oil 
Emulsifi er 3 Lecithin 
Miscellaneous 1 Colorant, antioxidant 
Source : From ref. 26 . 
TABLE 9 Example Chewing Gum Formulations 
Ingredient (%) Sugar Gum Sugar - Free Gum 
Gum base 19.4 25.0 
Corn syrup 19.8 — 
Sorbitol, 70% — 15.0 
Sugar 59.7 — 
Glycerin 0.5 6.5 
Sorbitol — 52.3 
Flavor 0.6 1.2 
Source : From ref. 26 . 

4.1.10.2 Manufacture of Chewing Gum 
The majority of chewing gum delivery systems today are manufactured using conventional 
gum processes. The gum base is softened or melted and placed in a kettle 
mixer where sweeteners, syrups, active ingredients, and other excipients are added 
at a defi ned time. The gum is then sent to a series of rollers that form it into a thin, 
wide ribbon. During this process, a light coating of an antisticking agent can be 
added (e.g., magnesium stearate, calcium carbonate, or fi nely powdered sugar or 
sugar substitute). Finally, the gum is cut to the desired size and cooled at a carefully 
controlled temperature and humidity. 
As the heating process involved in conventional methods may limit the applicability 
of the process for formulation of thermally labile drugs, directly compressible, 
free - fl owing powdered gums such as Pharmagum (SPI Pharma) and MedGumBase 
(Gumbase Co) have been proposed to simplify the process. These formulations 
can be compacted into a gum tablet using a conventional tablet press and have 
the potential to simplify the manufacture, facilitating inclusion of a wider range 
of drugs. 
4.1.10.3 Drug Release from Chewing Gums 
Until recently, the release of substances from chewing gums during mastication was 
studied using a panel of tasters and chew - out studies. During the mastication process, 
the medication contained within the gum product should be released into the saliva 
and is either absorbed through the buccal mucosa or swallowed and absorbed via 
the GI tract. The need for, and value of, in vitro drug release testing is well established 
for a range of dosage forms, however, standard dissolution apparatus is not 
suitable for monitoring release of drug from chewing gums as mastication is essential 
in order to provide a renewable surface for drug release after chew action. A 
number of devices to mimic the chewing action have been reported [26 – 28] . In 2000, 
the European Pharmacopoeia produced a monograph describing a suitable apparatus 
for studying the in vitro release of drug substances from chewing gums [25] . 
The chewing machine consists of a temperature - controlled chewing chamber in 
which the gum piece is chewed by two electronically controlled horizontal pistons 
driven by compressed air. The two pistons transmit twisting and pressing forces to 
the gum while a third vertical piston operates alternately to the two horizontal 
pistons to ensure that the gum stays in the right place (see Figure 1 ). The temperature 
of the chamber can be maintained at 37 ° C ± 0.5 ° C and the chew rate varied. 
Other adjustable settings include the volume of the medium, distance between the 
jaws and the twisting movement. The European Pharmacopoeia recommends using 
20 mL of unspecifi ed buffer in a chewing chamber of 40 mL and a chew rate of 60 
strokes per minute. This apparatus has been used to study release of nicotine from 
commercial gums and directly compressible gums [26] . 
Factors affecting the release of medicament from chewing gum can be divided 
into three groups: the physicochemical properties of the drug, the gum properties, 
and chew - related factors, including rate and frequency. Drugs can be incorporated 
into gums as solids or liquids. For most pharmaceuticals, aqueous solubility of the 
drug will be a major factor affecting the release rate. In order for drugs to be 
CHEWING GUMS 257

258 SOLID DOSAGE FORMS 
released, the gum would need to become hydrated; the drugs can then dissolve and 
diffuse through the gum base under the action of chewing. 
For treatment of local conditions, a release period less than 1 h may be desirable, 
but a faster release may be required if a rapid onset of action is required for a systemically 
absorbed formulation. There are a number of strategies that can be undertaken 
in order to achieve the desired release rate. Decreasing the amount of the 
gum base will enhance the release of lipophilic drugs and addition of excipients 
designed to promote release can also be considered. Release can be sustained using, 
for example, ion exchange resins as described for nicotine - containing gums. Changes 
in gum texture as a consequence of changes in excipient levels provide a further 
challenge to controlling the release of drugs. A quantitative measure of gum texture 
during the process is possible using texture analysis techniques [26] . 
4.1.10.4 Applications for Chewing Gums 
The promotion of sugar - free gums to counteract dental caries by stimulation of 
saliva secretion has led to a more widespread use and acceptance of gums. Medicated 
gums for delivery of dental products to the oral cavity are marketed in a 
number of countries, for example, fl uoride - containing gums as an alternative to 
mouthwashes and tablets or chlorhexidine gum for treatment of gingivitis. The 
potential use of medicated chewing gums in the treatment of oral infections has also 
been reported. Gums have been prepared containing antifungal agents such as 
nystatin [29] and miconazole [30] or antibiotics, such as penicillin and metronidazole 
for the treatment of oral gingivitis [31] . 
Chewing gum is also useful as a delivery system for agents intended for systemic 
delivery. Drug that is released from the gum within the oral cavity can act locally, 
be absorbed via the buccal mucosa, or swallowed with the saliva. The buccal mucosa 
is well vascularized, and if a drug is absorbed by this route, then fi rst - pass metabolism 
could be avoided. Associated increases in bioavailability can permit the use of 
lower dosages. Like orally disintegrating tablets, chewing gum is a convenient dosage 
form; it can be administered without water and to those who have diffi culty swallowing. 
Although medicated gums are generally intended to be chewed for 10 – 
30 min and can therefore be designed for sustained release, a fast onset of action 
can result either from buccal absorption or as a consequence of the active being 
FIGURE 1 Schematic of chewing chamber of in vitro chewing apparatus [26] . 
Chewing pistons 
Base of chewing chamber 
Piston 
Chewing chamber 

already dissolved in the saliva prior to swallowing. Guidance can be given regarding 
chewing conditions (e.g., time, frequency), but factors such as the force of chewing 
and salivary fl ow will impact on drug release and the fraction of drug absorbed via 
the oral mucosa. Released drug can be swallowed with the saliva, therefore leading 
to multiple absorption sites, which can result in variable pharmacokinetics. 
Along with nicotine replacement patches, nicotine chewing gum for smoking 
cessation therapy has met with major sales success. The principal active ingredient 
of currently marketed nicotine chewing gums is nicotine polacrilex USP. The nicotine 
is loaded at approximately 18% w/w on an ion exchange resin (Amberlite 
IRP64). Recent product variations have been launched with improved fl avors such 
as mint and fruit, rather than the original peppery fl avoring, designed to reduce the 
unpleasant taste and burning sensation arising from nicotine itself and fl avored 
coated gums that are sweeter and easier to chew. 
Other applications for chewing gum formulations include delivery of antacids 
such as calcium carbonate, antiemetics for travel sickness, and vitamins and minerals. 
However, the potential for a buccal delivery, a fast onset of action, and the opportunity 
for product line extension makes it an attractive alternative delivery form for 
other applications. 
4.1.11 ORALLY DISINTEGRATING TABLETS 
The demand for fast - dissolving/disintegrating tablets or fast - melting tablets that can 
dissolve or disintegrate in the mouth has been growing particularly for those with 
diffi culty swallowing tablets such as the elderly and children. They are referred to 
using a range of terminologies: fast dissolving, orodispersible, and fast melting and 
the FDA has adopted the term orally disintegrating tablets (ODTs). Patients with 
persistent nausea or those who have little or no access to water could also benefi t 
from ODTs. Other advantages include product differentiation and market expansion, 
and applications exist in the veterinary market for oral administration to 
animals. 
Orally disintegrating tablets disintegrate and/or dissolve rapidly in the saliva 
without the need for water, within seconds to minutes. Some tablets are designed 
to dissolve rapidly in saliva, within a few seconds, and are true fast - dissolving tablets. 
Others contain agents to enhance the rate of tablet disintegration in the oral cavity 
and are more appropriately termed fast - disintegrating tablets, as they may take up 
to a minute to completely disintegrate. Increased bioavailability using such formulations 
is sometimes possible if there is suffi cient absorption via the oral cavity prior 
to swallowing [32] . However, if the amount of swallowed drug varies, there is the 
potential for inconsistent bioavailability. Patented orally disintegrating tablet technologies 
include OraSolv, DuraSolv, Zydis, FlashTab, WOWTAB, and others. They 
are generally prepared using freeze drying, compaction, or molding. Examples of 
marketed products, excipients, and technologies used are given in Table 10 . 
Platform technologies based on freeze drying include Zydis (Cardinal Health) 
and Quicksolv (Janssen Pharmaceutica). Zydis was the fi rst ODT to be successfully 
launched, and it is ideal for poorly soluble drugs. It can incorporate doses up to 
400 mg, but high loadings can extend disintegration time. The porous matrix consists 
of a network of water - soluble carriers and active ingredient. The maximum dose for 
ORALLY DISINTEGRATING TABLETS 259

260 SOLID DOSAGE FORMS 
TABLE 10 Examples of Marketed ODT Products and Technologies 
Name (Company) Examples Ingredients a Technology 
Zydis 
(Cardinal Health) 
Claritin 
Reditab 
Micronized loratadine (10 mg) , citric 
acid, gelatin, mannitol, mint fl avor 
Freeze 
drying 
Zydis 
(Cardinal Health) 
Zofran 
ODT 
Ondansetron (4 or 8 mg) , aspartame, 
gelatin, mannitol, methylparaben 
sodium, propylparaben sodium, 
strawberry fl avor 
Freeze 
drying 
Zydis 
(Cardinal Health) 
Zyprexa 
Zydis 
Olanzapine (5, 10, 15, or 20 mg) , gelatin, 
mannitol, aspartame, methylparaben 
sodium, propylparaben sodium 
Freeze 
drying 
Oralsolv 
(CIMA Labs Inc.) 
Remeron 
Soltab 
Mirtazepine (15, 30, or 45 mg) , 
aspartame, citric acid, crospovidone, 
hydroxypropyl methylcellulose, 
magnesium stearate, mannitol, 
microcrystalline cellulose, 
polymethacrylate, povidone, sodium 
bicarbonate, starch, sucrose, orange 
fl avor 
Compression 
Durasolv 
(CIMA Labs Inc.) 
Zomig ZMT Zolmitriptan (2.5 mg) , mannitol, 
microcrystalline cellulose, 
crospovidone, aspartame, sodium 
bicarbonate, citric acid, anhydrous, 
colloidal silicon dioxide, magnesium 
stearate, orange fl avor 
Compression 
WOWTAB 
(Yamanouchi 
Pharma 
Technologies, 
Inc.) 
Benadryl 
Allergy & 
Sinus 
Fastmelt 
Diphenhydramine citrate (19 mg), 
pseudoephedrine HCl (30 mg), 
aspartame, citric acid, D & C red no. 7 
calcium lake, ethylcellulose, fl avor, 
lactitol, magnesium stearate, 
mannitol, and stearic acid 
Compression 
molded 
tablet 
Flashtab 
(Prographarm/ 
Ethypharm) 
Excedrin 
Quicktabs 
Acetaminophen (500 mg), caffeine 
(65 mg) , aminoalkyl methacrylate 
copolymers, citric acid, colloidal 
silicon dioxide, crospovidone, 
distilled acetylated monoglycerides, 
ethylcellulose, fl avors, magnesium 
stearate, mannitol, methacrylester 
copolymer, polyvinyl acetate, 
povidone, propylene glycol, propyl 
gallate, silica gel, sodium lauryl 
sulfate, sucralose, talc 
Compression 
a Active ingredients appear in italics. 
water - soluble drugs is 60 mg, and particle sizes of drug and excipients should be 
below 50 . m. 
Excipients used in the formulation usually include a mixture of a water - soluble 
polymer and a crystalline sugar. Mannitol and natural polysaccharides such as gelatin 
and alginates are used. Microencapsulation and complexation with ion exchange 
resins can be combined with additional fl avors and sweeteners for taste masking of 
bitter drugs. The fairly complex nature of manufacture and scale - up contributes to 

a relatively high manufacturing cost. Manufacture comprises three stages. The production 
sequence begins with the bulk preparation of an aqueous drug solution or 
suspension and subsequent precise dosing into preformed blisters. It is the blister 
that actually forms the tablet shape and is, therefore, an integral component of the 
total product package. The second phase of manufacturing entails passing the fi lled 
blisters through a cryogenic freezing process to control the ultimate size of the ice 
crystals. This aids in ensuring porosity and the product is freeze dried. The fi nal phase 
of production involves sealing the open blisters via a heat - seal process to ensure 
stability and protect the fragile tablet during removal by the patient. 
The manufacture of Flashdose (Fuisz Technologies/Biovail) is patented as 
Shearform process and utilizes a unique spinning mechanism to produce a fl osslike 
or shear - form crystalline structure, much like cotton candy. The matrix comprises 
saccharides or polysaccharides which are subjected to simultaneous melting and 
centrifugal force and then partially recrystallized [33] . High temperatures are 
involved so the technology is only suitable for thermostable agents. Drug can then 
be incorporated, either as coated or uncoated microspheres, into the sugar and the 
formulation is compressed into a tablet. Manufacture of the microspheres is patented 
as Ceform and will help with taste masking. The fi nal product has a very high 
surface area for dissolution and it disperses and dissolves quickly once placed onto 
the tongue. Like freeze - drying processes, the manufacture is expensive and resultant 
formulations are friable and moisture sensitive, therefore requiring specialized 
packaging. 
Most commercial ODTs have been developed using mannitol as the bulk excipient 
of choice because of its extremely low hygroscopicity, excellent compatibility, 
good compressibility, better sweetness, and relatively slower dissolution kinetics. 
Although lactose also has a relatively low aqueous solubility compared with other 
excipients that have acceptable palatabilities, the dispersibility of a bulk excipient 
is more important than its aqueous solubility for a successful ODT formulation. 
Many of the initially marketed ODTs were prepared by the wet granulation of 
mannitol followed by direct compression. However, added functionality mannitols 
are now available to simplify the process of ODT manufacturing by direct 
compression. 
Direct compression is, as for normal tablets, the most straightforward process for 
manufacturing ODTs. Conventional equipment can be used and high doses can be 
incorporated. The excipients play a major role in the successful formulation and 
superdisintegrants, hydrophilic polymers, and effervescent compounds are included. 
Patented technologies include Orasolv and Durasolv (Cima Labs) and Ziplets 
(Eurand). The OraSolv technology is best described as a fast disintegrating, slightly 
effervescing tablet; the tablet matrix dissolves in less than one minute, leaving 
coated drug powder. Both the coating and the effervescence contribute to taste 
masking in OraSolv. The tablet is prepared by direct compression but at a low pressure, 
yielding a weaker and more brittle tablet in comparison with conventional 
tablets. For that reason, Cima developed a special handling and packaging system 
for OraSolv called Packsolv. Acidic compounds such as citric or fumaric acid are 
included in the formulation together with a carbonate or bicarbonate. An advantage 
that goes along with the low degree of compaction of OraSolv is that the particle 
coating used for taste masking is not compromised by fracture during processing. 
DuraSolv is Cima ’ s second - generation fast - dissolving/disintegrating tablet formulation 
and is also produced using direct compression but using higher compaction 
ORALLY DISINTEGRATING TABLETS 261

262 SOLID DOSAGE FORMS 
pressures during tableting, resulting in a stronger product. It is thus produced in a 
faster and more cost - effective manner and may not require specialized packaging. 
Large amounts of fi nely milled conventional fi llers are used (mannitol, lactose) 
while the effervescing agents are reduced. It is best suited to potent drugs, requiring 
only low doses, and the taste - masking coating can be disturbed following 
compaction. DuraSolv is currently available in two products: NuLev and Zomig 
ZMT. 
Compression following wet or dry granulation is also employed in the manufacture 
of ODTs. Patented formulations include WOWTAB and Flashtab. WOWTAB 
relies on a combination of low moldable sahharides (mannitol, glucose, sucrose) 
with a highly moldable saccharide (malitol, sorbitol, maltose) using conventional 
granulation and tableting techniques to form a tablet of suitable mechanical properties 
with desired disintegration. It is manufactured by compression of molded granules, 
can accommodate a high level of drug loading (up to 50% in some cases), and 
can be packed using conventional methodology. 
Flashtab (Ethypharm) is the technology behind Exedrin QuickTabs and uses 
swellable agents and disintegrants along with sugars and polyalcohols to achieve a 
fast dispersible formulation. The manufacture involves either wet or dry granulation 
of the excipients, blending with the active followed by direct compression. 
4.1.11.1 Dissolution Testing of ODT s 
Taste masking (drug coating) is very often an essential feature of ODTs and thus 
can also be the rate - determining mechanism for dissolution/release. If taste masking 
is not an issue, then the development of dissolution methods is comparable to the 
approach taken for conventional tablets and pharmacopeial conditions should be 
used [34] . Due to the nature of the product, the dissolution of orally disintegrating 
tablets is very fast when using USP monograph conditions, and slower paddle speeds 
can be used to obtain a profi le. Other media such as 0.1 N HCl can also be used. 
USP 2 paddle apparatus is the most suitable and common choice for orally disintegrating 
tablets, with a paddle speed of 50 rpm commonly used [34] . Faster agitation 
rates may be necessary in the case of sample mounding. The method can be applied 
to the ODTs (fi nished product) as well as to the bulk intermediate (in the case of 
coated drug powder/granulate). A potential diffi culty for in vitro dissolution testing 
may arise from fl oating particles [35] . Similarly, diffi culties can arise using USP I 
due to trapping of disintegrated fragments. 
A single - point specifi cation is considered appropriate for ODTs with fast dissolution 
properties. For ODTs that dissolve very quickly, a disintegration test may be 
used in lieu of a dissolution test if it is shown to be a good discriminating method. 
If taste masking (using a polymer coating) is a key aspect of the dosage form, a 
multipoint profi le in a neutral pH medium with early points of analysis (e.g., . 5 min) 
may be recommended [34] . 
4.1.12 SOLID DOSAGE FORMS FOR NONORAL ROUTES 
Although the majority of tablets and capsules are intended for oral delivery, there 
are a number of other delivery routes suitable for drug delivery by these formula

tions. Some buccal formulations have been discussed above, and tablets can also be 
administered via the rectal and vaginal routes for local and systemic treatment. 
Many types of product have been designed for vaginal administration with 
creams, gels, and pessaries being most popular, although powders and tablets have 
also been used. Despite the effectiveness of systemic vaginal absorption, the majority 
of products administered by this route are for the treatment of localized infections, 
especially Candida albicans , (e.g., Canestan vaginal tablets). Estradiol tablets 
(Vagifem) were also designed for delivery via vaginal route to address patient preference 
issues with vaginal creams. The formulations are administered with an applicator 
and are designed to dissolve or erode slowly in the vaginal secretions [36] . 
Bioadhesion as a means of retaining the formulation at the site of delivery is widely 
accepted to retain formulations in the buccal cavity [37] and has also been reported 
for the vaginal route [38] . An increased residence time may improve drug absorption 
by these routes. 
REFERENCES 
1. Amidon , G. L. , Lennernas , H. , Shah , V. P. , and Crison , J. R. ( 1995 ), A theoretical basis for 
a biopharmaceutic drug classifi cation: The correlation of in vitro drug product dissolution 
and in vivo bioavailability , Pharm. Res. , 12 , 413 – 420 . 
2. Waiver of in - vivo bioavailability and bioequivalence studies for immediate release solid 
oral dosage forms based on a biopharmaceutics classifi cation system, available: http:// 
www.fda.gov/cder/OPS/BCS_guidance.html . 
3. Waiver of in vivo bioequivalence studies for immediate release solid oral dosage forms 
based on a biopharmaceutics classifi cation system, available: http://www.fda.gov/cder/ 
guidance/index.html . 
4. Zhao , C. , Jain , A. , Hailemariam , L. , Suresh , P. , Akkisetty , P. , Joglekar , G. , Venkatasubramanian 
, V. , Reklaitis , G.V. , Morris , K. , and Basu , P. ( 2006 ), Toward intelligent 
decision support for pharmaceutical product development , J. Pharm. Innovation , 1 , 
23 – 35 . 
5. Banker , G. S. , and Rhodes , C. T. ( 2002 ), Modern Pharmaceutics , 4th ed. , Drugs and the 
Pharmaceutical Sciences 121, Marcel Dekker , New York . 
6. Rowe , R. C. , Sheskey , P. J. , and Weller , P. J. ( 2001 ), Handbook of Pharmaceutical Excipients 
, 4th ed. , Pharmaceutical Press , London . 
7. Weiner , M. , and Bernstein I. L. ( 1989 ), Adverse Reactions to Drug Formulation Agents: A 
Handbook of Excipients , Marcel Dekker , New York , pp. 93 – 94 . 
8. Lachman , L. , Lieberman , H. A. , and Kanig , J. L. ( 1986 ), The Theory and Practice of Industrial 
Pharmacy , 3rd ed. , Lea & Febiger , Philadelphia . 
9. Augsburger , L. L. , Hahm, H. A. , Brzeczko , A. W. , and Shah, U. (2002), Superdisintegrants: 
Characterization and function , in Swarbrick , J. , and Boylan , J. V. , Eds., Encyclopedia of 
Pharmaceutical Technology , Vol. 3, 2nd ed. , Marcel Dekker , New York . 
10. Gibson , M. ( 2001 ), Pharmaceutical preformulation and formulation; a practical guide 
from candidate drug selection to commercial dosage form, IHS Health Group, 
Englewood, Colorado . 
11. Stanley , J. P. ( 1986 ), Soft gelatin capsules , in Lachman , L. , Lieberman , H. A. , and 
Kanig , J. L. , Eds., The Theory and Practice of Industrial Pharmacy , 3rd ed. , Lea & Febiger , 
Philadelphia . 
REFERENCES 263

264 SOLID DOSAGE FORMS 
12. The United States Pharmacopeia , 26th revision, United States Pharmacopeial Convention, 
Rockville, MD, 2003 . 
13. Gelatin Capsule Working Group, Collaborative development of two - tier dissolution 
testing for gelatin capsules and gelatin - coated tablets using enzyme - containing media, 
Pharmacop. Forum , 24(5), Sept./Oct. 1998 . 
14. Lee , R. E. , Effervescent tablets. Key facts about a unique effective dosage form, Tablets 
Capsules , available: http://www.amerilabtech.com/EffervescentTablets&KeyFacts.pdf , 
accessed Aug. 6, 2004 . 
15. Rotthauser , B. , Kraus , G. , and Schmidt , P. C. ( 1998 ), Optimization of an effervescent tablet 
formulations containing spray - dried l - leucine and polyethylene glycol 6000 as lubricants 
using a central composite design , Eur. J. Pharm. Biopharm. , 46 , 85 – 94 . 
16. Stahl , H. ( 2003 ), Effervescent dosage manufacturing , Pharm. Technol. Eur. , 4 , 25 – 28 . 
17. Lindberg , N. - O. , and Hansson , H. ( 2002 ), Effervescent pharmaceuticals , in Swarbrick , J. , 
and Boylan , J. V. , Eds., Encyclopedia of Pharmaceutical Technology , Vol. 2, 2nd ed. , Marcel 
Dekker , New York . 
18. Mendes , R. W. , and Bhargava , H. ( 2002 ), Lozenges , in Swarbrick J. , and Boylan , J. V. , 
Eds., Encyclopedia of Pharmaceutical Technology , Vol. 2, 2nd ed. , Marcel Dekker , New 
York . 
19. Richards , R. M. E. , Xing , J. Z. , and Weir , L. F. C. ( 1996 ), The effect of formulation on the 
antimicrobial activity of cetylpyridinium chloride in candy based lozenges , Pharm. Res. , 
13 , 583 – 587 . 
20. Bolhuis , G. , and Armstrong , A. N. ( 2006 ), Excipients for direct compaction — An update , 
Pharm. Dev. Technol. , 11 , 111 – 124 . 
21. Bowe , K. E. ( 1998 ), Recent advances in sugar - based excipients , Pharm. Sci. Technol. 
Today , 1 ( 4 ), 166 – 173 . 
22. FDA guidance for industry: Bioavailability and bioequivalence studies for orally 
administered drug products — General considerations, Oct. 2000 . 
23. Murray , O. J. , Dang , W. , and Bergstrom , D. ( 2004 ), Using an electronic tongue to optimize 
taste - masking in a lyophilized orally disintegrating tablet formulation , Pharm. Technol. 
Outsourcing Res. , 42 – 52 . 
24. Zheng , J. Y. , and Keeney , M. P. ( 2004 ), Taste masking analysis in pharmaceutical formulation 
development using an electronic tongue , Int. J. Pharm. , 310 , 118 – 124 . 
25. European Pharmacopoeia , 4th ed., European Directorate for the Quality of Medicines, 
Strasbourg, 2001 . 
26. Morjaria , Y. , Irwin , W. J. , Barnett , P. X. , Chan , R. S. , and Conway , B. R. ( 2004 ), In vitro 
release of nicotine from chewing gum formulations , Dissolution Technol. , 11 , 12 – 15 . 
27. Rider , J. N. , Brunson , E. L. , Chambliss , W. G. , Cleary , R. W. , Hikal , A. H. , Rider , P. H. , 
Walker , L. A. , Wyandt , C. M. , and Jones , A. B. ( 1992 ), Development and evaluation of a 
novel dissolution apparatus for medicated chewing gum products , Pharm. Res. , 9 , 
255 – 260 . 
28. Kvist , C. , Andersson , S. - B. , Fors , S. , Wennergren , B. , and Berglund , J. ( 1999 ), Apparatus 
for studying in vitro release from medicated chewing gums , Int. J. Pharm. , 89 , 57 – 65 . 
29. Andersen , T. , Gram - Hansen , M. , Pedersen , M. , and Rassing , M. R. ( 1990 ), Chewing gum 
as a drug delivery system for nystatin. Infl uence of solubilising agents on the release of 
water - soluble drugs , Drug Dev. Ind. Pharm. , 16 , 1985 – 1994 . 
30. Pedersen , M. , and Rassing , M. R. ( 1991 ), Miconazole chewing gum as a drug delivery 
system test of release promoting additives , Drug Dev. Ind. Pharm. , 17 , 411 – 420 . 
31. Emslie , R. D. ( 1967 ), Treatment of acute ulcerative gingivitis. A clinical trial using chewing 
gums containing metronidazole or penicillin , Br. Dent. J. , 122 , 307 – 308 . 

32. Habib , W. , Khankari , R. , and Hontz , J. ( 2002 ), Fast - dissolve drug delivery system , Crit. 
Rev. Therap. Drug Carrier Syst. , 17 , 61 – 72 . 
33. Dobetti, L. (2001), Fast-melting tablets: Developments and technologies , Pharm. Technol. 
Drug Deliv. Suppl. , 44 – 50 . 
34. Klanke , J. (2003), Dissolution testing of orally disintegrating tablets , Dissolution Technol. , 
10 ( 2 ), 6 – 8 . 
35. Siewert , M. , Dressman , J. , Brown , C. , and Shah , V. P. ( 2003 ), FIP/AAPS. Guidelines for 
dissolution/in vitro release testing of novel/special dosage forms , Dissolution Technol. , 
10 ( 1 ), 6 – 15 . 
36. Conine , J. W. , and Pikal , M. J. ( 1989 ), Special tablets , in Lieberman , H. A. , Lachman , L. , 
and Schwartz , J. B. Eds., Pharmaeutical Dosage Forms: Tablets , Vol. 1, 2nd ed. , Marcel 
Dekker , New York . 
37. Sudhakar , Y. , Kuotsu , K. , and Bandyopadhyay , A. K. ( 2006 ), Buccal bioadhesive drug 
delivery — A promising option for orally less effi cient drugs , J. Controlled Release , 114 , 
15 – 40 . 
38. Hussain , A. , and Ahsan , F. ( 2005 ), The vagina as a route for systemic drug delivery , J. 
Controlled Release , 103 , 301 – 313 . 
REFERENCES 265


267 
4.2 
SEMISOLID DOSAGES: OINTMENTS, 
CREAMS, AND GELS 
Ravichandran Mahalingam , Xiaoling Li , and Bhaskara R. Jasti 
University of the Pacifi c, Stockton, California 
Contents 
4.2.1 Introduction 
4.2.2 Ointments and Creams 
4.2.2.1 Defi nition 
4.2.2.2 Bases 
4.2.2.3 Preparation and Packaging 
4.2.2.4 Evaluation 
4.2.2.5 Typical Pharmacopeial/Commercial Examples 
4.2.3 Gels 
4.2.3.1 Defi nition 
4.2.3.2 Characteristics 
4.2.3.3 Classifi cation 
4.2.3.4 Stimuli - Responsive Hydrogels 
4.2.3.5 Gelling Agents 
4.2.3.6 Preparation and Packaging 
4.2.3.7 Evaluation 
4.2.3.8 Typical Pharmacopeial and Commercial Examples 
4.2.4 Regulatory Requirements for Semisolids 
References 
4.2.1 INTRODUCTION 
Semisolid dosage forms are traditionally used for treating topical ailments. The vast 
majority of them are meant for skin applications. They are also used for treating 
ophthalmic, nasal, buccal, rectal, and vaginal ailments. Various categories of drugs 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

268 SEMISOLID DOSAGES: OINTMENTS, CREAMS, AND GELS 
such as antibacterials, antifungals, antivirals, antipruritics, local anesthetics, anti - 
infl ammatories, analgesics, keratolytics, astringents, and mydriatic agents are incorporated 
into these products. Drugs incorporated into semisolids either show their 
activity on the surface layers of tissues or penetrate into internal layers to reach the 
site of action. For example, an antiseptic ointment should be able to penetrate the 
skin layers and reach the deep - seated infections in order to prevent the growth of 
microbes and heal the wound. 
Systemic entry of drugs from these products is limited due to various physicochemical 
properties of dosage forms and biological factors. The barrier nature of 
most surface biological layers such as skin, cornea and conjunctiva of the eye, and 
mucosa of nose, mouth, rectum, and vagina greatly limits their entry into the systemic 
circulation. Systemic delivery of drugs from topical dosages is however feasible by 
suitable formulation modifi cations. Semisolid dosage forms are also used in nontherapeutic 
conditions for providing protective and lubricating functions. They 
protect the skin against external environments such as air, moisture, and sun rays and 
hence their components do not necessarily penetrate the skin layers. Cold creams 
and vanishing creams are classic examples of such semisolid preparations. 
The formulation, evaluation, and regulatory feature of the three most commonly 
used semisolid dosage forms, ointments, creams, and gels, are described in this 
chapter. 
4.2.2 OINTMENTS AND CREAMS 
4.2.2.1 Defi nition 
Ointments are semisolid preparations intended for topical application. They are 
used to provide protective and emollient effects on the skin or carry medicaments 
for treating certain topical ailments. They are also used to deliver drugs into eye, 
nose, vagina, and rectum. Ointments intended for ophthalmic purposes are required 
to be sterile. When applied to the eyes, they reside in the conjunctival sac for prolonged 
periods compared to solutions and suspensions and improve the fraction of 
drug absorbed across ocular tissues. Ophthalmic ointments are preferred for nighttime 
applications as they spread over the entire corneal and conjunctival surface 
and cause blurred vision. 
Creams are basically ointments which are made less greasy by incorporation of 
water. Presence of water in creams makes them act as emulsions and therefore are 
sometimes referred as semisolid emulsions. Hydrophilic creams contain large 
amounts of water in their external phase (e.g., vanishing cream) and hydrophobic 
creams contain water in the internal phase (e.g., cold cream). An emulsifying agent 
is used to disperse the aqueous phase in the oily phase or vice versa. As with ointments, 
creams are formulated to provide protective, emollient actions or deliver 
drugs to surface or interior layers of skin, rectum, and vagina. Creams are softer 
than ointments and are preferred because of their easy removal from containers 
and good spreadability over the absorption site. 
4.2.2.2 Bases 
Bases are classifi ed based on their composition and physical characteristics. The U.S. 
Pharmacopeia (USP) classifi es ointment bases as hydrocarbon bases (oleaginous 

OINTMENTS AND CREAMS 269 
bases), absorption bases, water - removable bases, and water - soluble bases (water - 
miscible bases) [1] . 
Hydrocarbon bases are made of oleaginous materials. They provide emollient 
and protective properties and remain in the skin for prolonged periods. It is diffi cult 
to incorporate aqueous phases into hydrocarbon bases. However, powders can be 
incorporated into these bases with the aid of liquid petrolatum. Removal of hydrocarbon 
bases from the skin is diffi cult due to their oily nature. Petrolatum USP, white 
petrolatum USP, yellow ointment USP, and white ointment USP are examples of 
hydrocarbon bases. 
Absorption bases contain small amounts of water. They provide relatively less 
emollient properties than hydrocarbon bases. Similar to hydrocarbon bases, absorption 
bases are also diffi cult to remove from the skin due to their hydrophobic nature. 
Hydrophilic petrolatum USP and lanolin USP are examples of absorption bases. 
Water - removable bases are basically oil - in - water emulsions. Unlike hydrocarbon 
and absorption bases, a large proportion of aqueous phase can be incorporated into 
water - removable bases with the aid of suitable emulsifying agents. It is easy to 
remove these bases from the skin due to their hydrophilic nature. Hydrophilic ointment 
USP is an example of a water - removable ointment base. 
Water - soluble bases do not contain any oily or oleaginous phase. Solids can be 
easily incorporated into these bases. They may be completely removed from the 
skin due to their water solubility. Polyethylene glycol (PEG) ointment National 
Formulary (NF) is an example of a water - soluble base. 
Selection of an appropriate base for an ointment or cream formulation depends 
on the type of activity desired (e.g., topical or percutaneous absorption), compatibility 
with other components, physicochemical and microbial stability of the product, 
ease of manufacture, pourability and spreadability of the formulation, duration of 
contact, chances of hypersensitivity reactions, and ease of washing from the site of 
application. In addition, bases that are used in ophthalmic preparations should be 
nonirritating and should soften at body temperatures. White petrolatum and liquid 
petrolatum are generally used in ophthalmic preparations. Table 1 summarizes 
TABLE 1 Some Compendial Bases Used in Ointments and Creams 
Name Synonyms 
Offi cial 
Compendia Specifi cations 
Carnauba wax Caranda wax, 
Brazil wax 
BP, JP, 
PhEur, 
USPNF 
Melting range 80 – 88 ° C a ; iodine value 
5 – 14b ; acid value 2 – 7; saponifi cation 
value 78 – 95; total ash . 0.25% 
Cetyl alcohol Cetanol, Avol, 
Lipocol C 
BP, JP, 
PhEur, 
USPNF 
Melting range 47 – 53 ° C b ; residue on 
ignition . 0.05% b ; iodine value . 5.0; 
acid value . 2.0; saponifi cation value 
. 2.0 a 
Cetyl ester wax Crodamol SS, 
Ritachol SS, 
Starfol wax 
USPNF Melting range 43 – 47 ° C; acid value 
. 5.0; saponifi cation value 109 – 120; 
iodine value . 1.0 
Emulsifying 
wax 
Collone HV, 
Crodex A, 
Lipowax PA 
BP Saponifi cation value . 2.0; iodine value 
. 3.0 c

270 SEMISOLID DOSAGES: OINTMENTS, CREAMS, AND GELS 
Name Synonyms 
Offi cial 
Compendia Specifi cations 
Hydrous lanolin Hydrous wool 
fat, Lipolan 
BP, JP, 
PhEur 
Melting range 38 – 44 ° C; acid value 
. 0.8; saponifi cation value 67 – 79; 
nonvolatile matter 72.5 – 77.5%; 
iodine value 18 – 36b 
Lanolin Wool fat, 
purifi ed wool 
fat, Corona 
BP, JP, 
PhEur, 
USPNF 
Melting range 38 – 44 ° C; loss on drying 
. 0.25%; residue on ignition . 0.1%; 
iodine value 18 – 36; acid value . 1.0 b 
Lanolin 
alcohols 
Argowax, 
Ritawax, wool 
wax alcohol 
BP, PhEur, 
USPNF 
Melting range . 56 ° C; loss on drying 
. 0.50%; residue on ignition . 0.15%; 
acid value . 2.0; saponifi cation value 
. 12 
Microcrystalline 
wax 
Petroleum 
ceresin 
USPNF Melting range 54 – 102 ° C; residue on 
ignition . 0.10% 
Paraffi n Paraffi n wax, 
hard wax, 
hard paraffi n 
BP, JP, 
PhEur, 
USPNF 
Melting range 47 – 65 ° C 
Petrolatum Yellow soft 
paraffi n, 
yellow 
petroleum 
jelly 
BP, JP, 
PhEur, 
USPNF 
Melting range 38 – 60 ° C; residue on 
ignition . 0.1% 
Poloxamer Polyethylene – 
propylene 
glycol, Lutrol, 
Pluronic 
BP, PhEur, 
USPNF 
Melting point . 50 ° C 
Polyethylene 
glycol (PEG) 
Macrogol, 
Carbowax, 
PEG, Lutrol 
BP, JP, 
PhEur, 
USPNF 
Melting range of PEG 1000, 37 – 40 ° C; 
melting range of PEG 8000, 60 – 
63 ° C; residue on ignition . 0.1% 
Stearic acid Emersol, 
Hystrene 
BP, JP, 
PhEur, 
USPNF 
Melting range . 54 ° C; iodine value . 4.0 
Stearyl alcohol Lipocol S, 
Cachalot, Rita 
SA 
BP, JP, 
PhEur, 
USPNF 
Melting range 55 – 60 ° C; residue on 
ignition 0.05% b ; iodine value . 2.0; 
acid value . 2.0; saponifi cation value 
. 2.0 a 
White wax Bleached wax BP, JP, 
PhEur, 
USPNF 
Melting range 62 – 65 ° C; acid value 17 – 
24; saponifi cation value 87 – 104a 
Yellow wax Refi ned wax BP, JP, 
PhEur, 
USPNF 
Acid value 17 – 22 a ; saponifi cation value 
87 – 102a 
Note : BP, British Pharmacopoeia; JP, Japanese Pharmacopoeia; PhEur, European Pharmacopoeia; 
USPNF, U.S. Pharmacopeia/National Formulary. All are USPNF specifi cations, except as indicated 
below. 
a European Pharmacopoeia. 
b Japanese Pharmacopoeia. 
c British Pharmacopoeia. 
TABLE 1 Continued

OINTMENTS AND CREAMS 271 
compendial status, synonym, and specifi cations of some of the bases used in ointments 
and creams. 
The following sections describe the source, physicochemical properties, formulation 
considerations, stability, incompatibility, storage, and hypersensitivity reactions 
of some of these bases. 
Lanolin Lanolin is a refi ned, decolorized, and deodorized material obtained from 
sheep wool. It is available as a pale yellow, waxy material with a characteristic odor. 
It is extensively used in the preparation of hydrophobic ointments and water - in - oil 
creams. As lanolin is prone to oxidation, antioxidants such as butylated hydroxytoluene 
are generally included. Although lanolin is insoluble in water, it is miscible 
with water up to 1 : 2 ratio. This property favors in preparing physically stable creams. 
Addition of soft paraffi n or vegetable oil improves the emollient property of lanolin 
preparations. Exposure of lanolin to higher temperature usually leads to discoloration 
and rancidlike odor, and hence prolonged heating is avoided during the 
preparation and preservation of lanolin - containing preparations. Gamma sterilization 
or fi ltration sterilization is usually employed for sterilizing ophthalmic ointments 
containing lanolin. Lanolin and some of its derivatives are reported to cause 
hypersensitivity reactions and therefore are avoided in patients with known hypersensitivity. 
One of the reasons for hypersensitivity reactions is free fatty alcohols. 
Modifi ed lanolins containing reduced levels of free fatty alcohols are commercially 
available [2, 3] . 
Hydrous Lanolin Incorporation of about 25 – 30% of water into lanolin gives 
hydrous lanolin. Gradual addition of water into molten lanolin with constant stirring 
helps in water incorporation. It is available as a pale yellow, oily material with a 
characteristic odor. The water uptake capacity of hydrous lanolin is higher than 
lanolin, and it is used for preparing topical hydrophobic ointments or water - in - oil 
creams with larger aqueous phase. Exposure of these preparations to higher temperatures 
results in separations of oily and aqueous layers. Addition of antioxidants 
and preservation in well - fi lled, airtight, light - resistant containers in a cool and dry 
place improve the stability of lanolin products. Well - preserved preparations can be 
stored up to two years. Hydrous lanolin that contains free fatty alcohols is avoided 
in hypersensitive patients [2, 3] . 
Lanolin Alcohols Lanolin alcohol is prepared from lanolin by the saponifi cation 
process and is used as a hydrophobic vehicle in pharmaceutical ointments and 
creams. It is composed of steroidal and triterpene alcohols and is available as a 
brittle solid material pale yellow in color with a faint characteristic odor. The brittle 
powder becomes plastic under warm conditions. It is practically insoluble in water 
and soluble in boiling ethanol. Lanolin alcohol possesses emollient properties, which 
makes it suitable for preparing dry - skin ointments, eye ointments, and water - in - oil 
creams. Creams containing lanolin alcohols do not show surface darkening and 
do not produce objectionable odor compared to lanolin - containing preparations. 
Inclusion of about 0.1% antioxidant, however, minimizes the oxidation on storage. 
Preparations containing lanolin alcohols can be stored up to two years if preserved 
in well - fi lled, well - closed, light - resistant containers in a cool and dry place. As with 

272 SEMISOLID DOSAGES: OINTMENTS, CREAMS, AND GELS 
other lanolin bases, hypersensitivity reactions may occur in some individuals while 
using preparations containing lanolin alcohols [2, 3] . 
Petrolatum Petrolatum is also known as yellow soft paraffi n. It is an inert material 
obtained from petroleum, which contains branched and unbranched hydrocarbons. 
It is available as soft oily material and appears pale yellow to yellow in color. Various 
grades of petrolatum are commercially available with varying physical properties. 
All these grades are generally insoluble in water and possess emollient properties. 
Concentrations up to 30% are used in creams. Petrolatum shows phase transitions 
on heating to about 35 ° C. As it possesses a higher coeffi cient of thermal expansion, 
prolonged heating is avoided during processing. The presence of minor impurities 
can oxidize petrolatum and discolor the product. Antioxidants are therefore added 
to prevent such physical changes in preparations during storage. Butylated hydroxyanisole, 
butylated hydroxytoluene, or . - tocopherol is generally incorporated as an 
antioxidant in petrolatum products. In addition, use of well - closed, airtight, light - 
resistant containers and storage in a cool and dry place improve stability of preparations. 
Minor quantities of polycyclic aromatic hydrocarbon impurities in petrolatum 
sometimes cause hypersensitivity reactions. Substituting yellow soft paraffi n with 
white soft paraffi n reduces such reactions [4] . 
Petrolatum and Lanolin Alcohols Various quantities of lanolin alcohols are mixed 
with petrolatum to form these mixtures. Wool ointment British Pharmacopoeia (BP) 
2001 contains 6% lanolin alcohols and 10% petrolatum. These proportions can be 
varied to alter physical properties such as consistency and melting range. They are 
available as soft solids pale ivory in color and possess a characteristic odor. These 
mixtures are insoluble in water, and concentrations ranging 5 – 50% are used for 
preparing hydrophobic ointments. They are also used for preparing water - in - oil 
emollient creams. Preparations containing petrolatum and lanolin alcohols need to 
be preserved in airtight, well - closed, light - resistant containers in a cool and dry place 
to avoid oxidation of impurities and discoloration. Antioxidants improve the stability 
of these products. Although these mixtures are safe for topical applications, 
hypersensitivity reactions may occur in some individuals due to the presence of 
lanolin alcohol [5] . 
Paraffi n Paraffi n is obtained by distillation of crude petroleum followed by puri- 
fi cation processes. The purifi ed fraction contains saturated hydrocarbons. Paraffi n is 
available as a white color solid and does not possess any specifi c odor or taste. Different 
purity grades are available. Use of highly purifi ed grades can avoid batch - to - 
batch variations in formulations, especially the hardness, melting behavior, and 
malleability. Paraffi n is insoluble in water and is generally used to prepare hydrophobic 
topical ointments and water - in - oil creams. Repeated heating and congealing 
are avoided during formulation as they change the physical properties of paraffi n. 
These preparations need to be preserved in well - closed container at room temperature. 
Synthetic paraffi ns, which melt between 96 and 105 ° C, are sometimes used to 
increase the melting point and stiffness of formulations [6] . 
Polyethylene Glycol Also known as macrogol, PEG is synthesized by condensation 
of ethylene oxide and water under suitable reaction conditions. Based on the 

OINTMENTS AND CREAMS 273 
number of oxyethylene groups present, their molecular weights vary from few hundreds 
to several thousands. Usually the number that follows PEG represents their 
average molecular weight. They are available as liquids or solids based on molecular 
weight. PEGs 600 or less are liquids, whereas PEGs above 1000 are solids. PEG 
liquids are usually clear or pale yellow in color. Their viscosity increases with 
increase in molecular weight. Solid PEGs are usually white in color and available 
as pastes, waxy fl akes, or free - fl owing solids based on their molecular weight. Table 
2 shows the physicochemical properties of some PEGs. 
PEGs are hydrophilic materials and are extensively used in the preparation of 
hydrophilic ointments and creams. They are nonirritants and are easily washed 
from skin surfaces. Products with varying consistency are prepared by mixing different 
grades of PEGs. Excessive heating is avoided while melting PEGs. This will 
prevent oxidation and discoloration of products. In addition, use of purifi ed grades 
that are free from peroxide impurities, inclusion of suitable antioxidants, and 
heating under nitrogen atmosphere can minimize the oxidation. PEGs are prone 
to etherifi cation or esterfi cation reactions due to the presence of two terminal 
hydroxyl groups. They are incompatible with some antibiotics, antimicrobial preservatives, 
iron, tannic acid, and salicylic acid and also interact with plastic containers 
made of polyvinyl chloride and polyethylene. PEG - containing products are 
usually packed in aluminum, glass, or stainless steel containers to avoid such interactions. 
Although low - molecular - weight PEGs are hygroscopic, they do not promote 
microbial growth. PEG - containing products are generally stored in well - closed 
containers in a cool, dry place. These products can cause stinging sensation on 
mucus and some hypersensitivity reactions, especially when applied onto open 
wounds [7, 8] . 
Stearic Acid Stearic acid is obtained by hydrolysis of fat or hydrogenation of 
vegetable oils. Compendial stearic acid contains a mixture of stearic acid and palmitic 
acids. It is available as powder or crystalline solid which is white to yellowish 
white in color and possesses a characteristic odor. Although stearic acid is insoluble 
TABLE 2 Properties of Different Grades of PEG 
Property 
By Grade 
200 400 600 1000 2000 3000 4000 8000 
Physical 
state 
Liquid Liquid Liquid Solid Solid Solid Solid Solid 
Average 
molecular 
weight 
190 – 210 380 – 420 570 – 613 950 – 1050 1800 – 2200 2700 – 3300 3000 – 4800 7000 – 9000 
Melting 
( ° C) 
— — — 37 – 40 45 – 50 48 – 54 50 – 58 60 – 63 
Density 
(g/cm3 ) 
1.11 – 1.14 1.11 – 1.14 1.11 – 1.14 1.15 – 1.21 1.15 – 1.21 1.15 – 1.21 1.15 – 1.21 1.15 – 1.21 
Kinematic 
viscositya 
(cS) 
3.9 – 4.8 6.8 – 8.0 9.9 – 11.3 16.0 – 19.0 38 – 49 67 – 93 110 – 158 470 – 900 
a At 98.9 ° C. 

274 SEMISOLID DOSAGES: OINTMENTS, CREAMS, AND GELS 
in water, partially neutralized grades form a cream base when combined with about 
10 times its weight of aqueous solvents. The appearance and consistency of these 
grades are based on the proportion of alkali or triethanolamine used for neutralization. 
Concentrations up to 20% are used for formulating creams and ointments. 
Different grades of stearic acids are commercially available with varying stearic acid 
content, melting temperature, and other physical properties. A suitable antioxidant 
is included in formulations containing stearic acid. As stearic acid interacts with 
metals, it is avoided in preparations which contain salts, especially divalent metals 
such as calcium and zinc. It also reacts with metal hydroxides and some drugs. Compatibility 
evaluation between stearic acid and other formulation components is 
therefore essential when formulating newer products with stearic acid [9] . 
Carnauba Wax Carnauba wax contains a mixture of esters of acids and hydroxyacids 
isolated from Brazilian carnauba palm. It also contains various resins, hydrocarbons, 
acids, polyhydric alcohols, and water. It is available as lumps, powder, or 
fl akes which are brown to pale yellow in color and possesses a characteristic odor. 
Carnauba wax is practically insoluble in water and melts at 80 – 88 ° C. Being a hard 
material, it improves the stiffness of topical preparations [6] . 
Cetyl Alcohol Cetyl alcohol is obtained by hydrogenolysis or esterfi cation of fatty 
acids and contains not less than 90% cetyl alcohol along with other aliphatic alcohols. 
It is available as fl akes or granules white in color and possesses a characteristic 
odor. Different grades are commercially available with varying proportions of cetyl 
alcohol, stearyl alcohol, and related alcohols. Although insoluble in water, cetyl 
alcohol has good water - absorptive and emulsifying properties. This property makes 
it suitable for preparing emollient ointments and creams. Its viscosity - enhancing 
properties reduce coalescence of dispersed phase and improves the physical stability 
of creams. Concentrations ranging from 2 to 10% are used in topical preparations 
to impart emollient, emulsifying, water - absorptive, and stiffening properties. Mixtures 
of petrolatum and cetyl alcohol are sometimes used for preparing creams. Such 
mixtures minimize the quantity of additional emulsifying agents in preparations. 
Although cetyl alcohol forms stable preparations, it is incompatible with strong 
oxidizing materials and some drugs. Compatibility studies are therefore conducted 
when including cetyl alcohol into formulations. Highly purifi ed grades are free from 
hypersensitivity reactions [3, 10] . 
Emulsifying Wax Emulsifying wax, also known as anionic emulsifying wax, is a 
mixture of cetostearyl alcohol, sodium lauryl sulfate, and purifi ed water. Emulsifying 
wax BP contains about 90% cetostearyl alcohol, 10% sodium lauryl sulfate, and 4% 
purifi ed water. Emulsifying wax USP contains nonionic surfactants. It is available 
as fl akes or solids which are white to pale yellow in color and possesses a characteristic 
odor. Although emulsifying wax is insoluble in water, its emulsifying properties 
help in preparing hydrophilic oil - in - water emulsions. Ointment bases are 
prepared by mixing up to 50% emulsifying wax with liquid or soft paraffi ns. At 
concentrations up to 10%, it forms creams. Although emulsifying wax is compatible 
with many acids and alkalis, it is incompatible with many cationic materials and 
polyvalent metal salts. Stainless steel vessels are preferred for mixing operations. 

OINTMENTS AND CREAMS 275 
Preparations containing emulsifying wax are preserved in well - closed container in 
a cool, dry place [11] . 
Cetyl Esters Wax Cetyl esters wax is obtained by esterifi cation of some fatty alcohols 
and fatty acids. It is available as crystalline fl akes which are white to off - white 
in color and possesses a characteristic aromatic odor. It is insoluble in water and 
has emollient and stiffening properties. About 10% of cetyl ester wax is used for 
preparing hydrophobic creams and about 20% is used for preparing topical ointments. 
Various grades of cetyl esters wax are available commercially and vary in 
their fatty alcohol and fatty acids content and melting range. As this wax is incompatible 
with strong acids and bases, it should be avoided in certain formulations. 
Cetyl ester wax – containing products are stored in well - closed containers in a cool, 
dry place [6] . 
Hydrogenated Castor Oil It is used as stiffening agent in hydrophobic ointments 
and creams due to its higher melting point. Hydrogenated castor oil contains triglyceride 
of hydroxystearic acid and is available as white color fl akes or powder. It 
is insoluble in water and melts at 85 – 88 ° C. Different grades with varying compositions 
and physical properties are commercially available. Products can be prepared 
at higher temperatures, as hydrogenated castor oil is stable up to 150 ° C. It is compatible 
with other waxes obtained from vegetable and animal sources. Preparations 
containing hydrogenated castor oil need to be preserved in well - closed containers 
in a cool and dry place [12] . 
Microcrystalline Wax Microcrystalline wax is obtained from petroleum by solvent 
fractionation and dewaxing procedures. It contains many straight - chain and 
branched - chain alkanes, with carbon chain lengths ranging from 41 to 57. It is available 
as fi ne fl akes or crystals which are white or yellow in color. Microcrystalline 
wax is insoluble in water and possesses a wide melting range (54 – 102 ° C). High - 
melting and stiffening properties of microcrystalline wax make it suitable for preparing 
ointments and cream with higher consistency. Acids, alkalis, oxygen, and light 
do not affect its stability [6] . 
Stearyl Alcohol Reduction of ethyl stearate in the presence of lithium aluminum 
hydride yields stearyl alcohol, which contains not less than 90% of 1 - octadecanol. 
It is available as fl akes or granules which are white in color and possesses a characteristic 
odor. It is insoluble in water and melts at 55 – 60 ° C. Stearyl alcohol has stiffening, 
viscosity - enhancing, and emollient properties and hence is used in the 
preparation of hydrophobic ointments and creams. Its weak emulsifying properties 
help in improving the water - holding capacity of ointments. Hypersensitivity reactions 
are sometimes observed due to the presence of some minor impurities. Stearyl 
alcohol preparations are compatible with acids and alkalis and are preserved in 
well - closed containers in a cool and dry place [6] . 
White Wax White wax is a bleached form of yellow wax which is usually obtained 
from the honeycomb of bees and hence is known as bleached wax or white bees wax . 
It contains about 70% esters of straight - chain monohydric alcohols, 15% free acids, 

276 SEMISOLID DOSAGES: OINTMENTS, CREAMS, AND GELS 
12% carbohydrates, and 1% free wax alcohols and stearic esters of fatty acids. It is 
available as granules or sheets which are white in color and possesses a characteristic 
odor. White wax is insoluble in water and melts between 61 and 65 ° C. It has stiffening 
and viscosity - enhancing properties and therefore is used in hydrophobic ointments 
and oil - in - water creams. Although it is thermally stable, heating to above 
150 ° C results in reduction of its acid value. White wax is incompatible with oxidizing 
agents. The presence of small quantities of impurities results in hypersensitivity 
reactions in rare occasions. Preparations are stored in well - closed, light - resistant 
containers in a cool, dry place [13] . 
Yellow Wax Yellow wax, also known as yellow beeswax, is obtained from honey 
combs. It contains about 70% esters of straight - chain monohydric alcohols, 15% 
free acids, 12% carbohydrates, and 1% free wax alcohols and stearic esters of fatty 
acids. It is available as noncrystalline pieces which are yellow in color and possesses 
a characteristic odor. It is practically insoluble in water and melts at 61 – 65 ° C. It is 
used in the preparation of hydrophobic ointments and water - in - oil creams because 
of its viscosity - enhancing properties. Concentrations up to 20% are used for producing 
ointments and creams. It is incompatible with oxidizing agents. Esterifi cation 
occurs while heating to 150 ° C and hence should be avoided during preparation. 
Hypersensitivity reactions sometimes occur on topical application of yellow wax – 
containing ointments and creams due to the presence of some minor impurities. 
These products are preserved in well - closed, light - resistant containers [13] . 
Combinations of bases are sometimes used to acquire better stability. Gelling 
agents such as carbomers and PEG are also included in some ointment and cream 
preparations. Table 3 shows examples of cream bases used in some commercial 
cream preparations. 
4.2.2.3 Preparation and Packaging 
In addition to the base and drug, ointments and creams may also contain other 
components such as stabilizers, preservatives, and levigating agents. Usually levigation 
and fusion methods are employed for incorporating these components into the 
base. Levigation involves simple mixing of base and other components over an ointment 
slab using a stainless steel ointment spatula. A fusion process is employed only 
when the components are stable at fusion temperatures. Ointments and creams 
containing white wax, yellow wax, paraffi n, stearyl alcohol, and high - molecular - 
weight PEGs are generally prepared by the fusion process. Selection of levigation 
or the fusion method depends on the type base, the quantity of other components, 
and their solubility and stability characteristics. 
Oleaginous ointments are prepared by both levigation and fusion processes. 
Small quantities of powders are incorporated into hydrocarbon bases with the aid 
of a levigating agent such as liquid petrolatum, which helps in wetting of powders. 
The powder component is mixed with the levigating agent by trituration and is then 
incorporated into the base by spatulation. All solid components are milled to fi ner 
size and screened before incorporating into the base to avoid gritty sensation of the 
fi nal product. Roller mills are used for producing large quantities of ointments in 
pharmaceutical industries. Uniform mixing can be obtained by the geometric dilution 
procedure, which usually involves stepwise dilution of solids into the ointment 

OINTMENTS AND CREAMS 277 
TABLE 3 Cream Bases Present in Some Commercial Creams 
Commercial Name Drug Cream Base (s) Used 
Dritho - Calp, 
Psoriatec 
Anthralin, 0.5%, 1.0% White petrolatum, cetostearyl alcohol 
Temovate E Clobetasol propionate, 
0.05% 
Propylene glycol, glyceryl monostearate, 
cetostearyl alcohol, glyceryl stearate, 
PEG 100 stearate, white wax 
Eurax Crotamiton, 10% Petrolatum, propylene glycol, cetyl 
alcohol, carbomer - 934 
Topicort Desoximetasone, 
0.25% 
White petrolatum USP, isopropyl 
myristate NF, lanolin alcohols NF, 
mineral oil USP, cetostearyl alcohol NF 
Apexicon, Maxifl or, 
Psorcon 
Difl orasone diacetate, 
0.05% 
Hydrophilic vanishing cream base of 
propylene glycol, stearyl alcohol, cetyl 
alcohol 
Lidex Cream, Vanos Fluocinonide, 0.05%, 
0.10% 
Polyethylene glycol 8000, propylene 
glycol, stearyl alcohol 
Carac Fluorouracil, 0.5%, 
1.0%, 5.0% 
Carbomer - 940, PEG 400, propylene 
glycol, stearic acid 
Halog Halcinonide, 0.1% Polyethylene and mineral oil gel base 
with PEG 400, PEG 6000, PEG 300, 
PEG 1450 
Cortaid, Anusol-Hc, 
Proctosol HC 
Hydrocortisone, 2.5% 
water washable 
Petrolatum, stearyl alcohol, propylene 
glycol, carbomer - 934 
Monistat - Derm Miconazole nitrate, 
2% 
Water - miscible base consisting of pegoxol 
7 stearate, peglicol 5 oleate, mineral oil, 
butylated hydroxyanisole 
base. The fusion method is followed when the drugs and other solids are soluble in 
the ointment bases. The base is liquefi ed, and the soluble components are dissolved 
in the molten base. The mixture is then allowed to congeal by cooling. Fusion is 
performed using steam - jacketed vessels or a porcelain dish. The congealed mixture 
is then spatulated or triturated to obtain a smooth texture. Care is taken to avoid 
thermal degradation of the base or other components during the fusion process. 
Absorption - type ointments and creams are prepared by incorporating large 
quantities of water into hydrocarbon bases with the aid of a hydrophobic emulsifying 
agent. Water - insoluble drugs are added by mechanical addition or fusion methods. 
As with oleaginous ointments, levigating agents are also included to improve wetting 
of solids. Water - soluble or water - miscible agents such as alcohol, glycerin, or propylene 
glycol are used if the drug needs to be incorporated into the internal aqueous 
phase. If the drug needs to be incorporated into the external oily phase, mineral oils 
are used as the levigating agent. Incorporation of water - soluble components is 
achieved by slowly adding the aqueous drug solution to the hydrophobic base using 
pill tile and spatula. If the proportion of aqueous phase is larger, inclusion of additional 
quantities of emulsifi er and application of heat may be needed to achieve 
uniform dispersion. Care must be taken to avoid excessive heating as it can result 
in evaporation aqueous phase and precipitation of water - soluble components and 
formation of stiff and waxy product. 

278 SEMISOLID DOSAGES: OINTMENTS, CREAMS, AND GELS 
Water - removable ointments and creams are basically hydrophilic - type emulsions. 
They are prepared by fusion followed by mechanical addition approach. Hydrocarbon 
components are melted together and added to the aqueous phase that 
contains water - soluble components with constant stirring until the mixture congeals. 
A hydrophilic emulsifying agent is included in the aqueous phase in order to obtain 
stable oil - in - water dispersion. Sodium lauryl sulfate is used in the preparation of 
hydrophilic ointment USP. 
Water - soluble ointments and creams do not contain any oily phase. Both water - 
soluble and water - insoluble components are incorporated into water - soluble bases 
by both levigation and fusion methods. If the drug and other components are water 
soluble, they are dissolved in a small quantity of water and incorporated into the 
base by simple mixing over an ointment slab. If the components are insoluble in 
water, aqueous levigating agents such as glycerin, propylene glycol, or a liquid PEG 
are used. The hydrophobic components are mixed with the levigating agent and then 
incorporated into the base. Heat aids incorporation of a large quantity of hydrophobic 
components. 
A wide range of machines are available for the large - scale production of ointments 
and creams. Each of these machines is designed to perform certain unit 
operations, such as milling, separation, mixing, emulsifi cation, and deaeration. 
Milling is performed to reduce the size of actives and other additives. Various fl uid 
energy mills, impact mills, cutter mills, compression mills, screening mills, and tumbling 
mills are used for this purpose. Alpine, Bepex, Fluid Air, and Sturtevant are 
some of the manufacturers of these mills. Separators are employed for separating 
materials of different size, shape, and densities. Either centrifugal separators or 
vibratory shakers are used for separation. Mixing of the actives and other formulation 
components with the ointment or cream base is performed using various types 
of low - shear mixers, high - shear mixers, roller mills, and static mixers. Mixers with 
heating provisions are also used to aid in the melting of bases and mixing of components. 
Chemineer, Fryma, Gate, IKA, Koruma (Romaco), Moorhouse - Cowles, 
Ross, and Stokes Merrill are some of the manufacturers of semisolids mixers. 
Creams are produced with the help of low - shear and high - shear emulsifi ers. 
These emulsifi ers are used to disperse the hydrophilic components in the hydrophobic 
dispersion phase (e.g., water - in - oil creams) or oleaginous materials in aqueous 
dispersion medium (oil - in - water creams). Bematek, Fryma, Koruma (Romaco), 
Lightnin, Moorhouse, and Ross supply various types of emulsifi ers. Entrapment of 
air into the fi nal product due to mixing processes is a common issue in the large - 
scale manufacturing of semisolid dosage forms. Various offl ine and in - line deaeration 
procedures are adopted to minimize this issue. Effective deaeration is generally 
achieved by using vacuum vessel deaerators. Some of the recent large - scale machines 
are designed to perform heating, high - shear mixing, scrapping, and deaeration processes 
in a single vessel. Figure 1 shows the design feature of a semisolid production 
machine manufactured by Ross. 
Various low - and high - shear shifters are used to transfer materials from the production 
vessel to the packaging machines. In the packaging area, various types of 
holders (e.g., pneumatic, gravity, and auger holders), fi llers (e.g., piston, peristaltic 
pump, gear pump, orifi ce, and auger fi llers), and sealers (e.g., heat, torque, microwave, 
indication, and mechanical crimping sealers) are used to complete the unit 

OINTMENTS AND CREAMS 279 
operations. These equipments are supplied by various manufacturers, namely Bosch, 
Bonafacci, Erweka, Fryma - Maschinenbau, IWKA, Kalish, and Norden. 
Sterility of ointments, especially those intended for ophthalmic use, is achieved 
by aseptic handling and processing. Improper processing, handling, packing, or use 
of ophthalmic ointments lead to microbial contaminations and eventually result in 
ocular infections. In general, the empty containers are separately sterilized and fi lled 
under aseptic condition. Final product sterilization by moist heat sterilization or 
gaseous sterilization is ineffective because of product viscosity. Dry - heat sterilization 
is associated with stability issues. Strict aseptic procedures are therefore practiced 
when processing ophthalmic preparations. Antimicrobial preservatives such as 
benzalkonium chloride, phenyl mercuric acetate, chlorobutanol, or a combination 
of methyl paraben and propyl paraben are included in ophthalmic ointments to 
retain microbial stability. 
Packaging An ideal container should protect the product from the external atmosphere 
such as heat, humidity, and particulates, be nonreactive with the product 
components, and be easy to use, light in weight, and economic [14] . As tubes made 
of aluminum and plastic meet most of these qualities, they are extensively used for 
packaging semisolids. Aluminum tubes with special internal epoxy coatings are 
commercially available for improving the compatibility and stability of products. 
Various modifi ed plastic materials are used for making ointment tubes. Tubes made 
FIGURE 1 Semisolid production machine with heat jacketed vessel, high - shear mixer, 
scrapper, vacuum attachments, and control station. (Courtesy of Ross, Inc.) 

280 SEMISOLID DOSAGES: OINTMENTS, CREAMS, AND GELS 
of low - density polyethylene (LDPE) are generally soft and fl exible and offer good 
moisture protection. Tubes made of high - density polyethylene (HDPE) are relatively 
harder but offer high moisture protection. Polypropylene containers offer 
high heat resistance. Plastic containers made of polyethylene terephthalate (PET) 
are transparent and provide superior chemical compatibility. Ointments meant for 
ophthalmic, nasal, rectal, and vaginal applications are supplied with special application 
tips for the ease of product administration. 
A recent method known as blow fi ll sealing (BFS) performs fabrication of 
container, fi lling of product, and sealing operations in a single stage and hence is 
gaining greater attention. The products can be sterile fi lled, which makes BFS a 
cost - effective alternative for aseptic fi lling. All plastic materials are suitable for 
BFS processing. In most cases, monolayered LDPE materials are used for making 
small - size containers. If the product is not compatible with the LDPE or sensitive 
to oxygen, barrier layers are added to the container wall by coextrusion methods. 
As the container is formed inside the BFS machine, upstream handling problems 
are avoided. The BFS machine can hand the container off to any secondary packaging 
operation that needs to be performed. Typically a secondary overwrap is added 
to the containers prior to cartooning. An additional advantage of BFS containers is 
the integrated design of the applicator into the product container. Figure 2 shows 
some of the custom - designed BFS containers for topical products. 
4.2.2.4 Evaluation 
Ointments and creams are evaluated for various pharmacopeial and nonpharmacopeial 
tests to ascertain their physicochemical, microbial, in vitro, and in vivo 
characteristics. These tests help in retaining their quality and minimizing the batch - 
to - batch variations. The USP recommends storage and labeling, microbial screening, 
minimum fi ll, and assays for most ointments and creams. Tables 4 and 5 summarize 
the compendial requirements for some pharmacopeial ointments and creams. 
FIGURE 2 Custom - designed LDPE containers made by BFS process for packaging topical 
products. (Courtesy of Rommelag USA, Inc.) 

OINTMENTS AND CREAMS 281 
TABLE 4 USP Specifi cations for Some Offi cial Ointments 
Drug Quality Control Tests Packaging and Storage Requirements 
Acyclovir Staphylococcus aureus, 
Pseudomonas aeruginosa , 
minimum fi ll, limit of 
guanine, and assay 
Tight containers; store between 15 and 
25 ° C in a dry place 
Alclometasone 
dipropionate 
S. aureus, P. aeruginosa , 
minimum fi ll, and assay 
Collapsible tubes or tight containers, 
store at controlled room temperature 
Amphotericin B Minimum fi ll, water, and 
assay 
Collapsible tubes or other well - closed 
containers 
Anthralin Assay Tight containers; in a cool place; protect 
from light 
Bacitracin Minimum fi ll, water, and 
assay 
Well - closed containers containing not 
more than 60 g; controlled temperature 
Benzocaine S. aureus, P. aeruginosa , 
minimum fi ll, and assay 
Tight containers; protect from light; avoid 
prolonged exposure to temperatures 
exceeding 30 ° C 
Betamethasone 
valerate 
S. aureus, P. aeruginosa , 
minimum fi ll, and assay 
Collapsible tubes or tight containers; 
avoid exposure to excessive heat. 
Clioquinol Assay Collapsible tubes or tight, light - resistant 
containers 
Clobetasol 
propionate 
S. aureus, P. aeruginosa, 
Escherichia coli, 
Salmonella species, total 
aerobic microbial count, 
minimum fi ll, and assay 
Collapsible tubes or in tight containers; 
store at controlled room temperature; 
do not refrigerate 
Erythromycin Minimum fi ll, water, and 
assay 
Collapsible tubes or in tight containers at 
controlled room temperature 
Fluocinolone 
acetonide 
S. aureus, P. aeruginosa , and 
assay 
Tight containers 
Gentamycin 
sulfate 
Minimum fi ll, water, and 
assay 
Collapsible tubes or in tight containers; 
avoid exposure to excessive heat 
Hydrocortisone 
valerate 
S. aureus, P. aeruginosa , 
total microbial count, 
minimum fi ll, and assay 
Tight container; store at room 
temperature 
Ichthammol Assay Collapsible tubes or in tight containers; 
avoid prolonged exposure to 
temperatures exceeding 30 ° C 
Lidocaine S. aureus, P. aeruginosa , 
minimum fi ll, and assay 
Tight containers 
Mometasone 
furoate 
S. aureus, P. aeruginosa, E. 
coli, Salmonella species, 
minimum fi ll, and assay 
Well - closed containers 
Nitrofurazone Completeness of solution 
and assay 
Tight, light - resistant containers; avoid 
exposure to direct sunlight, strong 
fl uorescent lighting, and excessive heat 
Nitroglycerine Minimum fi ll, homogeneity, 
and assay 
Tight containers 
Nystatin Minimum fi ll, water, and 
assay 
Well - closed containers at controlled 
room temperature 
Tetracycline 
hydrochloride 
Minimum fi ll, water, and 
assay 
Well - closed containers at controlled 
room temperature 
Zinc oxide Minimum fi ll, calcium, 
magnesium, other foreign 
substances, and assay 
Tight containers; avoid prolonged 
exposure to temperatures exceeding 
30 ° C 

282 SEMISOLID DOSAGES: OINTMENTS, CREAMS, AND GELS 
TABLE 5 USP Specifi cations for Some Offi cial Creams 
Cream Quality Control Tests Packaging and Storage Requirements 
Alclometasone 
dipropionate 
Microbial limits, minimum 
fi ll, and assay 
Collapsible tubes or tight containers; store 
at controlled room temperature 
Amphotericin B Minimum fi ll and assay Collapsible tubes or other well - closed 
containers 
Benzocaine Microbial limits, minimum 
fi ll, and assay 
Tight containers, protected from light, 
and avoid prolonged exposure to 
temperatures exceeding 30 ° C 
Betamethasone 
dipropionate 
Minimum fi ll and assay Collapsible tubes or tight containers; store 
at 25 ° C; excursions permitted between 
15 and 30; protect from freezing 
Ciclopirox 
olamine 
Minimum fi ll, pH, content 
of benzyl alcohol, and 
assay 
Collapsible tubes at controlled room 
temperature 
Clobetasol 
propionate 
Microbial limits, minimum 
fi ll, pH, and assay 
Collapsible tubes or tight containers; store 
at controlled room temperature; do not 
refrigerate 
Clotrimazole Assay Collapsible tubes or tight containers at a 
temperature between 2 and 30 ° C 
Desoximetasone Minimum fi ll, pH, and 
assay 
Collapsible tubes at controlled room 
temperature 
Dibucaine Microbial limits, minimum 
fi ll, and assay 
Collapsible tubes or in tight, light - resistant 
containers 
Dienestrol Minimum fi ll and assay Collapsible tubes or in tight containers 
Difl orasone 
diacetate 
Microbial limits, minimum 
fi ll, and assay 
Collapsible tubes, preferably at controlled 
room temperature 
Fluocinolone 
acetonide 
Microbial limits, minimum 
fi ll, and assay 
Collapsible tubes or in tight containers 
Fluorouracil Microbial limits, minimum 
fi ll, and assay 
Tight containers and stored at controlled 
room temperature 
Gentamycin 
sulfate 
Minimum fi ll and assay Collapsible tubes or in other tight 
containers; avoid exposure to excessive 
heat 
Hydrocortisone 
butyrate 
Microbial limits, minimum 
fi ll, pH, and assay 
Well - closed containers 
Hydroquinone Minimum fi ll and assay Well - closed, light - resistant containers 
Lindane pH and assay Tight containers 
Meclocycline 
sulfosalicylate 
Minimum fi ll and assay Tight containers, protected from light 
Miconazole 
nitrate 
Minimum fi ll and assay Collapsible tubes or tight containers; store 
at controlled room temperature 
Monobenzone Assay Tight containers; avoid exposure to 
temperatures higher than 30 ° C 
Nystatin Minimum fi ll and assay Collapsible tubes or in other tight 
containers; avoid exposure to excessive 
heat 
Prednisolone Minimum fi ll and assay Collapsible tubes or in tight containers 
Tetracaine 
hydrochloride 
Microbial limits, minimum 
fi ll, pH between 3.2 and 
3.8, and assay 
Collapsible, lined metal tubes 
Triamcinolone 
acetonide 
Microbial limits, minimum 
fi ll, and assay 
Tight containers 

OINTMENTS AND CREAMS 283 
Packaging and Storage The USP recommends packaging and storage requirements 
for each offi cial ointment and cream. Generally collapsible tubes, tight containers, 
or other well - closed containers are recommended for packing. They are 
stored in either a cool place or at controlled room temperatures. In some cases, 
special storage conditions are recommended: for example, protect from light, avoid 
exposure to excessive heat, avoid exposure to direct sunlight, avoid strong fl uorescent 
lighting, do not refrigerate, and avoid prolonged exposure to temperatures 
exceeding 30 ° C. 
Minimum Fill This test is performed to compare the weight or volume of product 
fi lled into each container with their labeled weight or volume. It helps in assessing 
the content uniformity of product. A minimum - fi ll test is applied only to those 
containers that contain not more than 150 g or mL of preparation. It is performed 
in two steps. Initially, labels from the product containers are removed. After washing 
and drying the surface, their weights are recorded ( W1 ). In the second step, the entire 
product from each container is removed. After cleaning and drying, the weight of 
empty containers is recorded ( W2 ). The difference between total weight ( W1 ) and 
empty - container weight ( W2 ) gives the weight of product. The USP recommends 
that the average net content of 10 containers should not be less than the labeled 
amount. If the product weight is less than 60 g or mL, the net content of any single 
container should not be less than 90% of the labeled amount. If the product weight 
is between 60 and 150 g or mL, the net content of any single container should not 
be less than 95% of the labeled amount. If these limits are not met, the test is 
repeated with an additional 20 containers. All semisolid topical preparations should 
meet these specifi cations [15] . 
Water Content The presence of minor quantities of water may alter the microbial, 
physical, and chemical stability of ointments and creams. Titrimetric methods 
(method I) are usually performed for determining the water content in these preparations. 
These methods are based on the quantitative reaction between water and 
anhydrous solution of sulfur and iodine in the presence of a buffer that can react 
with hydrogen ions. Special titration setups and reagents (Karl Fischer, KF) are used 
in these determinations. In the direct method (method Ia), about 35 mL of methanol 
is titrated with suffi cient quantity of KF reagent to the electrometric or visual endpoint 
(color change from canary yellow to amber). This blank titration helps to 
consume any moisture that may be present in the reaction medium. A known quantity 
of test material (ointment or cream) is added to the reaction medium, mixed, 
and again titrated with KF reagent to the reaction endpoint. The water content is 
determined by considering the volume of KF reagent consumed and its water 
equivalence factor. In the residual titration method (method Ib), a known excess 
quantity of KF reagent is added to the titration vessel, which is then back titrated 
with standardized water to the electrometric or visual endpoint. In the coulometric 
titration method (method Ic), the sample is dissolved in anhydrous methanol and 
injected into the reaction vessel that contains the anolyte, and the coulometric reaction 
is performed until the reaction endpoint. In some cases, methanol is replaced 
with other solvents. The maximum allowable limit of water in ointment preparations 
varies between 0.5 and 1.0%. The limit of water in bacitracin, chlortetracycline 
hydrochloride, and nystatin ointments is not more than 0.5%, whereas amphotericin 

284 SEMISOLID DOSAGES: OINTMENTS, CREAMS, AND GELS 
B, erythromycin, gentamycin sulfate, neomycin sulfate, and tetracycline hydrochloride 
ointments may contain up to 1% moisture [15] . 
Metal Particles This test is required only for ophthalmic ointments. The presence 
of metal particles will irritate the corneal or conjunctival surfaces of the eye. It is 
performed using 10 ointment tubes. The content from each tube is completely 
removed onto a clean 60 - mm - diameter petridish which possesses a fl at bottom. The 
lid is closed and the product is heated at 85 ° C for 2 h. Once the product is melted 
and distributed uniformly, it is cooled to room temperature. The lid is removed after 
solidifi cation. The bottom surface is then viewed through an optical microscope at 
30. magnifi cation. The viewing surface is illuminated using an external light source 
positioned at 45 ° on the top. The entire bottom surface of the ointment is examined, 
and the number of particles 50 . m or above are counted using a calibrated eyepiece 
micrometer. The USP recommends that the number of such particles in 10 tubes 
should not exceed 50, with not more than 8 particles in any individual tube. If these 
limits are not met, the test is repeated with an additional 20 tubes. In this case, the 
total number of particles in 30 tubes should not exceed 150, and not more than 3 
tubes are allowed to contain more than 8 particles [15] . 
Leakage Test This test is mandatory for ophthalmic ointments, which evaluates 
the intactness of the ointment tube and its seal. Ten sealed containers are selected, 
and their exterior surfaces are cleaned. They are horizontally placed over absorbent 
blotting paper and maintained at 60 ± 3 ° C for 8 h. The test passes if leakage is not 
observed from any tube. If leakage is observed, the test is repeated with an additional 
20 tubes. The test passes if not more than 1 tube shows leakage out of 30 
tubes [15] . 
Sterility Tests Ophthalmic semisolids should be free from anaerobic and aerobic 
bacteria and fungi. Sterility tests are therefore performed by the membrane fi ltration 
technique or direct - inoculation techniques. In the membrane fi ltration method, 
a solution of test product (1%) is prepared in isopropyl myristate and allowed to 
penetrate through cellulose nitrate fi lter with pore size less than 0.45 . m. If necessary, 
gradual suction or pressure is applied to aid fi ltration. The membrane is then 
washed three times with 100 - mL quantities of sterile diluting and rinsing fl uid and 
transferred aseptically into fl uid thioglycolate (FTG) and soybean – casein digest 
(SBCD) medium. The membrane is fi nally incubated for 14 days. Growth on FTG 
medium indicates the presence of anaerobic and aerobic bacteria, and SBCD 
medium indicates fungi and aerobic bacteria. Absence of any growth in both these 
media establishes the sterility of the product. In the direct - inoculation technique, 1 
part of the product is diluted with 10 parts of sterile diluting and rinsing fl uid with 
the help of an emulsifying agent and incubated in FTG and SBCD media for 14 
days. In both techniques, the number of test articles is based on the batch size of 
the product. If the batch size is less than 200 the containers, either 5% of the containers 
or 2 containers (whichever is greater) are used. If the batch size is more than 
200, 10 containers are used for sterility testing [15] . 
Microbial Screening Semisolid preparations are required to be free from any 
microbial contamination. Hence, most of the topical ointments are screened for the 

OINTMENTS AND CREAMS 285 
presence of Staphylococcus aureus and Pseudomonas aeruginosa . In some cases, 
screening for Escherichia coli, Salmonella species, and total aerobic microbial counts 
is recommended by the USP. For instance, clobetasol propionate ointment USP and 
mometasone furoate ointment USP are screened for all these organisms. In addition, 
preparations meant for rectal, vaginal, and urethral applications are tested for yeasts 
and molds [15] . 
Test for S. aureus and P. aeruginosa The test sample is mixed with about 100 mL 
of fl uid soybean – casein digest (FSBCD) medium and incubated. If microbial growth 
is observed, it is inoculated in agar medium by the streaking technique. Vogel – 
Johnson agar (VJA) medium is used for S. aureus screening, and cetrimide agar 
(CA) medium is used for screening P. aeruginosa . The petridishes are then closed, 
inverted, and incubated under appropriate conditions. The appearance of black 
colonies surrounded by a yellow zone over VJA medium and greenish colonies in 
CA medium indicates the presence of S. aureus and P. aeruginosa , respectively. 
Various other agar media are also available for screening these organisms. A coagulase 
test is then performed for confi rming the presence of S. aureus and oxidase and 
pigment tests for confi rming P. aeruginosa . 
Test for Salmonella Species and E. coli The test sample is mixed with about 100 mL 
of fl uid lactose (FL) medium and incubated. If microbial growth is observed, the 
contents are mixed and 1 mL is transferred to vessels containing 10 mL of fl uid 
selinite cystine (FSC) medium and fl uid tetrathionate (FT) medium and incubated 
for 12 – 24 h under appropriate conditions. To identify the presence of Salmonella , 
samples from the above two media are streaked over brilliant green agar (BGA) 
medium, xylose lysine desoxycholate agar (XLDA) medium, and bismuth sulfi te 
agar (BSA) medium and incubated. The appearance of small, transparent or pink - 
to - white opaque colonies over BGA medium, red colonies with or without black 
centers over XLDA medium, and black or green colonies over BSA medium indicates 
the presence of Salmonella . It is further confi rmed in triple sugar iron agar 
medium. The presence of E. coli is screened by streaking the samples from FL 
medium over MacConkey agar medium. The appearance of brick red colonies indicates 
the presence of E. coli . It is further confi rmed using Levine eosin methylene 
blue agar medium. 
total aerobic microbial counts The plate method or multiple - tube method is 
performed to estimate the total count. About 10 g or 10 mL of the test sample is 
dissolved or suspended in suffi cient volume of phosphate buffer (pH 7.2), fl uid 
soybean casein digest (FSBCD) medium, or fl uid casein digest – soy lecithin – 
polysorbate 20 medium to make the fi nal volume 100 mL. In the plate method, about 
1 mL of this diluted sample is mixed with molten soybean – casein digest agar 
(SBCDA) medium and solidifi ed at room temperature. The plates are inverted and 
incubated for two to three days. The number of colonies that are on the surface of 
nutrient media are counted. The multiple tube method is performed using sterile 
fl uid SBCD medium. The number of colonies formed should not exceed the limits 
specifi ed in an individual monograph. For example, clobetasol propionate ointment 
USP and hydrocortisone valerate ointment USP contains less than 100 colony - 
forming units (CFU) per gram of sample. 

286 SEMISOLID DOSAGES: OINTMENTS, CREAMS, AND GELS 
Test for Yeasts and Molds The plate method is used for testing molds and yeast in 
semisolids. The procedure is similar to that of the total count test. Instead of SBCDA 
medium, Sabouraud dextrose agar (SDA) medium or potato dextrose agar (PDA) 
medium is used. Samples are incubated for fi ve to seven days at 20 – 25 ° C to identify 
the presence of yeasts and molds. 
Assay The quantity of drug present in a unit weight or volume of ointment or 
cream is determined by various methods. Spectrophotometric, titrimetric, chromatographic, 
and in some cases microbial assays are performed. Selection of a particular 
method is based on the nature of drug, its concentration in the product, interference 
between the drug and other formulation components, and offi cial requirements. 
Although spectrophotometric methods are accurate and easy to perform, the complexity 
of ointment matrix sometimes reduced the specifi city of analysis compare 
to liquid chromatographic methods. The USP prescribes high - performance liquid 
chromatographic (HPLC) assays for many offi cial ointments due to its specifi city, 
accuracy, and precision. For example, amcinonide, anthralin, betamethasone dipropionate, 
clobetasol propionate, dibucaine, nitroglycerine, hydrocortisone, and triamcinolone 
acetonide are assayed by HPLC methods. These methods involve extraction 
of drug from the formulation matrix using suitable solvents followed by chromatographic 
separation using suitable reversed - phase columns followed by ultraviolet 
(UV) detection. Clioquinol preparation is assayed by gas chromatography. The USP 
also recommends potentiometric titrations (benzocaine, lidocaine, and ichthammol) 
and complexometric titrations (zinc oxide) for some semisolid preparations. 
Microbial assays are recommended for certain preparations containing antibiotics 
such as amphotericin B, bacitracin, chlortetracycline hydrochloride, gentamycin 
sulfate, neomycin sulfate, and nystatin. These tests evaluate the potency of an antibiotic 
by means of its inhibitory effects on specifi c microorganism. Two types of 
microbial assays are performed to determine the antibiotic potency. They are known 
as cylindrical plate or plate assays and turbidimetric or tube assays. The plate 
method measures the extent of growth inhibition of a particular microorganism in 
solidifi ed agar medium in the presence of the test antibiotic (commonly known as 
zone of inhibition ). The tube method measures the turbidity of a liquid medium that 
contains a particular organism in the presence and absence of the test antibiotic. 
These methods involve extracting drug from the formulation matrix, diluting the 
drug to a known concentration, and measuring the zone of inhibition or turbidity. 
In Vitro Drug Release Studies These studies are conducted to ascertain release 
of drug from the formulation matrix. Open - chamber diffusion cells such as Franz 
cells are used for performing in vitro studies. These cells consist of a donor side and 
a receiver side separated by a synthetic membrane such as cellulose acetate/nitrate 
mixed ester, polysulfone, or polytetrafl uoroethylene. The membranes are usually 
pretreated with the receiver fl uid to avoid any lag phase in drug release. The receiver 
side is fi lled with a known volume of release medium and is heated to 32 ± 0.5 ° C 
by circulating warm water through an outer jacket. Aqueous buffers are used for 
water - soluble drugs. Phosphate buffer solution of pH 5.4 is considered most appropriate 
for dermatological products as it mimics the pH of skin. Hydroalcoholic or 
other suitable medium may also be used for sparingly water soluble drugs. A known 
quantity of the test product is applied uniformly over the membrane on the donor 

OINTMENTS AND CREAMS 287 
side and samples are withdrawn from the receiver side at different time intervals. 
After each sampling, an equal volume of fresh medium is replaced to the receiver 
side. The sampling time points are different for different formulations; however, at 
least fi ve samples are withdrawn during the study period for determining the release 
rate. A typical sample time sequence for a 6 - h study is 0.5, 1.0, 2.0, 4.0, and 6.0 h. 
The receiver samples are analyzed by a suitable analytical method to quantify the 
amount of drug released from the formulation at different time intervals. The slope 
of the straight line which is obtained from a plot of cumulative amount drug release 
across 1 - cm 2 membrane versus the square root of time represents the release rate. 
Experiments are conducted in hexaplicate to obtain statistically signifi cant results 
[16] . 
In Vivo Bioequivalence Studies In vivo studies are conducted to establish the 
biological availability or activity of the drug from a topically applied semisolid formulation. 
Dermatopharmacokinetic studies, pharmacodynamic studies, or comparative 
clinical trials are generally conducted to assess the bioequivalence of topical 
products [16, 17] . 
Dermatopharmacokinetic (DPK) studies are applicable for topical semisolid 
products that contain antifungals, antivirals, corticosteroids, and antibiotics and 
vaginally applied products. They are not applicable for ophthalmic, otic, and other 
products that damage stratum corneum. DPK studies involve measurement of drug 
concentrations in stratum corneum, drug uptake, apparent steady state, and elimination 
after application of the test product onto skin. These studies are conducted in 
healthy human subjects adopting crossover design. The test and the reference products 
are applied onto eight to nine sites in the forearm. The surface area of each 
site is based on the strength of drug, extent of drug diffusion, exposure time, and 
sensitivity of the analytical technique. The application site is washed and allowed to 
normalize for at least 2 h prior to drug application. A known amount of product is 
applied onto these selected sites. At appropriate time intervals, the excess of drug 
from each area is removed using cotton swabs or soft tissue papers. Care is taken 
to avoid stratum corneum damage during sample collection. Stripping of stratum 
corneum is performed using adhesive tape - strips (e.g., D - Squame, Transpore). In the 
elimination phase, the excess drug is removed at the steady - state time point, and 
the stratum corneum is harvested at succeeding times over 24 h. The drug content 
from strips from each time point are extracted using suitable solvents and quantifi ed 
by a validated analytical method. A stratum corneum drug levels – time curve is 
developed, and pharmacokinetic parameters such as maximum concentration at 
steady state ( Cmax - ss ), time to reach Cmax - ss ( Tmax - ss ), and areas under the curve for the 
test and standard (AUC test and AUC reference ) are computed. DPK studies are performed 
in either one or two occasions. If performed in one occasion, both arms of 
a single subject are used to compare the test and reference products. If performed 
in two occasions, a wash - out period of at least 28 days is allowed to rejuvenate the 
harvested stratum corneum. 
Pharmacodynamic (PD) studies are also performed to estimate the bioavailability 
and bioequivalence of drugs from topically applied semisolids. In this case, 
known therapeutic responses from drug products such as skin blanching due to 
vasoconstrictor effects caused by corticosteroids and transepidermal water loss 
caused by retinoids are measured and compared between the test and reference 

288 SEMISOLID DOSAGES: OINTMENTS, CREAMS, AND GELS 
products. Comparative clinical studies are rarely conducted due to the diffi culties 
involved in performing the study, variability in study results, and their poor 
sensitivity. 
4.2.2.5 Typical Pharmacopeial/Commercial Examples 
The vast majority of topical ointments and creams are meant for dermatological 
applications. They are used to treat various skin conditions such as eczema, dermatitis, 
allergies, infl ammatory and pruritic manifestations, minor skin wounds, pain, 
insect bites, psoriasis, herpes and other infections of the skin (e.g., impetigo), acne, 
and precancerous and cancerous skin growths. Similarly, ophthalmic conditions such 
as infections, infl ammation, allergy, and dry - eye symptoms are treated with semisolid 
preparations. Products are also available for certain eye examinations. Vaginal preparations 
are available for treating genital herpes, yeast infections, and vaginosis 
caused by bacteria and to reduce menopausal symptoms (e.g., vaginal dryness), and 
rectal preparations are available for treating minor pain, itching, swelling, and discomfort 
caused by hemorrhoids and other problems of the anal area. Tables 6 and 
7 show some of the commercially available compendial ointment and cream preparations 
used for treating various topical ailments. 
4.2.3 GELS 
4.2.3.1 Defi nition 
Gels are semisolid preparations that contain small inorganic particles or large 
organic molecules interpenetrated by a liquid. Gels made of inorganic materials are 
usually two - phase systems where small discrete particles are dispersed throughout 
the dispersion medium. When the particle size of the dispersed phase is larger, they 
are referred to as magmas. Gels made of organic molecules are single - phase systems, 
where no apparent physical boundary is seen between the dispersed phase and the 
dispersion medium. In most cases, the dispersion medium is aqueous. Hydroalcoholic 
or oleaginous dispersion media are also used in some cases. Unlike dispersed 
systems like suspensions and emulsions, movement of the dispersed phase is 
restricted in gels because of the solvated organic macromolecules or interconnecting 
three - dimensional networks of particles. 
Gels are attractive delivery systems as they are simple to manufacture and 
suitable for administering drugs through skin, oral, buccal, ophthalmic, nasal, 
otic, and vaginal routes. They also provide intimate contact between the drug and 
the site of action or absorption. With the advancement in polymer science, gel - based 
systems that respond to specifi c biological or external stimuli like pH, temperature, 
ionic strength, enzymes, antigens, light, magnetic fi eld, ultrasound, and electric 
current are being designed and evaluated as smart delivery systems for various 
applications. 
4.2.3.2 Characteristics 
Gels may appear transparent or turbid based on the type of gelling agent used. They 
exhibit different physical properties, namely, imbibition, swelling, syneresis, and 

TABLE 6 Examples of Compendial/Commercial Ointments 
Drug a Category Indication 
Commercial 
Names 
Strength(s) 
Available 
(%) 
Acyclovir Antiviral Genital herpes, 
herpes infections 
of the skin, and 
oral herpes 
Zovirax 5 
Atropine 
sulfate (oph) 
Mydriatic Relax muscles in the 
eye, treat 
infl ammation of 
certain parts of 
the eye (uveal 
tract), and used 
for certain eye 
exams 
Atropisol, 
Isopto 
Atropine 
0.5, 1 
Bacitracin First - aid antibiotic Treat or prevent skin 
infections 
Baciguent 
Oint, 
Bacitracin 
Top 
500 units/g 
Bacitracin 
(oph) 
Antibiotic Treat or prevent eye 
infections 
Ak - Tracin, 
Bacticin 
500 units/g 
Benzocaine Antipruritic and 
local anesthetic 
Itching, minor skin 
wound pain, and 
insect bites 
Americaine 20 
Ciprofl oxacin 
(oph) 
Antibiotic Eye infections Ciloxan 0.3 
Clobetasol 
propionate 
Anti - infl ammatory 
agent 
Relieve 
infl ammatory 
and pruritic 
manifestations of 
corticosteroid - 
responsive 
dermatoses 
Temovate 
Ointment 
0.05 
Erythromycin Antibiotic Treatment of acne Akne - Mycin 2.0 
Erythromycin 
(oph) 
Antibiotic Infections of eye or 
ear 
Erythromycin 
Ophthalmic 
0.5 
Gentamicin 
sulfate (oph) 
Antibacterial Infections of eye or 
ear 
Gentamicin 
Sulfate 
0.3 
Hydrocortisone Anti - infl ammatory 
agent 
Minor pain, itching, 
swelling, and 
discomfort caused 
by hemorrhoids 
and other 
problems of 
anal area 
Cortaid, 
Anusol - HC, 
Proctosol 
HC 
2.5 
Mupirocin Antibiotic Treat certain skin 
infections (e.g., 
impetigo) 
Bactroban 2.0 
Sodium 
chloride 
(oph) 
Miscellaneous Treat fl uid 
accumulation in 
cornea of eye 
causing swelling 
Muro - 128, 
Sochlor 
2.0 
a oph: ophthalmic ointment. 
GELS 289

290 SEMISOLID DOSAGES: OINTMENTS, CREAMS, AND GELS 
TABLE 7 Examples of Compendial/Commercial Creams 
Drug a Category Indication 
Commercial 
Products 
Strength(s) 
Available 
(%) 
Alclometasone 
dipropionate 
Anti - 
infl ammatory 
Eczema, dermatitis, 
allergies, and rash 
Aclovate 0.05 
Amcinonide Anti - 
infl ammatory 
Lymphomas of the skin, 
atopic dermatitis, 
contact dermatitis, 
and skin rash 
Cyclocort 0.1 
Amphotericin B Antifungal 
antibiotic 
Treat skin infection due 
to a Candida yeast 
and diaper rash 
Fungizone 3.0 
Anthralin Keratolytic Long - term psoriasis Dritho - Calp, 
Psoriatec 
0.5, 1.0 
Betamethasone 
dipropionate 
Anti - 
infl ammatory 
Eczema, dermatitis, 
allergies, and rash 
Diprolene 
AF 
0.05 
Butoconazole 
nitrate (vag) 
Antifungal Vaginal yeast infections Gynazole - 1 2.0 
Clindamycin 
phosphate 
(vag) 
Antibiotic Vaginosis caused by 
bacteria 
Clindesse 2.0 
Clobetasol 
propionate 
Anti - 
infl ammatory 
Infl ammatory 
and pruritic 
manifestations of 
corticosteroid - 
responsive 
dermatoses 
Temovate E 
Cream 
0.05 
Clotrimazole Antifungal Ringworm of groin 
area, athlete ’ s foot, 
ringworm of the body, 
fungal infection of 
the skin with yellow 
patches, skin infection 
due to a candida 
yeast, and diaper rash 
Lotrimin 1.0 
Crotamiton Scabicidal and 
antipruritic 
Scabies infection and 
itching 
Eurax 10.0 
Desoximetasone Anti - 
infl ammatory 
Eczema, dermatitis, 
allergies, and rash 
Topicort 0.25 
Dienestrol Estrogen Reduce menopause 
symptoms such as 
vaginal dryness 
Ortho - 
Dienestrol 
0.01 
Difl orasone 
diacetate 
Anti - 
infl ammatory 
Eczema, dermatitis, 
allergies, and rash 
Apexicon, 
Maxifl or 
0.05 
Fluocinonide Anti - 
infl ammatory 
and 
antipruritic 
Psoriasis, eczema, 
dermatitis, allergies, 
and rash 
Lidex, Vanos 0.05, 0.10 
Fluorouracil Anticancer Precancerous and 
cancerous skin 
growths 
Fluoroplex, 
Carac, 
Efudex 
0.5, 1.0, 5.0 

thixotropy. Imbibition refers to the uptake of water or other liquids by gels without 
any considerable increase in its volume. Swelling refers to the increase in the volume 
of gel by uptake of water or other liquids. This property of most gels is infl uenced 
by temperature, pH, presence of electrolytes, and other formulation ingredients. 
Syneresis refers to the contraction or shrinkage of gels as a result of squeezing out 
of dispersion medium from the gel matrix. It is due to the excessive stretching of 
macromolecules and expansion of elastic forces during swelling. At equilibrium, the 
system still maintains its physical stability because the osmotic forces of swelling 
balance the expanded elastic forces of macromolecules. On cooling, the osmotic 
pressure of the system decreases and therefore the expanded elastic forces return 
to normal. This results in shrinkage of the stretched molecules and squeezing of 
dispersion medium from the gel matrix. Addition of osmotic agents such as sucrose, 
glucose, and other electrolytes helps in retaining higher osmotic pressure even at 
lower temperatures and avoids syneresis of gels. Thixotropy refers to the non - 
Newtonian fl ow nature of gels, which is characterized by a reversible gel - to - sol 
formation with no change in volume or temperature [18] . 
4.2.3.3 Classifi cation 
Gels are classifi ed as hydrogels and organogels based on the physical state of the 
gelling agent in the dispersion. Hydrogels are prepared with water - soluble materials 
or water - dispersible colloids. Organogels are prepared using water - insoluble oleaginous 
materials. 
Hydrogels Natural and synthetic gums such as tragacanth, sodium alginate, and 
pectin, inorganic materials such as alumina, bentonite, silica, and veegum, and 
organic materials such as cellulose polymers form hydrogels in water. They may 
either be dispersed as fi ne colloidal particles in aqueous phase or completely dissolve 
in water to gain gel structure. Gums and inorganic gelling agents form gel 
structure due to their viscosity - increasing nature. Organic gelling agents which are 
generally high - molecular - weight cellulose polymer derivatives produce gel structure 
Drug a Category Indication 
Commercial 
Products 
Strength(s) 
Available 
(%) 
Halcinonide Anti - 
infl ammatory 
and 
antipruritic 
Eczema, dermatitis, 
allergies, and rash 
Halog 0.1 
Mometasone 
furoate 
Anti - 
infl ammatory 
Eczema, dermatitis, 
allergies, and rash 
Elocon 0.1 
Naftifi ne 
hydrochloride 
Antifungal Jock itch, athlete ’ s feet, 
or ringworm 
Naftin 1 
Nystatin Antifungal Fungal skin infections Mycostatin 100,000 
units/g 
a vag, vaginal cream. 
TABLE 7 Continued 
GELS 291

292 SEMISOLID DOSAGES: OINTMENTS, CREAMS, AND GELS 
because of their swelling and chain entanglement properties. The swollen molecular 
chains are held together by secondary valence forces, which help in retaining their 
gel structure. The physical strength of the gel structure is based on the quantity of 
gelling agent, nature and molecular weight of gelling agent, product pH, and gelling 
temperature. Generally high - molecular - weight polymers at higher concentrations 
produce thick gels. The gel - forming temperature ( gel point ) varies with different 
polymers. Generally natural gums form gel at lower temperatures. Gelatin, a natural 
protein polymer, forms gel at about 30 ° C. If the temperature is increased, gel consistency 
is not obtained even at higher concentrations of gelatin. On the other hand, 
polymers such as methylcellulose gain gel structure only when the temperature is 
above 50 ° C due to its decreased solubility and precipitation. Knowledge of the gel 
point for each gelling agent is therefore essential for preparing physically stable 
hydrogels. 
Organogels Organogels are also known as oleaginous gels. They are prepared 
using water - insoluble lipids such as glycerol esters of fatty acids, which swell in water 
and form different types of lyotropic liquid crystals. Widely used glycerol esters of 
fatty acids include glycerol monooleate, glycerol monopalmito stearete, and glycerol 
monolinoleate. They generally exist as waxes at room temperature and form cubic 
liquid crystals in water and increase the viscosity of dispersion. Waxes such as carnauba 
wax, esparto wax, wool wax, and spermaceti are used in cosmetic organogel 
preparations. A large quantity of water is entrapped between the three - dimensional 
lipid bilayers. The equilibrium water content in organogels is about 35%. The structural 
properties of the lipid, quantity of water in the system, solubility of drug 
incorporated, and external temperature infl uence the nature of the liquid crystalline 
phase. The bipolar nature of organogels allows incorporation of both hydrophilic 
and lipophilic drugs. Release rates can be controlled by altering the hydrophilic and 
hydrophobic components. Biodegradability of these waxes by the lipase enzyme in 
the body makes organogels suitable for parenteral administration. 
The water present in the gel framework can be completely removed with some 
gelling agents. Gelatin sheets, acacia tears, and tragacanth ribbons are generally 
prepared by removal of water from their respective gel matrix. These dehydrated 
gel frameworks are called as xerogels. 
4.2.3.4 Stimuli - Responsive Hydrogels 
The three - dimensional networks of hydrophilic polymers absorb a large quantity of 
water and form soft structures which resemble biological tissues. Swelling properties 
of these hydrogels can be altered by various physicochemical parameters. Physical 
factors such as temperature, pH, and ionic strength of the swelling medium 
and chemical factors such as the structure of polymer (linear/branched) and chemical 
modifi cations (cross - linking) can be altered to tailor their swelling rate. This 
feature makes them very attractive for drug delivery and biomedical applications 
[19 – 23] . 
pH-Responsive Hydrogels Some polymers show pH - dependent swelling and 
gelling characteristics in aqueous media. A polymer that exhibits such phase transition 
properties is very useful from the point of drug delivery. Methacrylic acids 

(e.g., carbomers) that contain many carboxylic acid groups exist as solution at lower 
pH conditions. When the pH is increased, they undergo a sol - to - gel transition. This 
is because of the increase in the degree of ionization of acidic carboxylic groups at 
higher pH conditions, which in turn results in electrostatic repulsions between 
chains and, increased hydrophilicity and swelling. Conversely, polymers that contain 
amine - pendant groups swell at lower pH environment due to ionization and repulsion 
between polymer chains. The ionic strength of surrounding fl uids signifi cantly 
infl uences the equilibrium swelling of these pH - responsive polymers. Higher ionic 
strength favors gel – counter ionic interactions and reduces the osmotic swelling 
forces. 
Thermoresponsive Hydrogels A dispersion which exists as solution at room temperature 
and transforms into gel on instillation into a body cavity can improve the 
administration mode and help in modulating the drug release. Many polymers with 
thermoresponsive gelling properties are currently being synthesized and evaluated. 
A triblock copolymer that consists of polyethylene glycol – polylactic acid, glycolic 
acid – polyethylene glycerol (PEG – PLGA – PEG) is solution at room temperature 
and gels at body temperature. Poloxamers, which are made of triblock poly(ethylene 
oxide) – poly(propylene oxide) – poly(ethylene oxide), exhibit gelatin properties at 
body temperatures. Similarly, xyloglucan and xanthan gum aqueous dispersions are 
solutions at room temperature and become gel at body temperature. These are 
considered convenient alternatives for rectal suppository formulations which usually 
cause mucosal irritations due to their physical state. The physicochemical properties 
of these chemically modifi ed thermoresponsive hydrogels are altered by changing 
the ratio of hydrophilic and hydrophobic segments, block length, and polydispersity. 
ReGel by MacroMed contains a triblock copolymer PLGA – PEG – PLGA, undergoes 
sol - to - gel transition on intratumoral injection, and releases paclitaxel for six 
weeks. 
Ionic-Responsive Hydrogels Administration of sodium alginate aqueous drops 
into the eye results in alginate gelation due to its interaction with calcium ions in 
the tear fl uid. Alginate with high guluronic acid and deacetylated gellan gum 
(Gelrite) show sol - to - gel conversions in the eye due to their interaction with cations 
in the tear fl uid. Timolol maleate sterile ophthalmic gel - forming solution (Timoptic - 
XE) that contains Gelrite gellan gum is commercially available. 
4.2.3.5 Gelling Agents 
A large number of gelling agents are commercially available for the preparation of 
pharmaceutical gels. In general, these materials are high - molecular - weight compounds 
obtained from either natural sources or synthetic pathways. They are water 
dispersible, possess swelling properties, and improve the viscosity of dispersions. An 
ideal gelling agent should not interact with other formulation components and 
should be free from microbial attack. Changes in the temperature and pH during 
preparation and preservation should not alter its rheological properties. In addition, 
it should be economic, readily available, form colorless gels, provide cooling sensation 
on the site of application, and possess a pleasant odor. Based on these factors, 
gelling agents are selected for different formulations. Table 8 summarizes the 
GELS 293

294 SEMISOLID DOSAGES: OINTMENTS, CREAMS, AND GELS 
TABLE 8 Some Compendial Gelling Agents Used in Gels 
Name 
Molecular 
Weight 
Gelling 
Strength 
(%) Synonyms 
Offi cial 
Compendia 
Bentonite 359.16 10 – 20 Magnabrite, 
mineral soap, 
Polargel, Veegum 
HS 
BP, JP, PhEur, 
USPNF 
Carbomer 7 . 10 5 – 4 . 10 9 0.5 – 2.0 Acritamer, 
Carbopol, 
polyacrylic acid 
BP, PhEur, 
USPNF 
Carboxymethyl 
cellulose sodium 
90,000–700,000 3.0–6.0 Akucell, Aquasorb, 
Sodium CMC, 
Tylose CB 
BP, JP, PhEur, 
USPNF 
Carrageenan . 1,000,000 0.3 – 2.0 Gelcarin, Genu, 
Hygum Marine 
colloids 
USPNF 
Colloidal silicon 
dioxide 
60.08 2.0 – 10.0 Aerosil, colloidal 
silica, fumed 
silica 
BP, PhEur, 
USPNF 
Gelatin 15,000 – 25,0000 10.0 – 20.0 Cryogel, Solugel BP, JP, PhEur, 
USPNF 
Glyceryl behenate 1059.8 1.0 – 15.0 Docosanoic acid, 
glycerol behenate 
BP, PhEur, 
USPNF 
Guar gum . 220,000 1.0 – 5.0 Galactosal, Guar 
fl our, Jaguar gum, 
Meyproguar 
BP, PhEur, 
USPNF 
Hydroxypropyl 
cellulose 
50,000–1,250,000 2.0 – 5.0 Hyprolose, klucel, 
Methocel 
BP, JP, PhEur, 
USPNF 
Hydroxypropylmethyl 
cellulose 
10,000–1,500,000 1.0 – 10.0 Hypromellose BP, JP, PhEur, 
USPNF 
Magnesium aluminum 
silicate 
— 5.0 – 15.0 Veegum, 
aluminosilicic 
acid, Carrisorb, 
Magnabite 
BP, PhEur, 
USPNF 
Methylcellulose 10,000 – 220,000 1.0 – 5.0 Benecel, Methocel, 
Metolose 
BP, JP, PhEur, 
USPNF 
Poloxamer 2090 – 17,400 15.0 – 20.0 Lutrol, Monolan, 
Pluronic, 
Supronic 
BP, phEur, 
USPNF 
Polyvinyl alcohol . 20,000 – 200,000 2.5 – 10.0 Airvol, Elvanol, 
PVA, vinyl 
alcohol 
USP 
Povidone 2500–3,000,000 2.0 – 20.0 Kollidon, Plasdone, 
Polyvidone, PVP 
BP, JP, PhEur, 
USPNF 
Sodium alginate 20,000 – 240,000 10.0 – 20.0 Algin, alginic acid, 
sodium salt, 
Protanal 
BP, PhEur, 
USPNF 
Tragacanth 840,000 1.0 – 8.0 Gum Benjamin, 
Gum dragon, 
Trag, Tragant 
BP, JP, PhEur, 
USPNF 
Note : BP, British Pharmacopoeia; JP, Japanese Pharmacopoeia; PhEur, European Pharmacopoeia; USPNF, U.S. 
Pharmacopeia/National Formulary 

molecular weight, gelling strength, synonyms, and compendial status of some of 
these agents. 
The following sections briefl y describe the source, physicochemical properties, 
formulation, and preservation of some pharmacopeial gelling agents. 
Alginic Acid Alginic acid is tasteless and odorless and occurs as a yellowish white 
fi brous powder. The main source for this naturally occurring hydrophilic colloidal 
polysaccharide is different species of brown sea weed, known as Phaeophyceae. It 
consists of a mixture of d - mannuronic acid and l - glucuronic acids. It is used in gels 
due to its thickening and swelling properties. Alginic acid is insoluble in water; 
however, it absorbs 200 – 300 times its own weight of water and swells. The viscosity 
of alginic acid gels can be altered by changing the molecular weight and concentration. 
Addition of calcium salts increases the viscosity of alginic acid gels. Its viscosity 
decreases at higher temperature. Depolymerization due to microbial attack also 
results in viscosity reduction. Inclusion of an antimicrobial preservative avoids 
depolymerization and viscosity reduction during storage [6] . 
Bentonite Bentonite is a naturally occurring colloidal hydrated aluminum silicate 
and contains traces of calcium, magnesium, and iron. It is odorless, available as fi ne 
crystalline powder, and is cream to grayish in color. The particles are negatively 
charged. Its high water uptake and swelling and thickening properties make it suitable 
for preparing gels. It swells to about 12 - fold when it comes in contact with 
water. The viscosity of bentonite dispersion increases with increase in concentration. 
The gel - forming properties increase with addition of alkaline materials such as 
magnesium oxide and decrease with addition of alcohol or electrolytes. Use of hot 
water and stirring improve wetting and dispersion of bentonite particles in the 
preparation of the gel. Mixing with magnesium oxide or zinc oxide prior to addition 
helps in dispersion of bentonite in water. Prior trituration of bentonite with glycerin 
also helps in easy dispersibility in water. These dispersions are generally left for 
about 24 h to complete the swelling process. At lower concentration (10%) bentonite 
suspension exhibits the properties of shear thinning systems and at high concentrations 
(about 50 – 60%) it forms gel with fi nite yield strength [24] . 
Carbomer Carbomers are one of the widely used gelling agents in topical preparations 
due to their extensive swelling properties. They are obtained by cross - 
linking acrylic acid with allyl sucrose or allyl pentaerythritol. Various grades with 
varying degree of cross - linking and molecular weight are commercially available. 
Carbomers are generally available as hygroscopic powders, are white in color, and 
possess a characteristic odor. Presence of about 60% carboxylic acid in its composition 
makes them acidic. Carbomer 934P, 971P, 974P, and so on, are used for preparing 
clear gels. Aqueous dispersions of carbomers are usually low viscous, and on 
neutralization they form high - viscous gels. Basic materials such as sodium hydroxide, 
potassium hydroxide, sodium bicarbonate, and borax are being used for neutralizing 
carbomer dispersions. About 0.4 g of sodium hydroxide is used to neutralize 
1 g of carbomer dispersion. The viscosity of gels depends on the molecular weight 
of carbomer and its degree of cross - linking. Inclusion of antioxidants, protection 
from light, and preservation at room temperature help in retaining their viscosity 
for prolonged periods. Microbial stability of carbopol gels can be improved by 
GELS 295

296 SEMISOLID DOSAGES: OINTMENTS, CREAMS, AND GELS 
adding antimicrobial preservatives. These gels are prone to discoloration in the 
presence of large amounts of electrolytes, strong acids, and cationic polymers. Glass, 
plastic, and resin - lined containers which possess good corrosion - resistant properties 
are used for packing carbomer gels [6] . 
Carboxymethylcellulose Calcium (Calcium CMC) A calcium salt of polycarboxymethyl 
ether of cellulose, calcium CMC is obtained by carboxymethylation of 
cellulose and conversion into calcium salt. Different molecular grades are prepared 
by changing the degree of carboxymethylation. It is available as a fi ne powder, white 
to yellowish white in color, and hygroscopic in nature. Calcium CMC has swelling 
and viscosity - enhancing properties in water. It can swell twice its volume in water 
[25] . 
Carboxymethylcellulose Sodium (Sodium CMC) A sodium salt of polycarboxymethyl 
ether of cellulose, sodium CMC is obtained by treating alkaline cellulose 
with sodium monochloroacetate. It is available as white - colored granular powder. 
Various viscosity grades of sodium CMC commercially available basically differ in 
their degree of substitution. The degree of substitution represents the average 
number of hydroxyl groups that are substituted per anhydroglucose unit. It is readily 
dispersible in water and forms clear gels. The aqueous solubility of CMC sodium is 
governed by the degree of substitution. Higher concentrations generally yield 
thicker gels. Although the viscosity of gels is stable over a wide range of pH (4 – 10), 
a fall in pH below 2 or a rise to above 10 results in physical instability and viscosity 
reduction. Higher viscosity is obtained at neutral pH conditions. Exposure of gels 
to higher temperature also results in viscosity reduction. Preservation at optimum 
temperature and inclusion of an antimicrobial preservative improve the physical 
and microbial stability of CMC sodium gels [25] . 
Carrageenan Extraction of some red seaweed belonging to the Rhodophyceae 
class with water or aqueous alkali yields carrageenan. It is a hydrocolloid and mainly 
contains sodium, potassium, calcium, magnesium, and ammonium sulfate esters of 
galactose and copolymers of 3, 6 - anhydrogalactose. They differ in their ester sulfate 
and anhydrogalactose content. It is available as a coarse to fi ne powder which is 
yellow to brown in color. It is odorless and tasteless. Carrageenan is soluble in hot 
water and forms gels at 0.3 – 2.0%. . - Carrageenan and . - carrageenan show good 
gelling properties [26] . 
Colloidal Silicon Dioxide Colloidal silicon dioxide is a fumed silica obtained by 
vapor hydrolysis of chlorosilanes. It is available as nongritty amorphous powder 
which is bluish white in color. It is tasteless and odorless and possesses low tapped 
density. Although insoluble in water, it readily forms a colloidal dispersion due to 
its fi ne particle size, higher surface area, and water - adsorbing properties. The bulk 
density and particle size of colloidal silicon dioxide can be altered by changing the 
method of manufacture. Transparent gels can be formed by mixing with other materials 
that possess similar refractive index. Under acidic and neutral pH conditions, 
it shows viscosity - increasing properties. This property is lost at higher pH conditions 
because of its dissolution. Viscosity of gels is not generally affected by temperature 
[27] . 

Ethylcellulose Ethylcellulose is a synthetic polymer made of . - anhydroglucose 
units connected by acetyl linkages. It is obtained by ethylating alkaline cellulose 
solution with chloroethane. Ethylcellulose is available as a free - fl owing powder 
which is tasteless and white in color. Although it is insoluble in water, it is incorporated 
into topical preparations due to its viscosity - enhancing properties. Ethanol or 
a mixture of ethanol and toluene (2 : 8) is used as a solvent. A decrease in the ratio 
of alcohol increases the viscosity. The viscosity of the dispersion is increased by 
increasing the concentration of ethylcellulose or by using a high - molecular - weight 
material. As ethylcellulose is prone to photo - oxidation at higher temperature, and 
gels are prepared and preserved at room temperature and dispensed in airtight 
containers [28] . 
Gelatin Gelatin is a protein obtained by acid or alkali hydrolysis of animal tissues 
that contain large amounts of collagen. Based on the method of manufacture, it is 
named type A or type B gelatin. Type A is obtained by partial acid hydrolysis and 
type B is obtained by partial alkaline hydrolysis. They differ in their pH, density, 
and isoelectric point. Gelatin is available as yellow - colored powder or granules. It 
swells in water and improves the viscosity of dispersions. Different molecular weights 
and particle size grades are commercially available. Gels can be prepared by dissolving 
gelatin in hot water and cooling to 35 ° C. Temperature greatly infl uences the 
viscosity and stability of gelatin dispersions. It transforms to a gel at temperatures 
above 40 ° C and undergoes depolymerization above 50 ° C. The viscosity of gelatin 
gel is also affected by microbes [29] . 
Guar Gum Guar gum is a high - molecular - weight polysaccharide obtained from 
the endosperms of guar plant. It mainly contains d - galactan and d - mannan. It is 
available as powder which is odorless and white to yellowish white in color. It readily 
disperses in water and forms viscous gels. The viscosity of gel is infl uenced by the 
particle size of material, pH of the dispersion, rate of agitation, swelling time, and 
temperature. Viscosity reduces on long - time heating. Maximum viscosity can be 
achieved within 2 – 4 h. Gels are stable at pH between 7 and 9 and show liquifi cation 
below pH 7. Addition of antimicrobial preservatives improves the microbial stability 
of guar gum gels. Rheological properties of these gels can be modifi ed by incorporating 
other plant hydrocolloids such as tragacanth and xanthan gum [30] . 
Hydroxyethyl Cellulose ( HEC) HEC is a partially substituted poly(hydroxyethyl) 
ether of cellulose. It is obtained by treating alkali cellulose with ethylene oxide. HEC 
is available as a powder and appears light tan to white in color. Different viscosity 
grades of HEC are commercially available which differ in their molecular weights. 
Clear gels are prepared by dissolving HEC in hot or cold water. Dispersions can be 
prepared quickly by altering the stirring rate of dispersion, temperature, and pH. 
Slow stirring at room temperature during the initial stages favors wetting. Increasing 
the temperature at this stage increases the rate of dispersion. Although HEC dispersions 
are stable over a wide pH range, maintaining basic pH improves the dispersion. 
The preservation temperature, formulation pH, and microbial attack infl uence the 
rheological properties of HEC dispersions. Viscosity reduces at higher temperature, 
but reverts to the original value on returning to room temperature. Lower 
and higher pH of the vehicle usually results in hydrolysis or oxidation of HEC, 
GELS 297

298 SEMISOLID DOSAGES: OINTMENTS, CREAMS, AND GELS 
respectively. Some of the enzymes secreted by microbes decrease the viscosity of 
HEC dispersions. The presence of higher levels of electrolytes may also destabilize 
HEC dispersions. Inclusion of a suitable antimicrobial preservative is essential to 
retain the viscosity of HEC gels [31] . 
Hydroxyethylmethyl Cellulose ( HEMC) HEMC is a partially o - methylated and 
o - (2 - hydroxyethylated) cellulose. It is available as powder or granules which are 
white, grayish white, or yellowish white in color. Various viscosity grades of HEMC 
are commercially available, and form viscous colloidal dispersions or gels in cold 
water which has a pH in the range of 5.5 – 8 [6] . 
Hydroxypropyl Cellulose ( HPC) HPC is a partially substituted poly(hydroxypropyl) 
ether of cellulose. It is obtained by treating alkali cellulose with propylene oxide at 
higher temperatures. It is available as tasteless and odorless powder which is yellowish 
or white in color. Different viscosity grades are commercially available. 
Gradual addition of HPC powder into vigorously stirred water yields clear viscous 
dispersions or gels below 38 ° C. Increase in temperature destabilizes the dispersion 
and leads to precipitation. The viscosity of dispersions can be increased by increasing 
the concentration of HPC or by using high - molecular - weight grades. Inclusion 
of a cosolvent such as dichloromethane or methane produces viscous dispersion or 
gels with modifi ed texture. The viscosity of HPC dispersions can be increased by 
mixing with an anionic polymer. High concentrations of electrolytes destabilize 
HPC dispersions. HPC dispersions are neutral in pH (6 – 8). They undergo acid 
hydrolysis at lower pH and oxidation at higher pH. Both processes decrease the 
dispersion viscosity. In addition, certain enzymes produced by microbes degrade 
HPC and reduce its viscosity. Addition of an antimicrobial preservative is therefore 
recommended for HPC gels. Preservation of these gels from light can further 
improve its physical stability [25] . 
Hydroxypropylmethyl Cellulose ( HPMC) HPMC is a partly o - methylated and 
o - (2 - hydroxypropylated) cellulose obtained by treating alkali cellulose with chloromethane 
and propylene oxide. It is available as odorless and tasteless granular or 
fi brous powder which is creamy white or white in color. HPMC is soluble in cold 
water. Aqueous dispersions are prepared by dispersing material in about 25% hot 
water (80 ° C) under vigorous stirring. On complete hydration of HPMC, a suffi cient 
quantity of cold water is added and mixed. The gel point of HPMC dispersions varies 
from 50 to 90 ° C. Gels are stable over a wide pH range (3 – 11). The viscosity HPMC 
dispersions depends on the concentration of material used, its molecular weight, 
vehicle composition, presence of preservatives, and so on. Viscous gels can be prepared 
using high concentrations of high - molecular - weight grades. Inclusion of 
organic solvents such as ethanol or dichloromethane improves the viscosity. Addition 
of an antimicrobial preservative (e.g., benzalkonium chloride) minimizes microbial 
spoilage of HPMC gels [25] . 
Glyceryl Behenate Glyceryl behenate is a mixture of glycerides of fatty acids 
which is obtained by esterifi cation of glycerin with behenic acid. It may also contain 
arachidic acid, stearic acid, erucic acid, lignoceric acid, and palmitic acid. It is available 
as a waxy mass or powder, possesses a faint odor, and is white in color. It is 

practically insoluble in water and soluble in dichloromethane and chloroform. It is 
used as a viscosity - increasing agent in silicon gels [6] . 
Glyceryl Monooleate ( GMO) GMO is a mixture of glycerides of fatty acids 
obtained by esterifi cation of glycerol with oleic acid. It may also contain linoleic 
acid, palmitic acid, stearic acid, linolenic acid, arachidic acid, and eicosenoic acid. It 
is available as a partially solidifi ed or oily liquid. GMO is insoluble in water. Self - 
emulsifying grades that contain an anionic surfactant disperse easily and swell in 
water. The nonemulsifying grades are used as emollients in topical preparations and 
self - emulsifying grades are used as emulsifi ers in aqueous emulsions [6] . 
Magnesium Aluminum Silicate ( MAS) MAS is also known as veegum. It is a 
polymeric complex of magnesium, aluminum, silicon, oxygen, and water and is 
obtained from silicate ores. Based on the ratio of aluminum and magnesium and 
viscosity, it is classifi ed as types IA, IB, IC, and IIA. It is available as fi ne powder 
that is odorless, tasteless, and off - white to creamy white in color. Although MAS is 
insoluble in water, it swells to a large extent and produces viscous colloidal dispersions. 
Use of higher concentration, addition of electrolytes, and heating of dispersion 
usually improve the viscosity [32] . 
Methylcellulose ( MC) MC is a long - chain cellulose polymer with methoxyl substitutions 
at positions 2, 3, and 6 of the anhydroglucose ring. It is synthesized by 
methylating alkali cellulose with methyl chloride. The degree of substitution of 
methoxy groups infl uences the molecular weight, viscosity, and solubility characteristics 
of MC. It is available as powder or granules and is odorless, tasteless, and white 
to yellowish white in color. MC is insoluble in hot water but slowly swells and forms 
viscous colloidal dispersions in cold water. Gels can be prepared by initially mixing 
the methylcellulose with half the volume of hot water ( . 70 ° C) followed by addition 
of the remaining volume of cold water. Viscosity of these dispersions can be increased 
by using high - concentration or high - molecular - weight grades of methylcellulose. 
Higher processing or preservation temperatures reduce the viscosity of formulations, 
which regain their original state on cooling to room temperature. MC aqueous 
dispersions show pH values of 5 – 8. Reduction in pH to less than 3 leads to acid - 
catalyzed hydrolysis of glucose – glucose linkages and results in low viscosity. Antimicrobial 
preservatives are generally included to enhance the microbial stability of 
dispersions. Salting out is observed when high concentrations of electrolytes are 
added to methylcellulose dispersions. The viscosity of methylcellulose dispersions is 
also infl uenced by the presence of formulation excipients and drugs [25] . 
Poloxamer Poloxamers are copolymers of ethylene oxide and propylene oxide. 
Different molecular weight grades that are different in physical form, solubility, and 
melting point are available. Poloxamer 124 is a colorless liquid, whereas poloxamers 
188, 237, 338, and 407 are solids at room temperature. All poloxamer grades are 
freely soluble in water and form gels at higher concentrations. The pH of aqueous 
liquids ranges between 5 and 7.5 [33] . 
Polyethylene Oxide Polyethylene oxide is a nonionic homopolymer of ethylene 
oxide synthesized by polymerization of ethylene oxide. It is available as a free - 
GELS 299

300 SEMISOLID DOSAGES: OINTMENTS, CREAMS, AND GELS 
fl owing powder white to off - white in color with a slight ammonia odor. Various 
molecular weight grades of polyethylene oxide are commercially available. They 
swell in water and form viscous dispersions or gels based on the concentration and 
grade used. Inclusion of alcohol improves the rheological stability of polyethylene 
oxide dispersions [6] . 
Polyvinyl Alcohol ( PVA ) PVA is a synthetic polymer prepared by hydrolysis of 
polyvinyl acetate. It is available as a granular powder which is odorless and white 
in color. Mixing with water at room temperature, heating for about 5 min at 90 ° C, 
followed by cooling with constant mixing yield aqueous dispersions or gels. Higher 
viscosities can be obtained by using high - viscosity grades. Addition of borax improves 
the gelling properties of PVA, whereas inorganic salts destabilize these dispersions. 
The pH of PVA dispersions ranges between 5 and 8. Physical and chemical decompositions 
occur at lower and higher pH conditions. Incorporation of an antimicrobial 
preservative and storage at room temperature improve its stability [6] . 
Povidone Povidone is a synthetic polymer consisting of 1 - vinyl - 2 - pyrrolidinone 
units. It is available as a fi ne powder and appears white to creamy - white in color. 
Various molecular weight grades of povidone are available which differ in their 
degree of polymerization. Povidone is soluble in water and forms viscous solutions 
and gels based on the concentration and viscosity grade used. Decomposition occurs 
when dispersions are heated to about 150 ° C. The pH of aqueous dispersions ranges 
from 3 to 7. The microbial stability of povidone aqueous dispersions can be increased 
by adding preservatives [6] . 
Propylene Carbonate ( PC) PC is prepared by reacting propylene chlorohydrin 
with sodium bicarbonate. It is available as a clear liquid with a faint odor. Mixtures 
of PC and propylene glycol are good solvents for corticosteroids in topical preparations. 
It is incompatible with strong acids, bases, and amines. The pH of 10% aqueous 
dispersion is 6.0 – 7.5 [34] . 
Propylene Glycol Alginate ( PGA) PGA is a propylene glycol ester of alginic acid 
obtained by treating alginic acid with propylene oxide. It is available as granular or 
fi brous powder which is odorless, tasteless, and white to yellowish - white in color. 
PGA is soluble in water and forms viscous colloidal dispersions. The viscosity of 
these dispersions is based on the concentration of PGA, temperature, and pH. Its 
aqueous solubility decreases at higher temperatures. The aqueous dispersions are 
acidic in nature and more stable at pH 3 – 6. Higher pH leads to saponifi cation. As 
these dispersions are prone to microbial spoilage, antimicrobial preservatives are 
generally included [6] . 
Sodium Alginate Sodium alginate is obtained by extraction of alginic acid from 
brown seaweed followed by neutralization with sodium bicarbonate. Alginic acid is 
composed of d - mannuronic acid and l - guluronic acid. It is available as a powder 
which is tasteless, odorless, and white to yellowish - brown in color. Sodium alginate 
forms viscous gels in water. Dispersing agents such as glycerol, propylene glycol, 
sucrose, and alcohol are added to improve dispersion. The presence of low concentration 
of electrolytes improves the viscosity, whereas at high concentrations salting 

out takes place. The viscosity of gel is based on the concentration of sodium alginate, 
temperature, pH, and other additives. Various viscosity grades of sodium alginate 
are commercially available. Aqueous dispersions are stable at pH 4 – 10. Precipitation 
or decrease in viscosity is observed when the pH is below or above these values. 
Autoclaving or heating above 70 ° C results in depolymerization and decrease in 
viscosity. Inclusion of preservative is essential to maintain the microbial stability of 
sodium alginate topical gels [35] . 
Tragacanth Tragacanth is a polysaccharide polymer obtained from some Astragalus 
species. It is composed of two polysaccharides: bassorin (water insoluble) and 
tragacanthin (water soluble). It is available as odorless powder white to yellowish 
in color and possesses mucilaginous taste. Tragacanth swells about 10 times its 
weight in water and forms viscous solutions or gels. Tragacanth is usually added with 
vigorous stirring to avoid lump formation. Wetting agents such as glycerin, propylene 
glycol, and 95% ethanol are used in initial stages to improve wetting and dispersion 
of tragacanth in water. The viscosity of tragacanth dispersions is infl uenced by 
the processing temperature and formulation pH. High temperature usually increases 
the viscosity of gels. Tragacanth dispersions show higher viscosity at pH 8 and starts 
decreasing at higher pH. These gels usually contain preservatives such as benzoic 
acid or a combination of methyl and propyl parabens for effective preservation from 
microbial attack. The viscosity of tragacanth dispersions reduces in the presence of 
strong mineral and organic acids and sodium chloride [6] . 
4.2.3.6 Preparation and Packaging 
Gels are relatively easier to prepare compare to ointments and creams. In addition 
to the gelling agent, medicated gels contain drug, antimicrobial preservatives, stabilizers, 
dispersing agents, and permeation enhancers. Some of the factors discussed 
below are essential to obtain a uniform gel preparation. 
Order of Mixing The order of mixing of these ingredients with the gelling agent 
is based on their infl uence on the gelling process. If they are likely to infl uence the 
rate and extent of swelling of the gelling agent, they are mixed after the formation 
of gel. In the absence of such interference, the drug and other additives are mixed 
prior to the swelling process. In this case, the effects of mixing temperature, swelling 
duration, and other processing conditions on the physicochemical stability of the 
drug and additives are also considered. Ideally the drug and other additives are 
dissolved in the swelling solvent, and the swelling agent is added to this solution 
and allowed to swell. 
Gelling Medium Purifi ed water is the most widely used dispersion medium in the 
preparation of gels. Under certain circumstances, gels may also contain cosolvents 
or dispersing agents. A mixture of ethanol and toluene improves the dispersion of 
ethylcellulose, dichloromethane and methanol increase the viscosity of hydroxypropyl 
cellulose dispersions, alcohol improves the rheological stability of polyethylene 
oxide gels, and inclusion of glycerin, propylene glycol, sucrose, and alcohol improves 
the dispersion of sodium alginate dispersions. Borax is included in polyvinyl alcohol 
gels and magnesium oxide, zinc oxide, and glycerin are included in bentonite gel as 
GELS 301

302 SEMISOLID DOSAGES: OINTMENTS, CREAMS, AND GELS 
dispersing agents. Care should be taken to avoid the evaporation or degradation of 
these cosolvents and dispersing agents during the preparation of gels. 
Processing Conditions and Duration of Swelling The processing temperature, pH 
of the dispersion, and duration of swelling are critical parameters in the preparation 
of gels. These conditions vary with each gelling agent. For instance, hot water is 
preferred for gelatin and polyvinyl alcohol, and cold water is preferred for methylcellulose 
dispersions. Carbomers, guar gum, hydroxypropyl cellulose, poloxamer, 
and tragacanth form gels at weakly acidic or near - neutral pH conditions (pH 5 – 8). 
Gelling agents such as carboxymethyl cellulose sodium, hydroxypropylmethyl cellulose, 
and sodium alginate form gels over a wide pH range (4 – 10). Hydroxyethyl 
cellulose forms gel at alkaline pH condition. A swelling duration of about 24 – 48 h 
generally helps in obtaining homogeneous gels. Natural gums need about 24 h and 
cellulose polymers require about 48 h for complete hydration. 
Removal of Entrapped Air Entrapment of air bubbles in the gel matrix is a 
common issue, especially when the swelling process involves a mixing procedure or 
the drug and other additives are added after the swelling process. Positioning the 
propeller at the bottom of the mixing container minimizes this issue to a larger 
extent. Further removal of air bubbles can be achieved by long - term standing, low - 
temperature storage, sonication, or inclusion of silicon antifoaming agents. In large - 
scale production, vacuum vessel deaerators are used to remove the entrapped air. 
Packaging Being viscous and non - Newtonian systems, gels need high attention 
during packing into containers. Usually they are packed into squeeze tubes or jars 
made of plastic materials. Aluminum containers are also used when the product pH 
is slightly acidic. Pump dispensers and prefi lled syringes are sometimes used for 
packing gels. As most of the gels contain an aqueous phase, preservation in airtight 
containers helps in protecting them from microbial attack. Usually they are preserved 
at room temperature and protected from direct sunlight and moisture. 
In large - scale production, different mills, separators, mixers, deaerators, shifters, 
and packaging machines are used. Most of this equipment is similar to those discussed 
under ointments and creams. Figure 3 shows a “ one - bowl ” vacuum processing 
machine manufactured by FrymaKoruma - Rheinfelden (Romaco) for the 
preparation gels. Batch sizes ranging from 15 to 160 L are processed using this 
machine. It uses an extremely effi cient high - shear rotor/stator system for homogenizing 
and a counterrotating mixing system for macromixing. The raw materials are 
drawn into the multichamber system of the homogenizer by vacuum and then mixed 
and pumped into the homogenizing zone. The product which enters the vessel is 
mixed, sheared, and recirculated. All the entrapped air is removed during the recirculation. 
The machine also has an insulated jacket for controlling the processing 
temperature. 
4.2.3.7 Evaluation 
Various pharmacopeial and nonpharmacopeial tests are carried out to evaluate the 
physicochemical, microbial, in vitro, and in vivo characteristics of gels. These tests 
are meant for assessing the quality of gel formulations and minimizing the batch -

to - batch variations. Some of the tests recommended by the USP for gels include 
minimum fi ll, pH, viscosity, microbial screening, and assay. In some cases sterility 
and alcohol content are also specifi ed. The USP also recommends storage for each 
compendial gel formulation. Table 9 shows the quality control tests and storage 
requirements that are specifi ed for some pharmacopeial gels. The procedures for 
minimum fi ll, microbial screening, sterility, assay, in vitro drug release, and in vivo 
bioequivalence are similar to those of ointments and creams. The procedures for 
additional tests such as homogeneity, surface morphology, pH, alcohol content, 
rheological properties, bioadhesion, stability, and ex vivo penetration are described 
below. 
Homogeneity and Surface Morphology The homogeneity of gel formulations is 
usually assessed by visual inspection and the surface morphology by using scanning 
electron microscopy. Generally, the swollen gel is allowed to freeze in liquid nitrogen 
and then lyophilized by freeze drying. It is assumed that the morphologies of 
the swollen samples are retained during this process. The lyophilized hydrogel is 
gold sputter coated and viewed under an electron microscope. 
pH Many gelling agents show pH - dependent gelling behavior. They show highest 
viscosity at their gel point. Determination of pH is therefore important to maintain 
consistent quality. As conventional pH measurements are diffi cult and often give 
erratic results, special pH electrodes are used for viscous gels. Flat - surface digital 
FIGURE 3 Vacuum processing machine used for preparation of gels. (Courtesy of 
FrymaKoruma - Rheinfelden, Switzerland.) 
GELS 303

304 SEMISOLID DOSAGES: OINTMENTS, CREAMS, AND GELS 
TABLE 9 USP Specifi cations for Some Offi cial Gels 
Drug Quality Control Tests Packaging and Storage Requirements 
Aminobenzoic 
acid 
Minimum fi ll, pH (4.0 – 6.0), 
alcohol content, and assay 
Tight, light - resistant containers 
Benzocaine Microbial limits, minimum fi ll, 
and assay 
Well - closed containers 
Benzoyl peroxide pH (2.8 – 6.6), and assay Tight containers 
Betamethasone 
benzoate 
Microbial limits, minimum fi ll, 
and assay 
Tight containers; store at 25 ° C, 
excursions permitted between 15 
and 30 ° C; protect from freezing 
Clindamycin 
phosphate 
Minimum fi ll, pH (4.5 and 
6.5), and assay 
Tight containers 
Desoximetasone Minimum fi ll, alcohol content, 
and assay 
Collapsible tubes at controlled room 
temperature 
Dexamethasone Minimum fi ll and assay Collapsible tubes; keep tightly closed; 
avoid exposure to temperatures 
exceeding 30 ° C 
Dyclonine 
hydrochloride 
pH (2.0 and 4.0), and assay Collapsible, opaque plastic tubes or in 
tight, light - resistant glass containers 
Erythromycin Minimum fi ll and assay Tight containers 
Fluocinonide Minimum fi ll and assay Collapsible tubes or tight containers 
Hydrocortisone Minimum fi ll and assay Tight containers 
Lidocaine 
hydrochloride 
Sterility, minimum fi ll, pH 
(7.0 – 7.4), and assay 
Tight containers 
Metronidazole Minimum fi ll, pH (4.0 and 
6.5), and assay 
Laminated collapsible tubes at 
controlled room temperature 
Naftifi ne 
hydrochloride 
Microbial limits, minimum fi ll, 
pH (5.5 – 7.5), content of 
alcohol, and assay 
Tight containers 
Salicylic acid Alcohol content and assay Collapsible tubes or tight containers, 
preferably at controlled room 
temperature 
Sodium sulfi de pH (11.5 – 13.5) and assay Tight containers at controlled room 
temperature or in a cool place 
Stannous fl uoride Viscosity, pH (2.8 – 4.0), 
stannous ion content, total 
tin content, and assay 
Well - closed containers 
Tolnaftate Minimum fi ll and assay Tight containers 
pH electrodes from Crison, combination electrodes that contain a built - in temperature 
probe, a bridge electrolyte chamber and movable sleeve junction from 
Mettler, and combination pH puncture electrodes with spear - shaped tip from 
Mettler are some commercially available pH measurement systems for semisolid 
formulations. 
Alcohol Content Alcohol levels in some gel preparations are determined by gas 
chromatographic (GC) methods. Desoxymetasone gel USP and naftifi ne hydrochloride 
gel USP contain 18 – 24% and 40 – 45% (w/w) of ethyl alcohol, respectively. In a 
desoxymetasone gel, the sample is dissolved in methanol and injected into a gas 

chramatograph for quantitative analysis. Isopropyl alcohol is used as an internal 
standard. In naftifi ne hydrochloride gel, n - propyl alcohol is used as an internal 
standard and water is used as the diluting solvent [15] . 
Rheological Studies Viscosity measurement is often the quickest, most accurate, 
reliable method to charactreize gels. It gives an idea about the ease with which gels 
can be processed, handled, or used. Some of the commonly used tests for characterizing 
rheology of gels are yield stress, critical strain, and creep. Yield stress refers to 
the stress that must be exceeded to induce fl ow. This helps in characterizing the fl ow 
nature of non - Newtonian systems. Critical strain or gel strength refers to the minimum 
energy needed to disrupt the gel structure. When samples are subjected to increasing 
stress, viscosity is maintained as long as the gel structure is maintained. When the 
gel ’ s intermolecular forces are overcome by the oscillation stress, the sample breaks 
down and the viscosity falls. The higher the critical strain, the better the physical 
integrity of gel systems. Creep or recovery helps in assessing the strength of bonds 
in a gel structure. This is assessed by determining relaxation times, zero - shear viscosity, 
and viscoelastic properties. 
Based on the nature of the test material, different techniques are employed to 
measure the rheological parametrs of gels. Very sophisticated automatic equipment 
is commercially available for measurements. Cup - and - bob viscometers and cone - 
and - plate viscometers are widely used for viscous liquids and gels. They measure 
the frictional force that is created when gels start fl owing. These viscometers are 
usually calibrated with certifi ed viscosity standards before each measurement. 
General - purpose silicone fl uids which are less sensitive to temperature change are 
used as standards. The viscosity of gels is affected by the experimental temperature 
and shear rate and the gels exhibit liquid - or solidlike properties. Hence the viscosity 
of these non - Newtonian systems are recorded at several shear rates under controlled 
temperatures. The USP specifi es the operating conditions for each gel formulation. 
Commercially available viscometers include Brookfi eld rotational 
viscometers, Haake rheometers, Schott viscoeasy rotational viscometers, Malvern 
viscometers, and Ferranti - Shirley cone - and - plate viscometers. 
Bioadhesion This test is performed to assess the force of adhesion of a gel with 
biological membranes. The bioadhesive property is preferred for ophthalmic, nasal 
buccal, and gastroretentive gel formulations. Drugs applied as solutions, viscous 
solutions, and suspensions drain out from these biological locations within a short 
time and only a limited fraction of drug elicits the pharmacological activity. Products 
with higher bioadhesion thus help in increasing the contact time between drugs and 
absorbing surface and improve their availability. The bioadhesive properties of 
gels are measured using various custom - designed equipment. All the equipment, 
however, measures the force required to detach the gel from a biological surface 
under controlled experimental conditions (e.g., temperature, wetting level, contact 
time, contact force, surface area of tissue). A typical bioadhesion measurement 
system consists of a moving platform and a static platform. A tissue from a particular 
biological region is fi xed onto these platforms and a known quantity of the test 
product is uniformly applied to the tissue surface of the lower static platform. The 
upper moving platform is allowed to contact with the product surface with a known 
contact force. After allowing for a short contact time, the moving platform is 
GELS 305

306 SEMISOLID DOSAGES: OINTMENTS, CREAMS, AND GELS 
separated from the product with a constant rate. The force required to detach the 
mucosal surface from the product is recorded. The analog signals generated by 
precision load cells are then converted to digital signals through data acquisition 
systems and processed using specifi c software programs. 
Stability Studies Being dispersed systems containing water in their matrix, gels 
are prone to physical, chemical, and microbial stability issues. Syneresis is a commonly 
observed physical stability problem with gels. It involves squeezing out dispersion 
medium due to elastic contraction of polymeric gelling agents. This results 
in shrinkage of gels. Syneresis can be determined by heating the gels to a higher 
temperature followed by rapid cooling using an ice water bath at room temperature. 
The sample is preserved at 4 ° C for about a week, and water loss from the gel matrix 
is measured. Water loss is measured by weighing the mass of the gel matrix after 
centrifugation. Absence of syneresis indicates higher physical stability of gels. The 
chemical stability of drugs in the gel matrix is determined using stability - indicating 
analytical methods. Studies are conducted at accelerated temperature, moisture, and 
light conditions to determine the possible degradation of drug in the gel. 
Ex Vivo Penetration Ex vivo studies are carried out to examine the permeation 
of drug from gels through the skin or any other biological membrane. As with in 
vitro release studies, ex vivo penetration is conducted using vertical diffusion cells 
or modifi ed cells with fl ow - through design. In this case, the receiver side is fi lled 
with phosphate buffer solution of pH 7.4 to simulate the biological pH of human 
blood. Skin samples from different animal sources such as rats, rabbits, pigs, and 
human cadavers are used for screening dermatological products. The stratum 
corneum layer of the skin is separated from the dermis before mounting onto the 
diffusion cells. The epidermis is separated by immersing the skin sample in normal 
saline or purifi ed water which is maintained at 60 ° C for 2 min followed by immersion 
into cold water for 30 s. Careful peeling helps in the separation of the epidermis 
layer from the dermis. This layer is mounted between the donor and receiver sides 
and studies are conducted after application of test gel over the surface of the stratum 
corneum in the donor side. Samples are withdrawn at different time intervals and 
analyzed for drug permeation by suitable analytical techniques. 
4.2.3.8 Typical Pharmacopeial and Commercial Examples 
Gels are becoming popular dosage forms for delivering various categories of drugs 
for treating dermatological, oral, ophthalmic, vaginal, and other conditions. Many 
dermatological gels are used for treating mild to moderate acne, eczema, dermatitis, 
allergies, rash, and psoriasis and for removal of common warts. Oral gels are available 
for relieving painful mouth sores, treating tooth decay, preventing tooth plaque, 
and relieving infl ammation of the gums, and vaginal gels are available for treating 
certain type of vaginal infections (e.g., bacterial vaginosis). Some special types of 
gels are available for preventing or controlling pain during certain medical procedures, 
numbing and treating urinary tract infl ammation (urethritis), and numbing 
mucous membranes. Table 10 shows some of the commercially available compendial 
gels. 

TABLE 10 Examples of Compendial/Commercial Gels 
Drug a Category Indication 
Commercial 
Products 
Strength 
(%) 
Benzocaine Local 
anesthetic 
In mouth to relieve pain 
or irritation caused by 
many conditions 
Oratect Gel, 
Num Zit 
Gel 
7.5, 10 
Benzoyl 
peroxide 
Keratolytic Mild to moderate acne Persa - Gel, 5 
Benzagel 
10 
5.0, 10 
Betamethasone Anti - 
infl ammatory 
Eczema, dermatitis, 
allergies, and rash 
Diprolene 0.05 
Clindamycin 
phosphate 
(vag) 
Antibiotic Certain types of vaginal 
infection (e.g., bacterial 
vaginosis) 
Cleocin T 1.0 
Desoximetasone Anti - 
infl ammatory 
Eczema, dermatitis, 
allergies, and rash. 
Topicort 0.05 
Dyclonine 
hydrochloride 
Antipruritic 
and local 
anesthetic 
Relieve painful mouth 
sores 
Dyclone 0.5, 1.0 
Erythromycin Antibiotic Acne and skin infection 
due to bacteria 
Erygel 2.0 
Fluocinonide Anti - 
infl ammatory 
Psoriasis, eczema, 
dermatitis, allergies, 
and rash 
Lidex 0.05 
Lidocaine 
hydrochloride 
Local 
anesthetic 
Prevent and control pain 
during certain medical 
procedures, numb and 
treat urinary tract 
infl ammation 
(urethritis), and numb 
mucous membranes 
Xylocaine, 
Anestacon 
2.0 
Metronidazole 
(vag) 
Antifungal Certain types of bacterial 
infections in the vagina 
Metrogel 0.75, 1.0 
Naftifi ne 
hydrochloride 
Antifungal Fungal infections of skin 
such as jock itch, 
athlete ’ s feet, or 
ringworm 
Naftin 1.0 
Salicylic acid Keratolytic Removal of common 
warts 
Sal - Plant 
Gel 
17.0 
Stannous 
fl uoride 
Fluoride Treat tooth decay, prevent 
tooth plaque and 
infl ammation of gums 
Flo - Gel, 
Gel - Kam 
0.4 
Tolnaftate Antifungal Skin infections such as 
athlete ’ s foot, jock itch, 
and ringworm 
Tolnaftate 1.0 
a vag: vaginal gel. 
GELS 307

308 SEMISOLID DOSAGES: OINTMENTS, CREAMS, AND GELS 
TABLE 11 Description on SUPAC Guidelines for Nonsterile Semisolid Dosage Forms 
Type of Change Level Description 
Change in 
components 
and 
composition 
1 Partial deletion or deletion of color, fragrance, or fl avor; up to 
5% excipient change in approved amount; change in supplier 
for structure forming or technical - grade excipient 
2 Excipient changes from 5 to 10%; change in supplier for 
structure forming excipient, which is not covered under level 
1; change in technical grade of structure forming excipient; 
change in particle size of drug if drug is in suspension 
3 Qualitative and quantitative changes in excipient not covered 
under levels 1 and 2; any change in crystallinity of drug if 
drug is in suspension 
4.2.4 REGULATORY REQUIREMENTS FOR SEMISOLIDS 
Regulatory agencies such as the Center for Drug Evaluation and Research (CDER) 
have at the Food and Drug Administration (FDA) have set forth guidelines for 
various pharmaceutical activities to ensure the identity, strength, quality, safety, and 
effi cacy of semisolid drug products. A manufacturer of semisolid formulations needs 
to fulfi ll these requirements at the time of fi ling for investigational new drug (IND), 
abbreviated new drug application (ANDA), or abbreviated antibiotic drug application 
(AADA). Standard chemistry, manufacturing, and control (CMC) tests are 
necessitated for all dermatological drug products. Additional information on polymorphic 
form, particle size distribution, and other characteristics is needed for 
submitting an NDA. When an ANDA for a semisolid product is fi led, the manufacturer 
should meet the standards of compendial requirements if available and match 
the important in vitro and in vivo characteristics of the reference listed drug (RLD). 
If such information is not available, appropriate in vitro release methods are submitted 
to ensure batch - to - batch consistency. Even at later stages, if changes are 
made for an approved semisolid product with respect to its components, composition, 
equipment, process, batch size, and manufacturing site, the formulator should 
submit necessary details to the regulatory agency. 
A typical guideline that defi nes the types and levels of scale - up and postapproval 
changes (SUPAC) is outlined in Table 11 . Based on the type and level of change, 
the manufacturer needs to submit application and compendial product release 
requirements, executed batch records, accelerated and long - term stability data, identifi 
cation and assay for new preservative, preservative effectiveness test at lowest 
specifi ed level, validation methods to support absence of interference of preservative 
with other tests, in vitro release test, and in vivo bioequivalence data to the 
FDA. When changes are made with respect to the quality and quantity of excipients 
or crystallinity of drug, especially if the drug is in suspension, in vivo bioequivalence 
studies are recommended. As routine pharmacokinetic studies do not produce 
measurable quantities of drug in blood, plasma, urine, and other extracutaneous 
biological fl uids, dermatopharmacokinetic (DPK) studies and pharmacodynamic or 
comparative clinical trials are recommended to establish bioequivalence of topical 
products. Table 12 shows specifi c requirements for various SUPAC levels. If bioavailability 
or bioequivalence data of a highest strength product are already available, 

TABLE 12 SUPAC Requirements for Nonsterile Semisolid Dosage Forms 
Parameter 
Change 
Level 
Requirements a 
A B C D E F G H I J K 
Change in 
components 
and 
composition 
1 • • 
2 • • • • 
3 • • • • • 
Change in 
preservative 
components 
and 
composition 
1 • • 
2 • • 
3 • • • • • 
Change in 
manufacturing 
equipment 
1 • • 
2 • • • • 
Change in 
manufacturing 
process 
1 • 
2 • • • • 
Change in batch 
size 
1 • • • 
2 • • • 
REGULATORY REQUIREMENTS FOR SEMISOLIDS 309 
Type of Change Level Description 
Change in 
preservative 
components 
and 
composition 
1 Less than 10% quantitative change in preservative 
2 10 – 20% quantitative change in preservative 
3 Deletion or more than 20% quantitative change in 
preservative; inclusion of a different preservative 
Change in 
manufacturing 
equipment 
1 Change to automated or mechanical equipment for transfer of 
ingredients; use of alternative equipment of same design and 
operating principles 
2 Use of alternative equipment of different design and operating 
principles; change in type of mixing equipment 
Change in 
manufacturing 
process 
1 Changes in process within approved application ranges; 
addition of formulation additives 
2 Changes in process outside approved application ranges; 
process of combining phases 
Change in batch 
size 
1 Batch size changes upto 10 times of pivotal clinical trial or 
biobatch 
2 Batch size changes above 10 times of pivotal clinical trial or 
biobatch 
Change in 
manufacturing 
site 
1 Changes within existing facility 
2 Changes within same campus or facilities in adjacent city 
blocks 
3 Change to different campus; change to a contract manufacturer 
TABLE 11 Continued

310 SEMISOLID DOSAGES: OINTMENTS, CREAMS, AND GELS 
in vitro release data are used to evaluate the in vivo bioequivalence of a lower 
strength product [16] . 
REFERENCES 
1. Ansel , H. C. , Allen , L. V. , and Popovich , N. G. ( 1999 ), Pharmaceutical Dosage Forms and 
Drug Delivery Systems , 7th ed. , Lippincott Williams and Wilkins , Philadelphia , pp. 
245 – 250 . 
2. Breit , J. , and Bandmann , H.J. ( 1973 ), Dermatitis from lanolin , Br. J. Dermatol. , 88 , 
414 – 416 . 
3. Smolinske , S. C. ( 1992 ), Handbook of Food, Drug, and Cosmetic Excipients , CRC Press , 
Boca Raton, FL , pp. 225 – 229 . 
4. Barker , G. ( 1977 ), New trends in formulating with mineral oil and petrolatum . Cosmet. 
Toilet. 92 ( 1 ), 43 – 46 . 
5. Davis , S. S. ( 1969 ), Viscoelastic properties of pharmaceutical semisolids I: Ointment bases . 
J. Pharm. Sci. , 58 , 412 – 418 . 
6. Rowe , R. C. , Sheskey , P. J. , and Weller , P. J. ( 2003 ), Handbook of Pharmaceutical Excipients 
, 4 ed. , Pharmaceutical Press and American Pharmaceutical Association , IL , pp. 16 – 18 , 
89 – 92 , 260 – 263 , 287 – 288 , 417 – 418 , 491 – 492 , 508 – 513 , 524 – 525 , 618 – 619 , 654 – 656 , 
679 – 684 . 
7. Hadia , I. A. , Ugrine , H. E. , Farouk , A. M. , and Shayoub , M. ( 1989 ), Formulation of polyethylene 
glycol ointment bases suitable for tropical and subtropical climates I . Acta 
Pharm. Hung. , 59 , 137 – 142 . 
Parameter 
Change 
Level 
Requirements a 
A B C D E F G H I J K 
Change in 
manufacturing 
site 
1 • 
2 • • • • 
3 • • • • • 
a Only those highlighted with black circles: 
A: Application/compendial product release requirement. 
B: Executed batch records. 
C: long - term stability data for fi rst production batch. 
D: 3 - month accelerated stability data for 1 batch and long - term data for fi rst production batch. 
E: 3 - month accelerated stability data for 1 batch and long - term data for fi rst 3 production batches if signifi 
cant information is available or 3 - month accelerated stability data for 3 batches and long - term data 
for fi rst 3 production batches if signifi cant information is not available. 
F: 3 - month accelerated stability data for 1 batch and long - term data for fi rst production batch if signifi - 
cant information is available or 3 - month accelerated stability data for 3 batches and long - term data for 
fi rst 3 production batches if signifi cant information is not available. 
G: In vitro release test. 
H: In vivo bioequivalence. 
I: Preservative effectiveness test at lowest specifi ed preservative level. 
J: Identifi cation and assay for new preservative; validation methods to support absence of interference 
with other tests. 
K: Location of new site. 
TABLE 12 Continued

8. Fisher , A. A. ( 1978 ), Immediate and delayed allergic contact reactions to polyethylene 
glycol , Contact Dermatitis , 4 , 135 – 138 . 
9. Mores , L. R. ( 1980 ), Application of stearates in cosmetic creams and lotions , Cosmet. 
Toilet. 95 ( 3 ), 79 – 84 . 
10. Mapstone , G. E. ( 1974 ), Crystalization of cetyl alcohol from cosmetic emulsions , Cosmet. 
Perfum. , 89 ( 11 ), 31 – 33 . 
11. Eccleston , G. M. ( 1984 ), Properties of fatty alcohol mixed emulsifi ers and emulsifying 
waxes , in Florence , A. T. , Ed., Materials Used in Pharmaceutical Formulations: Critical 
Reports on Applied Chemistry , Vol. 6, Blackwell Scientifi c , Oxford, UK , pp. 124 – 156 . 
12. Kline , C. H. (1964), Thixcin, R-Thixotrope , Drug Cosmet. Ind. , 95 ( 6 ), 895 – 897 . 
13. Cronin , E. ( 1967 ), Contact dermatitis from cosmetics , J. Soc. Cosmet. Chem. , 18 , 
681 – 691 . 
14. Forcinio , H. ( 1998 ), Tubes: The ideal packaging for semisolid products , Pharm. Tech. , 22 , 
32 – 36 . 
15. The United States Pharmacopeia/The National Formulary , 28th/23rd ed., U.S. Pharmacopeial 
Convention, Rockville, HD, 2005 , pp. 2246 – 2255, 2378, 2434 – 2435, 2441, 2510 – 2512. 
16. Guidance for industry, Nonsterile semisolid dosage forms: Scale - up and postapproval 
changes , U.S. Food and Drug Administration, May 1997 , pp. 1 – 37. 
17. Shah, V. P. , Glynn, G. L. , and Yacobi, A. (1998), Bioequivalence of topical dosage forms — 
methods of evaluation of bioequivalence , Pharm. Res. , 15 , 167 – 171 . 
18. Allen , L. V. ( 2002 ), The Art, Science, and Technology of Pharmaceutical Compounding , 
American Pharmaceutical Association , Washington, DC , pp. 301 – 312 . 
19. Jeong , B. , Bae , Y. H. , and Kim , S. W. ( 1999 ), Thermoreversible gelation of PEG - PLGA - 
PEG triblock copolymers aqueous solutions , Macromolecules , 32 , 7064 – 7069 . 
20. Miyazaki , S. , Suisha , F. , Kawasaki , N. , Shirakawa , M. , Yamatoya , K. , and Attwood , D. 
( 1998 ), Thermally reversible xyloglucan gels as vehicles for rectal drug delivery , J. Controlled 
Release , 56 , 75 – 83 . 
21. Watanabe , K. , Yakou , S. , Takayama , K. , Isowa , K. , and Nagai , T. ( 1996 ), Rectal absorption 
and mucosal irritation of rectal gels containing buprenorphine hydrochloride prepared 
with water - soluble dietary fi bers, xanthan gum and locust bean gum , J. Controlled Release , 
38 , 29 – 37 . 
22. Cohen , S. , Lobel , E. , Trevgoda , A. , and Peled , T. ( 1997 ), A novel in situ — forming ophthalmic 
drug delivery system from alginates undergoing gelation in the eye , J. Controlled 
Release , 44 , 201 – 208 . 
23. Carlfors , J. , Edsman , K. , Petersson , R. , and Jornving , K. ( 1998 ), Rheological evaluation of 
Gelrite in situ gels for ophthalmic use , Eur. J. Pharm. Sci. , 6 , 113 – 119 . 
24. Altagracia , M. , Ford , I. , Garzon , M. L. , and Kravzov , J. ( 1987 ), A comparative mineralogical 
and physico - chemical study of some crude Mexican and pharmaceutical grade montmorillonites 
, Drug Dev. Ind. Pharm. , 13 , 2249 – 2262 . 
25. Doelker , E. ( 1993 ), Cellulose derivatives , Adv. Polym. Sci. , 107 , 199 – 265 . 
26. Lev , R. , Long , R. , and Mallonga , L. ( 1997 ), Evaluation of carrageenan as a base for topical 
gels , Pharm. Res. , 14 ( 11 ), 42 . 
27. Daniels , R. , Kerstiens , B. , Tishinger - Wagner , H. , and Rupprecht , H. ( 1986 ), The stability 
of drug absorbates on silica , Drug Dev. Ind. Pharm. , 12 , 2127 – 2156 . 
28. Ruiz - Martinez , A. , Zouaki , Y. , and Gallard - Lara , V. ( 2001 ), In vitro evaluation of benzylsalicylate 
polymer interaction in topical formulation , Pharm. Ind. , 63 , 985 – 988 . 
29. Ling , W. C. ( 1978 ), Thermal degradation of gelatin as applied to processing of gel mass , 
J. Pharm. Sci. , 67 , 218 – 223 . 
REFERENCES 311

312 SEMISOLID DOSAGES: OINTMENTS, CREAMS, AND GELS 
30. Goldstein , A. M. , Alter , E. N. , and Seaman , J. K. ( 1973 ), Guar gum , in Whistler , R. L. , Ed., 
Industrial Gums , 2nd ed. , Academic , New York , pp. 303 – 321 . 
31. Gauger , L. J. ( 1984 ), Hydroxyethylcellulose gel as a dinaprostone vehicle , Am. J. Hosp. 
Pharm. , 41 , 1761 – 1762 . 
32. Farley , C. A. , and Lund , W. ( 1976 ), Suspending agents for extemporaneous dispensing: 
Evaluation of alternatives to tragacanth , Pharm. J. , 216 , 562 – 566 . 
33. Cabana , A. , Ait - Kadi , A. , and Juhasz , J. ( 1997 ), Study of the gelation process of polyethylene 
oxide copolymer (poloxamer 407) aqueous solutions , J. Colloid Interface Sci. , 190 , 
307 – 312 . 
34. Burdick , K. H. , Haleblian , J. K. , Poulsen , B. J. , and Cobner , S. E. , ( 1973 ), Corticosteroid 
ointments: Comparison by two human bioassays , Curr. Ther. Res. , 15 , 233 – 242 . 
35. Pavics , L. ( 1970 ), Comparison of rheological properties of mucilages , Acta Pharm. Hung. , 
40 , 52 – 59 . 

313 
4.3 
LIQUID DOSAGE FORMS 
Maria V. Rubio - Bonilla 1 , Roberto Londono 1 , and Arcesio Rubio 2 
1 Washington State University, Pullman, Washington 
2 Caracas, Venezuela 
Contents 
4.3.1 Introduction 
4.3.2 Generalities 
4.3.2.1 Dosage Form 
4.3.2.2 Liquid Dosage Form 
4.3.2.3 Dispersed Systems 
4.3.2.4 Solutions 
4.3.2.5 Manufacturing of Nonparenteral Liquid Dosage Forms 
4.3.2.6 Optimizing Drug Development Strategies 
4.3.2.7 Unit Operation or Batch 
4.3.2.8 Batch Management 
4.3.2.9 Steps of Liquids Manufacturing Process 
4.3.2.10 Protocols 
4.3.3 Approaches 
4.3.4 Critical Aspects of Liquids Manufacturing Process 
4.3.4.1 Physical Plant 
4.3.4.2 Equipment 
4.3.4.3 Particle Size of Raw Materials 
4.3.4.4 Compounding: Effects of Heat and Process Time 
4.3.4.5 Uniformity of Oral Suspensions 
4.3.4.6 Uniformity of Emulsions 
4.3.4.7 Microbiological Quality 
4.3.4.8 Filling and Packing 
4.3.4.9 Stability 
4.3.4.10 Process Validation 
4.3.5 Liquid Dosage Forms 
References 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

314 LIQUID DOSAGE FORMS 
4.3.1 INTRODUCTION 
Liquid dosage forms are designed to provide the maximum therapeutic response in 
a target population with diffi culty swallowing tablets and capsules and/or to produce 
rapid therapeutic effects. The major ingredient in most liquid dosage forms is water. 
While it is the safest and most palatable solvent option, water quality is signifi cant 
for the stability of pharmaceutical dosage forms. Furthermore, the Food and 
Drug Administration (FDA) “ Guide to Inspections of Dosage Form Drug Manufacturer 
’ s — CGMPRs ” considers microbial contamination due to inappropriate 
design and control of purifi ed water systems as the most common problem of liquid 
dosage forms. Solutions and dispersions studied in this chapter are chemically, 
microbiologically, and/or physically unstable systems that require a high level of 
organizational management of manufacturing processes in order to maintain a state 
of apparent stability, at least until the expiration date [1] . 
The pharmaceutical industry manufactures dosage forms in large - scale formulations. 
The decision to scale up is based on the economics of the production process 
related to costs of materials, personnel, equipment, and control [2] . To reduce costs 
of wastes and to obtain high - quality and effi cacious drug products, the strategic plan 
to be applied during the process has to be developed carefully. In fact, the variables 
that affect product quality are identifi ed and understood in the process instead of 
tested into the fi nal product [3] . 
Commercial liquid dosage forms reach large - scale production after being preformulated 
at the laboratory level followed by formulation at the small scale and then 
at the pilot plant scale. Due to the complexity of the manufacturing process, scale - up 
from pilot to commercial production is not a simple extrapolation. The approaches 
to the four levels of production are different. Most of the formulation ingredients 
are analyzed, studied, and selected at the laboratory scale. While small - scale production 
is more focused on the liquid preparation procedure with higher amounts of 
ingredients, the main issues at the pilot plant scale are the design of infrastructure 
and reduction of costs. Commercial production introduces problems that are not a 
major issue on a small scale, for instance, materials handling and storage, pulverizing, 
mixing, dissipation of the generated heat during production, time control, personnel 
administration, and bottle - fi lling capabilities. Furthermore, purifi ed water is essential 
for the manufacturing of these products as well as on - site packing capabilities 
[2] . 
The organization and advance of the pharmaceutical industry should be focused 
on three main points of project development based on quality by design (QbD): 
product objective (design of experiment, DoE), production resources (process analytical 
technology, PAT), and product acceptability (quality system) [3] . Manufacturers 
of liquid dosage forms must ensure safety, effi cacy, stability, elegance, and 
acceptability of the fi nal drug product while achieving development and clinical 
milestones [1] . Achieving the desirable clinical attributes of the product effi cacy 
means ensuring potency stability by confi rming the functionability of the manufacturing 
process and quality system. The stability and safety of the product are 
goals to be reached through chemical and microbiological stability by establishing 
and updating manufacturing and quality control protocols of product development. 
Time control and aesthetic considerations refl ect the physical stability through 
product elegance and acceptability. Flavoring, sweetening, coloring, and texturing 

are both challenges and opportunities. They are challenges because “ no single 
correct method exists to solve signifi cant problems of elegance ” ; they are opportunities 
because “ they enable a pharmacist to prepare a product more easily 
accepted by the patient ” [4] . Although the most important characteristic of a 
dosage form is effi cacy, there are other characteristics that remain important subjects 
for the manufacturing of liquid dosage forms such as safety as well as chemical, 
physical, and microbiological stabilities. From a pharmaceutical point of view, 
stability problems are the main causes of safety complaints. Despite its signifi cance, 
some companies decide to outsource stability services [5] . To solve or minimize 
stability problems in drug products, it is necessary to analyze and enhance the 
development of critical control points in each operation of the full manufacturing 
process as well as expected variances and tolerance limits. 
Except some aqueous acids, water in aqueous solutions is an excellent media for 
microbiological growth, such as molds, yeast, and bacteria. Typical microorganisms 
affecting drug microbiological stability are Pseudomonas, Escherichia coli Salmonella 
, and Staphylococcus [1] . Defi cient methods or an insuffi cient preservative 
system may be the principal causes of microbiological contamination in the pharmaceutical 
industry of liquid manufacturing [6] . 
Chemical instability reactions appear with or without microbiological contribution 
through reactions such as hydrolysis, oxidation, isomerization, and epimerization. 
Interactions between ingredients and ingredients with container closure 
materials are established as the principal causes of these reactions [1] , for instance, 
the hydrolysis of cefotaxime sodium, the oxidation of vitamin C, the isomerization 
of epinephrine, and the epimerization of tetracycline [7] . 
In most cases, physical instabilities are consequences of previous chemical instabilities. 
Physical instabilities can arise principally from changes in uniformity of 
suspensions or emulsions, diffi culties related to dissolution of ingredients, and 
volume changes [6] . For instance, some cases where physical stability has been 
affected are cloudiness, fl occulence, fi lm formation, separation of phases, precipitation, 
crystal formation, droplets of fog forming on the inside of container, and swelling 
of the container [8] . 
Although commercial oral solution and emulsion dosage forms rarely present 
bioequivalence issues, some bioequivalence problems have been reported for oral 
suspensions such as phenytoin [9] . The possibility of microbiological contamination 
and physicochemical instabilities during the manufacturing process also needs 
to be carefully considered. To approach the stability problems of liquid dosage 
forms, in this chapter, the main critical aspects during the manufacturing process 
are based on FDA inspection. From physical plant systems to batching management 
and packing, the potential sources of microbiological, chemical, and physical instabilities 
will be analyzed using defi nitions, case - by - case explanations, and practical 
examples. 
Final product stability, which determines the therapeutic activity and uniformity 
among other characteristics of the fi nal product, refl ects the dynamic of the production 
process. Conceptualization of stability issues is important to determine the 
changes to enhance the design space as well as protocols of manufacturing and 
quality control [3] . An information technology (IT) solution like Enterprice Resource 
Planning (ERP) may support the pharmaceutical industry ’ s current challenges of 
organization [10] . 
INTRODUCTION 315

316 LIQUID DOSAGE FORMS 
4.3.2 GENERALITIES 
4.3.2.1 Dosage Form 
According to the FDA: “ A dosage form is the physical form in which a drug is produced 
and dispensed. In determining dosage form, FDA examines such factors as 
(1) Elegance: physical appearance of the drug product, (2) Stability: physical form 
of the drug product prior to dispensing to the patient, (3) Acceptability: the way the 
product is administered, (4) Effi cacy: frequency of dosing, and (5) Safety: how pharmacists 
and other health professionals might recognize and handle the product ” 
[11] . The term dosage form is different from “ dose, ” which is defi ned as a specifi c 
amount of a therapeutic agent that can be taken at one time or at intervals. 
4.3.2.2 Liquid Dosage Form 
The physical form of a drug product that is pourable displays Newtonian or pseudoplastic 
fl ow behavior and conforms to its container at room temperature. In contrast, 
a semisolid is not pourable and does not fl ow at low shear stress or conform 
to its container at room temperature [12] . According to its physical characteristics, 
liquid dosage forms may be dispersed systems or solutions. 
4.3.2.3 Dispersed Systems 
Dispersed systems are dosage forms composed of two or more phases, where one 
phase is distributed in another [2] . If a dispersed system is formed by liquid phases, 
then it is known as an “ emulsion. ” In contrast, the dispersed system is named a 
“ suspension ” when the liquid dosage form is accomplished by the distribution of a 
solid phase suspended in a liquid matrix. The solid phase of a suspension is usually 
the drug substance, which is insoluble or very poorly soluble in the matrix [12] . 
4.3.2.4 Solutions 
A solution refers two or more substances mixed homogeneously [2] . Although solubility 
refers to the concentration of a solute in a saturated solution at a specifi c 
temperature, in pharmacy, solution liquid dosage forms are unsatured to avoid 
crystallization of the drug by seeding of particles or changes of pH or temperature 
[13] . The precipitation of drug crystals is one of the most important physical instabilities 
of solutions that may affect its performance [14] . Water is the most used 
solvent in solutions manufacturing; however, there are also some commercial nonaqueous 
solutions in the pharmaceutical market [1] . 
4.3.2.5 Manufacturing of Nonparenteral Liquid Dosage Forms 
The manufacturing of liquid dosage forms with market - oriented planning includes 
the following stages with respect to special good manufacturing practice (GMP) 
requirements: planning of material requirements, liquid preparation, fi lling and 
packing, sales of drug products, vendor handling, and customer service [15] . From 
the viewpoint of product stability, each stage of the process includes critical batches 

that are more decisive than others. Also, each decisive batch contains one or several 
unit operations that are more critical than others. The FDA inspection focuses on 
those critical unit operations to ensure the safety and stability of the liquid dosage 
forms [6] . 
4.3.2.6 Optimizing Drug Development Strategies 
According to Sokoll [16] : The phases of drug development include discovery, preclinical 
development, clinical development, fi ling for licensure, approval/licensure and post - 
approval. Discovery typically includes basic research, drug identifi cation and early - 
stage process and analytical method development. . . . Emerging companies that review 
their pipeline objectively and strike a balance between properly resourcing and developing 
their lead candidates in the clinic while nurturing their next generation of drug 
candidates will have the best chance for success and sustainability. 
4.3.2.7 Unit Operation or Batch 
A “ batch ” job or operation is defi ned as a unit of work. Raw materials, semifi nished 
drug products (bulk), and fi nished drug products are handled in batches. Each different 
type of material used during the process, such as product packing, should be 
managed by batches. This applies also to process aids and operation facilities [15] . 
4.3.2.8 Batch Management 
The batch management of production simplifi es the process and makes it easier to 
control the status of transformation between raw and fi nal products [2] . Some of 
the data used to follow the material performance around and out of the product 
manufacturing process are batch - where - used - list, initial status, batch determinations, 
master data, and expiration date check [15] . 
The functionality of the overall process to manufacture liquid dosage forms 
depends on the successful linkage of one unit operation to another. To use mathematical 
formulations to scale up the manufacturing process, it is necessary to divide 
the process into stages, batches, and unit operations. Each single unit operation is 
scalable, but the composite manufacturing process is not. Production problems 
result from attempts to follow a process scale - up instead of a unit operation scale - 
up. By using mathematical formulations, it is possible to understand the level of 
similarity between two scale sizes. In addition, nonlinear similarities between two 
scale sizes might require the use of conversion factors to achieve an extrapolation 
point for the scale [2] . 
4.3.2.9 Steps of Liquids Manufacturing Process 
Establishing short - term goals makes it easier to measure effi ciency as well as evaluate 
the diffi culties [2] . Based on these concepts, the problems of manufacturing 
liquid dosage forms can be approached as problems in one or more batches of the 
following process steps [6, 15] : 
Planning of Material Requirements Research and development of protocols and 
selection of materials; acquisition and analysis of raw materials; physical plant 
GENERALITIES 317

318 LIQUID DOSAGE FORMS 
design, building, and installation; equipment selection and acquisition; personnel 
selection and initial training; and monitoring information system. 
Liquid Preparation Research and development of protocols concerning liquid 
compounding; scale - up of the bulk product compounding; physical plant control 
and maintenance; equipment maintenance and renovation; continuous 
training of personnel and personnel compensation plan; and supervision of 
system reports. 
Filling and Packing Research and development of protocols concerning fi lling 
and packing; scale - up of the fi nished drug product fi lling and packing; physical 
plant control and maintenance; equipment maintenance and renovation; continuous 
training of personnel and personnel compensation plan; and supervision 
of system reports. 
Sales of Drug Products Research and development of protocols concerning 
product storage; distribution process; continuous training of personnel and 
personnel compensation plan; and supervision of system reports. 
Vendor Handling Research and development protocols concerning precautions 
to maintain product stability; control of vendor stock; and sales system 
reports. 
Customer Service Research and development of protocols concerning home 
storage and handling to maintain product stability; relations with health insurance 
companies and health care professionals; educational materials for patient 
counseling; and customer service system reports. 
4.3.2.10 Protocols 
Protocols are patterns developed by repeating procedures and fi xing the identifi ed 
problems each time that the procedure is followed. Therefore, protocols are dynamic 
entities that originally can be developed at a laboratory level but must be adjusted 
in every new step of the scal - up process. When the manufacturing process moves 
up in scale, the number of people affected by the protocol increases geometrically. 
Initially, the information can be obtained from library references, personal tests, 
interpersonal training, and previous laboratory protocols. However, when the production 
is scaled up, the information required to fi ne tune the process comes from 
monitoring the process itself [2] . 
4.3.3 APPROACHES 
Quality by Design is a systemic approach that applies the scientifi c method to the 
process. QbD theory contains components of management, statistics, psychology, 
and sociology. The FDA ’ s new century has identifi ed the QbD approach as its “ key 
component ” based on process quality control before industry end results [3, 17] . 
The cooperation between industry members and regulators is increased when the 
industry explains clearly what it is doing and the agency can understand the formulation 
and production process. In these cases, regulatory relief appears when industry 
explores its issues and receives active guidance and programs from the FDA. 
The agency takes the role of facilitator, or even partner of the industry, in order to 
improve the strength of the process and formulation [3, 17] . 

To apply QbD as a systemic approach, the company starts by understanding, 
step by step, the space design, the design of the dosage form, the manufacturing 
process, and the critical process parameters to be controlled in order to reach the 
new building block which is the expectation of variances within those critical 
process parameters that can be accepted. This approach allows the establishment 
of priorities and fl exible boundaries in the process [3] . Infl exible specifi cations 
allow uncontrolled small variances that can follow the butterfl y effect of the theory 
of chaos by producing unpredictable large variations in the long - term behavior of 
the product shelf - life [18, 19] . In contrast, fl exibility, with knowledge of potential 
variances, reduces changes in the approved spaces and manufacturing protocols 
[3, 17] . 
According to the FDA [6] , critical parameters during the manufacturing process 
of nonparenteral liquid dosage forms may appear in the design of physical plant 
systems, equipment, protocols of usage and maintenance, raw materials, compounding, 
microbiological quality control, uniformity of suspensions and emulsions, and 
fi lling and packing [6] . 
Process isolation and installation of an appropriate air fi ltration system in the 
physical plant may reduce product exposition to chemical and microbiological 
contaminations. In addition, the use of a suitable dust removal system as well as 
a heating, ventilation, and air conditioning system (HVACS) may help to repress 
product chemical instabilities [6] . 
The equipment of sanitary design, including transfer lines, as well as appropriate 
cleaning and sanitization protocols may reduce chemical and microbiological contaminations 
in the fi nal product. Chemical instabilities may be reduced by weighting 
the right amount of liquids instead of using a volumetric measurement, avoiding the 
common use of connections between processes, and using appropriate batching 
equipment [6] . 
Particle sizes of raw materials are critical to control dissolution in solutions as 
well as uniformity in suspensions and emulsions. Temperature control during compounding 
is important since heat helps to support mixing and/or fi lling operations, 
but, in contrast, high - energy mixers may produce adverse levels of heat that affect 
product stability. Too much heat may cause chemical and physical instabilities such 
as change of particle size or crystallization of drugs in suspensions, dissolution and 
potency loss of drugs in suspensions, oxidation of components, and activation of 
microbiological growth after degradation of compounds as well as precipitation of 
dissolved compounds in solution [20] . In addition, uniformity of suspensions depends 
on viscosity and segregation factors while solubility, particle size, and crystalline 
form determine uniformity of emulsions. Application of pharmaceutical GMP for 
product processes and storage assures microbiological quality. A defi cient deionizer 
water - monitoring program and product preservative system facilitate microbial contamination. 
Filling uniformity is indispensable for potency uniformity of unit - dose 
products and depends on the mixing operation. Calibration of provided measuring 
devices and the use of clean containers will allow administering the right amount 
of the expected components in the liquid dosage form [6] . 
Principal product specifi cations are microbial limits and testing methods, particle 
size, viscosity, pH, and dissolution of components. Process validation requires control 
of critical parameters observed during compounding and scale - up. Product stability 
examination is based on chemical degradation of the active components and interac- 
APPROACHES 319

320 LIQUID DOSAGE FORMS 
tions with closure systems, physical consequences of moisture loss, and microbial 
contamination control [6] . 
4.3.4 CRITICAL ASPECTS OF LIQUIDS MANUFACTURING PROCESS 
4.3.4.1 Physical Plant 
Heating, Ventilation, and Air Conditioning System The manufacturer has to 
warrant adequate heating, ventilation, and air conditioning in places where labile 
drugs are processed [6] . 
The effect of long processing times at suboptimal temperatures should be considered 
at the production scale in terms of the consequences on the physical or chemical 
stability of individual ingredients and product. A pilot plant or production scale 
differs from laboratory scale in that their volume - to - surface - area ratio is relatively 
large. Thus, for prolonged suboptimal temperatures, jacketed vessels or immersion 
heaters or cooling units with rapid circulation times are absolutely necessary [2] . 
For heat - labile drugs, uncontrolled temperature increments can activate auto - 
oxidation chains when the drug product ingredients react with oxygen and generate 
free radicals but without drastic external interference. Vitamins, essential oils, and 
almost all fats and oils can be oxidized. A good example of a heat - labile drug solution 
is clindamycin, which has to be stored at room temperature and away from 
excess heat and moisture [19] . Auto - oxidation chains are fi nished when free radicals 
react with each other or with antioxidant molecules (quenching). The tocopherols, 
some esters of gallic acid, as well as BHA and BHT (butylated hydroxyanisole and 
butylated hydroxytoluene) are common antioxidants used in the pharmaceutical 
industry [1] . 
Isolation of Processes To minimize cross - contamination and microbiological contamination, 
the manufacturer may develop special procedures for the isolation of 
processes. The level of facilities isolation depends on the types of products to be 
manufactured. For instance, steroids and sulfas require more isolation than over - 
the - counter (OTC) oral products [6] . To minimize exposure of personnel to drug 
aerosols and loss of product, a sealed pressure vessel must be used to compound 
aerosol suspensions and emulsions [21] . An example of cross - contamination with 
steroids was the controversial case of a topical drug manufactured for the treatment 
of skin diseases. High - performance liquid chromatography/ultraviolet and mass 
spectrometry (HPLC/UV, HPLC/MS) techniques were used by the FDA for the 
detection of clobetasol propionate, a class 1 superpotent steroid, as an undeclared 
steroid in zinc pyrithione formulations. The product was forbidden and a warning 
was widely published [22] . 
Dust Removal System The effi ciency of the dust removal system depends on the 
amount and characteristics of dust generated during the addition of drug substance 
and powdered excipients to manufacturing vessels [6] . Pharmaceutical industries 
usually generate some type of dust or fume during processing. Important factors for 
selecting dust collectors are maintenance, surrogate test, economics, and containment. 
In addition, reentrainment of the fi ne particles, vertical or horizontal position, 
effi ciency, pressure resistant, service life time, as well as sealing capacity to work 

CRITICAL ASPECTS OF LIQUIDS MANUFACTURING PROCESS 321 
through the bag are signifi cant factors concerning fi lter selection of dust removal 
systems. Some examples of dust collection applications in the manufacture of liquid 
dosage forms are handling and pulverization of raw materials, spray dryers, and 
general room ventilation [23] . 
Air Filtration System The effi ciency of the air fi ltration system has to be demonstrated 
by surface or air - sampling data where the air is recirculated [6] . To monitor 
the levels of contamination in the air, there are commercial automatic samplers for 
microbiological contamination or gas presence. Air trace environmental samplers 
for pharmaceutical industries are based on the slit - to - agar impaction technique for 
the presence of viable microorganisms. Automatic samplers for compressed gas 
analyze the presence of a specifi ed gas in 1 m 3 by absorbing air at a fi xed fl ow rate 
for a sampling period of 1 h or a different adjusted time. These solutions to the 
sampling needs of the pharmaceutical industry are robust, require low maintenance, 
and are easy to use. This allows for validation of sampling data at the moment of 
application fi lling to support the process control. Sampling time and selection of 
microbiological growth media or analysis technique are important components to 
consider when developing a sampling plan [24] . 
4.3.4.2 Equipment 
Sanitary Design Pumps, valves, fl owmeters, and other equipment should be easily 
sanitized. Some examples of identifi ed sources of contamination are ball valves, 
packing in pumps, and pockets in fl owmeters [6] . 
The sanitary design and performance of equipment make it accessible for inspection, 
cleaning, and maintenance. It has to be cleanable at a microbiological level and 
its performance during normal operations should contribute to sanitary conditions. 
The materials used in the design have to assure hygienic compatibility with other 
equipment, the product, the environment, other systems such as electrical, hydraulics, 
steam, air, and water, as well as the method and products used for cleaning and 
sanitation. The equipment should be self - draining to assure product or liquid collection. 
Small niches, for example, pits, cracks, corrosion, recesses, open seams, gaps, 
lap seams, protruding ledges, inside threads, bolt rivets, and dead ends, as well as 
inaccessible cavities of equipment such as entrap and curlers must be eliminated 
whenever possible; otherwise they have to be permanently sealed. Enclosures, for 
example, push buttons, valve handles, switches, and touch screens, should be prepared 
for a hygienic design of maintenance. Standards have been developed by the 
American Meat Institute [25] . 
Standard Operating Procedures for Cleaning Production Equipments Current 
GMPs are defi ned as the basic principles, procedures, and resources required to guarantee 
an environment appropriate for manufacturing products of adequate quality 
[26] . To minimize cross - contamination and microbiological contamination, it is GMP 
for a manufacturer to create and pursue written standard operating procedures 
(SOPs) to clean and sanitize production equipment in a way that avoids contamination 
of in - progress and upcoming batches. When the drug is known as a potent generator 
of allergic reactions, such as steroids, antibiotics, or sulfas, cross - contamination 
becomes an issue of safety [20] . In addition, validation and data analysis procedures, 

322 LIQUID DOSAGE FORMS 
FIGURE 1 Mixing and fi lling lines for pharmaceutical dosage forms. Positive indoor pressure 
of 5 psi over outdoor pressure assures constant airfl ow from inside to outside in order 
to reduce entrance of contaminating agents. 
UV Rays 
Collector Tank 
Continuous or Batch 
GENERAL DIAGRAM “A” 
INDUSTRIAL MANUFACTURING PLANT 
FOR PHARMACEUTICAL LIQUID DOSAGE FORMS Decontamination 
Camera 
Restricted access area 
Pressure = Atmospheric pressure + 5 PSIG 
Bottling 
Equipment 
Packing 
Filters 
Compressor 
Homogenizator 
Main 
Pump 
Filters 
Mixer 
Dosing 
Pumps 
Primary Components Tanks 
Restriction 
Gate 
Compressor 
Purified 
Water 
Air 
recycle 
including drawings of the manufacturing and fi lling lines [6] , are especially important 
for clean - in - place (CIP) systems, as indicated in Figures 1 and 2 . 
Many companies have problems with standardizing operating procedures for 
cleaning steps and materials used [6] . Appropriate SOPs are necessary to determine 
the scope of the problem in investigations about possible cross - contaminations or 
mix - ups. The best approach to validate a SOP is to test it, use it as a training tool, 
and observe the results obtained by different persons. This includes the worst - case 
situation in order to enhance the step - by - step writing methodology as well as standardizing 
the materials used. A typical SOP contains a header to present the SOP 
title, date of issue, date of last review, total number of pages, responsible person, 
and approval signature. Typically, a SOP includes position of responsible person, 
SOP purpose and scope, defi nitions, equipment and materials, safety concerns, step - 
by - step procedure, explanation of critical steps, tables to keep data, copies of forms 
to fi ll, and references [26] . The forms to keep the records must show the date, time, 
product, and lot number of each batch processed. However, the most important 
points of the SOP are equipment identity, cleaning method(s) with documentation 
of critical cleaning steps, materials approved for cleaning that have to be easily 
removable, names and position of persons responsible for cleaning and inspection, 
inspection methods, and maintenance and cleaning history of the equipment [20] . 

CRITICAL ASPECTS OF LIQUIDS MANUFACTURING PROCESS 323 
Cleaning and Sanitizing Transfer Lines Pipes should be hard, easily cleaned, and 
sanitized. To avoid moisture collection and microbiological contamination, hoses 
should be stored in a way that allows them to drain rather than be looped. For 
example, transfer lines are an important source of contamination when fl exible 
hoses are handled by operators, lying on the fl oor, and after they are placed in 
transfer or batching tanks [6] . 
Heat is considered one of the most effi cient physical treatments for sanitizing 
pharmaceutical equipment and could be used for sanitizing hoses that have already 
been cleaned. The recirculation of hot water at a temperature of 95 o C for at least 
100 min allows bacteria elimination [14] . 
Due to the important amount of insoluble residues left on piping and transfer 
lines after emulsion manufacturing, such as topical creams and ointments, equipment 
cleaning becomes diffi cult to address. To avoid cross - contamination, some 
manufacturers have decided to dedicate lines and hoses to specifi c products. 
FIGURE 2 Mixing and fi lling lines for pharmaceutical dosage forms. Using this hydropneumatic 
system, instead of the mechanical system in Figure 1 , the liquid moves by the 
pressure generated in a compressed air tank. 
Hydropneumatic 
vessel 
UV Rays 
Restricted access area 
Pressure = Atmospheric pressure + 5 PSIG 
Compressor 
Collector Tank 
Continuous or Batch 
GENERAL DIAGRAM “B” 
INDUSTRIAL MANUFACTURING PLANT 
FOR PHARMACEUTICAL LIQUID DOSAGE FORMS 
OF LOW VISCOSITY 
Restriction 
Gate
Air 
Recycle 
Pressure 
pump 
Decontamination 
Camera 
Bottling 
Equipment 
Homogenizator Dosing 
Pumps 
Primary Components Tanks Purified 
Water 
Homogenizator
Mixing 
Control 
Booster 
Pump 
Mixer 
Packing 
Filters 
Compressor 
Restriction 
Gate 
Filters

324 LIQUID DOSAGE FORMS 
However, these decisions have to appear in the written production protocols and 
SOPs [20] . 
Sampling Cleaned Surfaces for Presence of Residues The cleaning method is 
validated by sampling the cleaned surfaces of the equipment for the existence of 
residues. The equipment characteristics and residue solubility are factors to support 
the selection of the sampling method to be used [6] . There are two acceptable 
general types of sampling methods: direct surface sampling by swabbing of surfaces 
and rinse sampling with a routine production in - process control. Although surface 
residues will not be identical on each part of the surface, statistically the most 
advantageous is direct surface sampling because it allows evaluation of the hardest 
areas to clean as well as insoluble or “ dried - out ” residues by physical removal. The 
type of sampling material and solvent used for extraction from the sampling material 
should be validated in order to determine their impact on the test data. The 
second method, rinse sampling, is used for larger surfaces or inaccessible systems. 
Contaminants that are physically occluded and insoluble residues are disadvantages 
of the rinse sampling method. To validate this cleaning process, direct measurement 
of the contaminant in the rinse water has to be tested instead of a simple 
test for water quality. Routine production in - process control is used as indirect 
testing for large equipment that has to be cleaned by the rinse sampling method. 
The uncleaned equipment has to give an unacceptable result for the indirect 
test [27] . 
Establishing Appropriate Limits on Levels of Postequipment Cleaning 
Residues Very low levels of residue are possible to be determined since technological 
advances offer more sensitive analytical methods. The manufacturer should 
know the toxicological information of the materials used and potential amounts of 
residues after exposure to the equipment surface. Accordingly, the manufacturer 
has to establish proper limits of residues after equipment cleaning and scientifi cally 
justify these limits. The established limits must be clinically and pharmaceutically 
safe, realistic, viable, and verifi able [20] . The sensitivity of the analytical method will 
determine the logic of the established limits since absence of residues could indicate 
a low sensitivity of the analytical method or a poor sampling procedure. Sometimes 
thin - layer chromatography (TLC) screening must be used in addition to chemical 
analyses. Some practical levels established by manufacturers include 10 ppm of 
chemicals, 1/1000 of the biological activity levels met on a normal therapeutic dose, 
and no visible residues of particles determined organoleptically [27] . 
Connections Connectors and manifolds should not be for common use. For 
example, sharing connectors in a water supply, premix, or raw material supply tanks 
may be a source of cross - contamination [6] . 
Time between Completion of Manufacturing and Initiation of Cleaning The time 
that may elapse from completion of a manufacturing operation to initiation of 
equipment cleaning should also be stated where excessive delay may affect the 
adequacy of the established cleaning procedure. For example, residual product may 
dry and become more diffi cult to clean [20] . 
SOPs are an example of defi ciency in many manufacturers regarding time limitations 
between batch cleaning and sanitization [6] . Lack of communication between 

CRITICAL ASPECTS OF LIQUIDS MANUFACTURING PROCESS 325 
departments responsible for the production at different levels is the main cause of 
time control problems. Typically each department, from human resources to fi nances, 
manufacturing, and warehouse, has its own computer system optimized for the 
particular ways that the department does its work. Therefore, time control becomes 
a primordial issue when labile materials are transferred from one department to 
another [28] . 
To facilitate communication between different departments, some useful softwares 
have been developed. For example, ERP is an integrated approach which may 
have positive payback if the manufacturer installs it correctly. An ERP is a type 
of software that can improve communication between planning and resources. The 
software attempts to integrate all departments and functions in a company onto a 
single computer system that can serve each particular need, such as fi nance, human 
resources, manufacturing management, process manufacturing management, inventory 
management, purchasing management, quality management, and sales management. 
Each department has its own software, except now the software is linked 
together, so that, for example, someone in manufacturing can look into the maintenance 
software to see if specifi c batch cleaning and sanitization have been scheduled 
or realized and someone in fi nance can review the warehouse software to see if a 
specifi c order has been shipped. The information is online and not in someone ’ s 
heads or on papers that can be misplaced. People in different departments may see 
the same information, update it if they are allowed to do, and make right decisions 
faster. However, the software is less important than the changes companies make 
in the ways they work. Reorganization and training are the keys of ERP ’ s success 
to fi x integration problems. There are three different ways to install an ERP: big 
bang, franchising strategy, and slam dunk. Big bang is the most ambitious way 
whereby companies install a single ERP across the entire company. By the franchising 
strategy, departments do not share many common processes across, whereas 
slam dunk is focused on just a few key processes [28] . 
Weight in Formulations Flow properties of liquids rarely vary due to their constant 
density at a constant temperature. Oral solutions and suspensions are formulated 
on a weight basis (gravimetry) in order to be able to measure the fi nal volume 
by weight before fi lling and packing. Volumetric measurements of liquid amounts 
to be used for manufacturing liquid dosage forms have shown greater variability 
than weighted liquids. For instance, the inaccurate measurement of the fi nal volume 
by using dip sticks or a line on a tank may cause further analytical errors and 
potency changes [6] . 
The importance of selecting gravimetry instead of volumetry to measure liquid 
amounts in the pharmaceutical industry of liquid dosage forms is well illustrated by 
the volume contraction of water – ethanol and volume expansion of ethyl acetate – 
carbon disulfi de liquid mixtures as well as a CS2 – ethyl acetate system. The National 
Formulary (NF) diluted alcohol is a typical example of the volume nonadditivity of 
liquid mixtures [29] . This solution is prepared by mixing equal volumes of alcohol 
[U.S. Pharmacopeia (USP)] USP and purifi ed water (USP). The fi nal volume of this 
solution is about 3% less than the sum of the individual volumes because of the 
contraction due to the mixing phenomenon [1] . In addition, molecular interactions 
of surfactants in mixed monolayers at the air – aqueous solution interface and in 
mixed micelles in aqueous media also cause some contraction of volume upon 
mixing [30] . 

326 LIQUID DOSAGE FORMS 
Location of Bottom Discharge Valve in Batching Tank The bottom discharge 
valve should be located exactly at the bottom of the tank. In some cases valves have 
been found to be several inches to a foot above the bottom of the tank [6] . 
For a tank suspected of having substantial deposits at the bottom, a fi ber - optic 
camera can be inserted in the tank to provide a view and positive confi rmation of 
the tank bottom condition. These camera and light vision systems are sanitary equipment 
able to provide a computational real - time visual inspection of the inside tank 
under process conditions or pressure vessel. In addition, they are used to control 
several parameters during the manufacturing process, such as product level and 
thickness, solids level, uniformity of suspensions, foam, and interface and/or cake 
detection [31] . 
Batching Equipment to Mix Solution Ingredients of solutions have to be completely 
dissolved. For instance, it has been observed that some low - solubility drugs 
or preservatives can be kept in the “ dead leg ” below the tank, and the initial samples 
have reduced potency [6] . When there is inadequate solubility of the drug in the 
chosen vehicle, the dose is unable to contain the correct amount of drug in a manageable 
size unit, that is, one teaspoonful or one tablespoonful. Thus, ingredients 
as well as handling and storage conditions should be chosen to manage the problem 
[14] . 
In solutions, the most important physical factors that infl uence the solubility of 
ingredients are type of fl uid, mixing equipment, and mixing operations. Generalized 
Newtonian fl uids are ideal fl uids for which the ratio of the shear rate to the shear 
stress is constant at a particular time. Unfortunately, in practice, usually liquid 
dosage forms and their ingredients are non - Newtonian fl uids in which the ratio of 
the shear rate to the shear stress varies. As a result, non - Newtonian fl uids may not 
have a well - defi ned viscosity [32] . 
When all the ingredients are miscible liquids, the combination and distribution 
of these components to obtain a homogeneous mixture are called blending. Whenever 
possible, ingredients should be added together and the impeller mixer often is 
located near the bottom of the vessel [21] . Mixing of high - viscosity materials requires 
higher velocity gradients in the mixing zone than regular blending operations. In 
fact, the fundamental laws of physics regarding the performance of Newtonian fl uids 
in the production process may be studied using computational tools. For example, 
VisiMix is a software that is routinely used to calculate shear rates [2] . 
Finally, if it is determined that there is a bigger problem of insolubility 
coming from the formulation, then addition of cosolvents, surfactants, as well as 
the preparation of the ionized form of an acid or base, drug derivatization, and 
solid - state manipulation are approaches to manipulating the solubility of the drug 
[14] . 
Batching Equipment to Mix Suspension In the case of suspensions, the fl ow necessary 
to overcome settling in a satisfactory suspension depends on the mixing 
equipment and is predicted by Stokes ’ s law. Thus, to use the Stokes ’ s law, suspensions 
are considered as Newtonian fl uids if the percentage of solids is below 50%. 
Mixing equipment uses a mechanical device that moves through the liquid at a given 
velocity. Dispersing and emulsifying equipment is categorized as “ high - shear ” mixing 
equipment. The maximum shear rate with such equipment occurs very close to the 

CRITICAL ASPECTS OF LIQUIDS MANUFACTURING PROCESS 327 
mixing impeller. Therefore, the diameter of the impeller and the impeller speed 
directly infl uence the power applied by the mixer to the liquid [21] . 
Batching Equipment to Mix Emulsion The most common problems of mixing 
emulsions are removing “ dead spots ” of the mixture and scrapping internal walls of 
the mixer. Dead spots are quantities of ingredients that are not mixed and become 
immobile. Where dead spots are present, that quantity of the formula has to be 
recirculated or removed and not used. If the inside walls of the mixer keep residual 
material, operators should use hard spatulas to scrape the walls; otherwise the 
residual material will become part of the next batch. In both cases, the result may 
be nonuniformity. Stainless steel mixers have to include blades made of hard plastic, 
such as Tefl on, to facilitate the scrapping of the mixer walls without damaging the 
mixer. Scrapper blades should be fl exible enough to remove internal material but 
not too rigid to avoid damaging the mixer [20] . The mixing will be successful if the 
macroscale mixing offers suffi cient fl ow of components in all areas in the mixing 
tank and the microscopic examination shows a correct particle size distribution 
[33] . 
4.3.4.3 Particle Size of Raw Materials 
Raw materials in Solution The types of raw materials used to be part of solutions 
are presented in Table 1 . They have different purposes and can be cosolvents, electrolytes, 
buffers, antioxidants, preservatives, coloring, fl avoring and sweetener agents, 
among others. 
Particle Size of Raw Materials in Solution Particle size is affected by the breaking 
process of the particle, crystal form, and/or salt form of the drug. The particle 
size can affect the rate of dissolution of raw materials in the manufacturing process. 
Raw materials of a fi ner particle size may dissolve faster because they have a larger 
surface area in contact with the solvent than those of a larger particle size when the 
product is compounded [6] . Mixing faster causes the particle to break down and 
dissolve more quickly. In addition, hydrated particles are less soluble than their 
anhydrous partners [37] . 
TABLE 1 Solutions: pharmaceutical excipients 
Purpose Agent 
Protecting the active product 
ingredients 
- Buffers 
- Antioxidants 
- Preservatives 
Maintaining the appearance - Colorings 
- Stabilizers 
- Cosolvents 
- Antimicrobial preservatives 
- Electrolytes 
Taste/Small Masking - Sweeteners 
- Flavorings 
Source : From ref. 4, 34, 35, 36 

328 LIQUID DOSAGE FORMS 
Solid drugs may occur as pure crystalline substances of defi nite identifi able shape 
or as amorphous particles without defi nite structure. In addition, when a drug particle 
is broken up, the total surface area is increased as well as its rate of dissolution. 
The amorphous form of a chemical is usually more soluble than the crystalline form 
while the crystalline form usually is more stable than the amorphous form [37] . 
Processing conditions used for providers to obtain raw materials can dramatically 
impact their quality and stability; for instance, the presence of different polymorphs 
may depend on the thermal history of freezing, concentration of solvents, and drying 
conditions [38] . The polymorphism of a crystalline form is the capacity of a chemical 
to form different types of crystals, depending on the conditions of temperature, 
solvents, and time followed for its crystallization. Among different polymorphs, only 
one crystalline form is stable at a given temperature and pressure. Over time, the 
other crystalline forms, called metastable forms, will be transformed into stable 
forms. Transformations longer than the shelf - life of metastable forms into stable 
forms of a drug are very common in fi nal products and compromise its stability and 
effi cacy to different extents depending on quality control [37] . 
While the metastable forms offer higher dissolution rates, many manufacturers 
use a particular amorphous, crystalline, salt, or ester form of a drug with the solubility 
needed to be dissolved in the established conditions, for instance, to prepare a 
chloramphenicol ophthalmic solution [39] . Thus, the selection of amorphous or 
crystalline form of a drug may be of considerable importance to facilitate the formulation, 
handling, and stability [37] . 
However, the dissolution rate of an equal sample of a slowly soluble raw material 
usually will increase with increasing temperature or rate of agitation as well as with 
reduce viscosity, changes of pH or nature of the solvent. In addition, other alternative 
mechanisms to enhance the solubility of insoluble drugs are: 1) hydrophilization: 
the reduction in contact angle or angle between the liquid and solid surface 
[40] , which can be accessed by intensive mixing of the hydrophobic drug with a small 
amount of methylcellulose solution [41] ; 2) the formation of microemulsions: by 
covering small particles with surfactants to obtain micromicelles that are visible only 
in the form of an opalescence; and, 3) the formation of complexing compounds: by 
adding a soluble substance to form soluble reversible complexes. However, the last 
method is used with some restrictions [42] . 
Raw Materials in Suspension The types of raw materials used to be part of suspensions 
are presented in Table 2 . They have different purposes and can be wetting 
agents, salt formation ingredients, buffers, polymers, suspending agents, fl occulating 
agents, electrolytes, antioxidants, poorly soluble Active Product Ingredients, preservatives, 
coloring, fl avoring and sweetener agents, among others. 
Particle Size of Drug in Suspension The physical stability of a suspension can be 
enhanced by controlling the particle size distribution [43] . Uncontrolled changes of 
drug particle size in a suspension affect the dissolution and absorption of the drug 
in the patient. Drug substances of fi ner particle size may be absorbed faster and 
bigger particles may not be absorbed. Aggregation or crystal growth is evaluated 
by particle size measurements using microscopy and a Coulter counter [21] or preferably 
techniques that allow samples to be investigated in the natural state. Allen 
[44] offers an academic and industrial discussion about particle characterization. 

CRITICAL ASPECTS OF LIQUIDS MANUFACTURING PROCESS 329 
Powder properties and behavior, sampling, numerous potential particle size measuring 
devices, available equipment as well as surface and pore size are his principal 
themes. 
Particles are usually very fi ne (1 – 50 . m). For instance, topical suspensions use 
less than 25 . m particle size [6] . The particle size of the drug is the most important 
consideration in the formulation of a suspension, since the sedimentation rate of 
disperse systems is affected by changes in particle size. Finer particles become interconnected 
and produce particle aggregation followed by the formation of nonresuspendable 
sediment, known as caking of the product. The two main causes of 
aggregation and caking are energetic bonding and bonding through shared material. 
A statistical wide distribution of particle sizes gives more compact packing and 
energetic bonding than narrower distributions. It has been observed that heat treatments 
can cause agglomeration of particles, not only due to energetic bonding but 
also by formation of crystal bridges. Also, when the application of shear forces to 
mix and homogenize the suspension uses too high energy inputs, then the probability 
for aggregation increases [43] . 
Examples of oral suspensions in which a specifi c and well - defi ned particle size 
specifi cation for the drug substance is important are phenytoin suspension, carbamazepine 
suspension, trimethoprim and sulfamethoxazole suspension, and hydrocortisone 
suspension [6] . 
There are some useful methods to improve the physical stability of a suspension, 
such as decreasing the salt concentration, addition of additives to regulate the 
osmolarity, as well as changes in excipient concentrations, unit operations in the 
process, origin and synthesis of the drug substance, polymorphic behavior of 
the drug substance crystals, and other particle characteristics. However, methods 
based on changes of the particle properties and the surfactants used are the most 
successful [43] . 
TABLE 2 Suspensions: pharmaceutical excipients 
Purpose Agent 
Facilitating the connection between Active 
Product Ingredient and vehicle 
- Wetting agents particle size ( > 0.1 . m) 
- Salt formation ingredients 
- Sugars 
Protecting the Active Product Ingredients - Buffering – systems 
- Polymers 
- Antioxidants 
- Poorly soluble drugs 
Maintaining the suspension appearance - Colorings 
- Suspending agent 
- Flocculating agent 
- Antimicrobial preservatives 
- Electrolytes 
Masking the unpleasant taste/smell - Sweeteners 
- Flavorings 
- Poorly soluble Active Product Ingredient 
Source : From ref. 4, 34, 35, 36 

330 LIQUID DOSAGE FORMS 
To approach physical stability problems of suspensions, effectiveness and stability 
of surfactants as well as salt concentrations must be checked with accelerated aging. 
In addition, unit operations affecting particle size distribution, surface area, and 
surfactant effectiveness should be approached, taking into account that different 
types of distributions, for instance, volume or number weighted, give a different 
average diameter for an equal sample [43] . 
Raw Materials in Emulsions The types of raw materials used to be part of emulsions 
are presented in table 3 . They have different purposes and can be buffers, 
polymers, emulsifying agents, penetration enhancers, gelling agents, stabilizers, antioxidants, 
preservatives, coloring, fl avoring and sweetener agents, among others. 
Particle Size in Emulsions When a solid drug is suspended in an emulsion, the 
liquid dosage form is known as a coarse dispersion. In addition, a colloidal dispersion 
has solid particles as small as 10 nm – 5 . m and is considered a liquid between 
a true solution and a coarse dispersion [44] . 
4.3.4.4 Compounding: Effects of Heat and Process Time 
Oxygen Oxygen removal for processing materials that require oxygen to degrade 
is possible by methods such as nitrogen purging, storage in sealed tanks, as well 
as special instructions for manufacturing operations [6] . For instance, sealing glass 
ampules containing a liquid dosage form with heat under an inert atmosphere is a 
packing mechanism used to prevent oxidation. Some aspects of oxygen sensitiveness 
that should be taken into account are the necessity of water and headspace deoxygenation 
in ampules before sealing, the avoidance of multidose vials that facilitate 
oxygen contact with the product after opened, and rubber stoppers for vial sealing 
that are permeable to oxygen as well as release additives to catalyze oxidative reac- 
TABLE 3 Emulsions: pharmaceutical excipients 
Purpose Agent 
Particle Size - Solid particles (10 nanometers to 5 micrometers size) 
- Droplet particles (0.1 – 1.0 micrometers size) 
Protecting the Active Product 
Ingredients 
- Buffering - Systems 
- Polymers 
- Antioxidants 
- Distribution pattern (O/W, W/O) 
Maintaining the appearance - Colorings 
- Emulsifying agents 
- Penetration enhancers 
- Gelling agents 
- Stabilizers 
- Antimicrobial preservatives 
Taste/smell Masking - Sweeteners 
- Flavorings 
- Relation oil vs. water 
Source : From ref. 4, 34, 35, 36 

CRITICAL ASPECTS OF LIQUIDS MANUFACTURING PROCESS 331 
tions. Rubber stoppers soften and get sticky over time because all rubber products 
degrade as sulfur bonds induced during vulcanization revert. Connors et al. [45] 
present the oxygen content of water at different temperatures and an interesting 
discussion of calculations for the case of captopril as an oxygen - sensitive drug. 
Dissolution of Drugs in Solutions Although some compounds, such as poloxamers, 
decrease their aqueous solubility with an increase in temperature [46] , usually, 
drugs dissolve more quickly when the temperature increases because particle vibration 
is augmented and the molecules move apart to form a liquid. Chemical instabilities 
by oxidation due to high temperature or prolonged periods of heat exposure 
can occur when trying to increase the dissolution of poorly soluble raw materials. 
To control such instabilities, charts of time and amount of temperature treatments 
to dissolve materials as well as tests of dissolution are required [6] . In addition, 
precipitations and other reactions may occur between salts in solution and can be 
anticipated by using heat - of - mixing data and activation energy calculations for 
decomposition reactions. Connors et al. [45] provide examples of calculations about 
effects of temperature on chemical stability of pharmaceuticals in solution. Regarding 
the instability of the product, the reasons to limit temperature amounts can go 
from controlling fi nal concentration changes to controlling burn - on/fouling when 
too - high temperatures are applied [45] . Usually salts are more soluble in water and 
alcohol than weak acids or bases. The reason salts are not always the best choice to 
increase the solubility of a drug is its permeability. Oral drug absorption depends 
not only on solubility and dissolution but also on permeability through the cellular 
membrane. Drugs have to be able to dissolve not only in the aqueous fl uids of the 
body before reaching the intestinal wall but also in the lipophilic environment of 
the cellular membrane in order to reach the internal part of the cell and interfere 
with its functionability. Therefore, the cosolvent approach is essential if the drug 
presents problems in dissolving in the media. The dielectric constant of a solvent is 
a relative measure of its polarity. Comparing the hydroxyl – carbon ratio of the 
solvent molecule allows establishing the relative polarity of the cosolvent as determined 
by its dielectric constant [47] . Remington describes the formulations of some 
solutions, such as the ferrous sulfate syrup, amantadine hydrochloride syrup, phenobarbital 
elixir, and theophylline elixir [1] . 
Potency of Drugs in Suspension To avoid degradation of the suspended drug substance 
by high temperature or prolonged periods of heat exposure, it is necessary 
to record the time and amount of temperature treatments on charts [6] . The rate 
of dissolution of a suspended drug increases with the increase in temperature. The 
potency stability of a suspended drug depends on the concentration of the dissolved 
drug since drug decomposition occurs only in solution [48] . The goal is to avoid 
the dissolution of suspensions. Changing the pH of the vehicle or replacing the drug 
with a less soluble molecule may result in enhanced potency stability of the suspended 
drug [48] . 
For instance, when the chemical stability of a suspension of ibuprofen powder 
and other ibuprofen – wax microspheres was studied with a modifi ed HPLC procedure 
for three months, the amount of drug released from the microspheres was 
affected by the medium pH, type of suspending agent, and storage temperature 
without observing chemical degradation of the drug [49] . 

332 LIQUID DOSAGE FORMS 
Temperature Uniformity in Emulsions During the preparation of emulsions, heat 
may be increased as part of the manufacturing protocol or mixing operation system. 
Temperature measurements should be monitored and documented continuously 
using a recording thermometer if the temperature control is critical or using a hand - 
held thermometer if it is not a critical factor. Temperature may be critical in the 
manufacturing process depending on the thermosensitivity of the drug product and 
excipients as well as the type of mixer used. To guarantee the temperature uniformity 
during the mixing operation, manufacturers may consider the relation between 
the container size, mixer speed, blade design, viscosity of the contents, and rate of 
heat transfer [20] . 
Fong - Spaven and Hollenbeck [50] studied the apparent viscosity as a function 
of the temperature from 25 to 75 ° C of an oil – water emulsion stabilized with 5% 
triethanolamine stearate (TEAS) using a Brookfi eld digital viscometer. They 
observed that the viscosity decreased when the temperature reached about 48 ° C, 
but surprisingly viscosity increased to a small peak at 54 ° C and then continued 
decreasing after that peak. The viscosity peak was attributed to a transitional gellike 
arrangement molecular structure of TEAS that is destroyed as soon as the temperature 
continues increasing, the TEAS crystalline form reappears, and viscosity again 
decreases [36] . 
Microbiological Control To avoid chemical instabilities that yield microbiological 
and physical instabilities, as a result of high temperature or prolonged periods of 
exposure, it is necessary to record the time and amount of temperature treatments 
on charts [6] . 
Product Uniformity Charts of storage and transfer operation times for the bulk 
product are required to control the risk of segregation. Transfers to the fi lling line 
and during the fi lling operation are the most critical moments to keep the suspension 
uniformity [6] . The implementation of an ERP for time scheduling is the best 
solution for time control and organization of resources. However, it could be diffi - 
cult due to the reluctance of people to change [10] . The constant fl ow of the liquid 
through the piping, the constant mixing of the bulk product in the tank, as well as 
the transfer of small amounts near the end of the fi lling process to a smaller tank 
during the fi lling process may minimize segregation risks [6] . 
Final Volume Excess heating produces variations of the fi nal volume over time 
[6] . Although increasing solute concentration can elevate the boiling point and 
reduce evaporation of water, changes in drug concentration are undesirable because 
they yield different fi nal products. Regarding the instability of the product, the 
reasons to limit temperature amounts can go from controlling fi nal concentration 
changes to controlling burn - on/fouling when too - high temperatures are applied 
[36] . 
A solution is a liquid at room temperature that passes into the gaseous state when 
heated at very high temperature, forming a vapor with determined vapor pressure, 
through a process called vaporization. The kinetic energy is not evenly distributed 
between the molecules of the liquid. When the liquid is in a closed container at a 
constant temperature, the molecules with the highest kinetic energy leave the surface 
of the liquid and become gas molecules. Some of the gas molecules remain as gas 
and others condense and return to the liquid. When, at a determined temperature, 

CRITICAL ASPECTS OF LIQUIDS MANUFACTURING PROCESS 333 
the rate of condensation equals the rate of vaporization, the equilibrium vapor pressure 
is reached. However, vapor pressure increases with increases in liquid temperature, 
resulting in more molecules leaving the liquid surface and becoming gas 
molecules [51] . 
Storage Charts of time and temperature of storage are important to control the 
increased levels of degradedness [6] . Shelf life is defi ned as the amount of time in 
storage that a product can maintain quality and is equivalent to the time taken to 
reach 90% of the composition claim or have 10% degradation. The availability of 
an expiration date is assumed under specifi ed conditions of temperature. Based on 
zero - and fi rst - order reaction calculations, Connors et al. [45] show the estimation 
methods to determine the shelf life of a drug product at temperatures different from 
the one specifi ed under standard conditions. 
4.3.4.5 Uniformity of Oral Suspensions 
Keeping the particles uniformly distributed throughout the dispersion is an important 
aspect of physical stability in suspensions. Based on Stokes ’ s law for dilute 
suspensions where the particles do not interfere with one another, there are different 
factors that control the velocity of particle sedimentation in a suspension, for 
instance, particle diameter, densities of the dispersed phase and the dispersion 
medium, as well as viscosity of the dispersion medium [36] . Remington describes 
the formulation of trisulfapyrimidines oral suspension [1] . In addition, Lieberman 
et al. [42, 48] are also good sources of typical formulations for suspensions. 
Viscosity Depending upon the viscosity, many suspensions require continuous or 
periodic agitation during the fi lling process [6] . 
Segregation in Transfer Lines When the stored bulk of a nonviscous product is 
transferred to fi lling equipment through delivery lines, some level of segregation is 
expected. The manufacturer has to write the procedures and diagrams for line setup 
prior to fi lling the product [6] . Delivery lines of suspensions increase the tendency 
of particles of the same size to assemble together. However, slightly increasing the 
global mixing in the lines can easily reverse the segregation without enhancing the 
global mixing [52] . Shear stress versus rate of shear can be plotted to determine 
the fl ow pattern of a specifi c suspension as pseudoplastic, Newtonian, or dilatant. 
The type of fl ow is determined by the slope of the plot. While shaking increases the 
yield stress and causes particles fl ow, the cessation of shear and rest rebuilds the 
order of the system. A good - quality suspension is known as a thixotrophic system 
and is obtained when the particles at rest avoid or show reduced sedimentation. The 
rheogram of a thixotrope system presents a typical hysteresis or curve representing 
different shear stresses over time [33] . 
Quality Control The GMPs for suspensions include testing samples at different 
checkpoints in the procedure, at the beginning, middle, and end, as well as samples 
from the bulk tank. The uniformity will be successful only if, on microscopic analysis, 
the components are dispersed to the expected particle size distribution established 
by product development. Visual and microscopic examinations should consist of 
looking for verifi cation of foam formation, segregation, and settling, although testing 

334 LIQUID DOSAGE FORMS 
for viscosity is important to determine agitation during the fi lling process. Samples 
used for tests should not be combined again with the lot [6, 33] . 
4.3.4.6 Uniformity of Emulsions 
Remington describes the following three typical formulas of emulsions: type A 
gelatin, mineral oil emulsion (USP), and oral emulsion (O/W) containing an insoluble 
drug [1] . In addition, Lieberman et al. [42, 50] are also good sources of typical 
formulations for emulsions. The components of the emulsion system may present 
physical and chemical instabilities refl ected on the distribution of an active ingredient, 
component migration from one phase to another, polymorphic changes in 
components, and chemical degradation of components [33] . 
Solubility The soluble active ingredient should be added to the liquid phase that 
will be its carrier vehicle. Data of solubility have to be determined as part of the 
process validation. Potency uniformity has to be tested by demonstrating satisfactory 
distribution in the emulsifi ed mix [20] . 
Particle Size Regarding globule diameter in emulsions, the size – frequency distribution 
of particles in an emulsion over time may be the only method for determining 
stability [36] . Drug activity and potency uniformity of insoluble active ingredients 
depend upon control of particle size and distribution in the mix [6] . In addition, 
aggregation of the internal phase droplets, formation of larger droplets, and phase 
separation are categorized as emulsion system instabilities that are refl ected in the 
particle size distribution of the emulsion. The measurement of particle size distribution 
over time allows the characterization of the emulsion stability and determines 
the rheological behavior of the emulsion. Well - accepted approaches to determine 
particle size distribution include microscopy, sedimentation, chromatography, and 
spectroscopy. However, these analyses are problematic in a multiphase emulsion 
[33] . 
Crystalline Form Uncontrolled temperature or shear can induce changes in component 
crystallinity or solubility. For this reason, analytes originally present in each 
phase of the product should be counted as well possible interactions with the container 
or closure and the processing equipment analyzed. Some techniques used to 
obtain information about the emulsion system and its components are microscopic 
examination, macro - and microlaser Raman, and rheological studies [33] . The FDA 
guidance offers the following example: “ in one instance, residual water remaining 
in the manufacturing vessel, used to produce an ophthalmic ointment, resulted in 
partial solubilization and subsequent recrystallization of the drug substance; the 
substance recrystallized in a larger particle size than expected and thereby raised 
questions about the product effi cacy ” [20] . 
4.3.4.7 Microbiological Quality 
Microbial Specifi cations These specifi cations are determined by the manufacturer. 
The USP Chapters 61, 62, and 1111 present the microbial limits to assess the 
signifi cance of microbial contamination in a dosage form [53] . However, the USP 

CRITICAL ASPECTS OF LIQUIDS MANUFACTURING PROCESS 335 
does not determine specifi c methods for water - insoluble topical products. The 
microbial specifi cations are presented as a manufacturer ’ s document that details the 
methods to isolate and identify the organisms as well as the number of organisms 
permitted and action levels to be taken when limits are exceed and the potential 
causes are investigated [6] . The Pharmaceutical Microbiology Newsletter (PMF) 
presents several articles to discuss topics such as microbial identifi cation, methods, 
data analysis, and preservation as well as topics related to USP and FDA regulations 
[54] . To minimize the differences about microbial limits and test methods, the USP 
is trying to harmonize the standards with the European Pharmacopoeia (EP) [55] . 
Microbial Test Methods The selected microbial test methods determine specifi c 
sampling and analytical procedures. When the product has a potential antimicrobial 
effect and/or preservative, the spread technique on microbial test plates must be 
validated. In addition, the personnel performing the analytical techniques have to 
be qualifi ed and adequately trained for this purpose [6] . 
Usually, total aerobic bacteria, molds, and yeasts are counted by using a standard 
plate count in order to test the microbial limits. The microbial limit test may be 
customized by performing a screening for the occurrence of Staphylococcus aureus, 
Pseudomonas aeruginosa, Pseudomonas cepacia, Escherichia coli, and Salmonella 
sp. [56] . 
Investigation of Exceeded Microbiological Limits A high number of organisms 
may indicate defi ciencies in the manufacturing process, such as excessive high 
temperature, component quality, inadequate preservative system, and/or container 
integrity. Information about the health hazards of all organisms isolated from the 
product has different meanings depending on the type of dosage form and group 
of patients to be treated. For instance, in oral liquids, pseudomonads are usually a 
high - risk contamination. Examples presented by the FDA are Nystatin antifungal 
suspension, used as prophylaxis in AIDS patients [57] ; antacids, with which P. aeruginosa 
contamination can promote gastric ulceration [58] ; and the presence of 
Pseudomona putida , which could indicate the presence of other signifi cant contaminants 
such as P. aeruginosa [6] . 
Deionizer Water - Monitoring Program Deionizing systems must be controlled in 
order to produce purifi ed water, required for liquid dosage forms and USP tests and 
assays [1] . The monitoring program has to include the manufacturer ’ s documentation 
about time between recharging and sanitizing, microbial quality and chlorine 
levels of feed water, establishment of water microbial quality specifi cations, conductivity 
monitoring intervals, methods of microbial testing, action levels when microbial 
limits are exceeded, description of sanitization and sterilization procedures for 
deionizer parts, and processing conditions such as temperature, fl ow rates, use 
and sanitization frequency, and regenerant chemicals for ion exchange resin beds 
[6, 59] . 
Effectiveness of Preservative Manufacturing controls and shelf life must ensure 
that the specifi ed preservative level is present and effective as part of the stability 
program [6] . Depending on the type of product, the selection of the preservative 
system is based on different considerations, such as site of use, interactions, 

336 LIQUID DOSAGE FORMS 
spectrum, stability, toxicity, cost, taste, odor, solubility, pH, and comfort. The 
USP and other organizations describe methods to validate the preservative 
system used in the dosage form. Compounds used as preservatives are alcohols, 
acids, esters, and quaternary ammonium compounds, among others. For instance, to 
preserve ophthalmic liquid dosage forms, these products are autoclaved or fi ltrated 
and require an antimicrobial preservative to resist contamination throughout 
their shelf life, such as chlorobutanol, benzalkonium chloride, or phenylmercuric 
nitrate [1] . 
4.3.4.8 Filling and Packing 
Constant Mixing during Filling Process Due to the tendency of suspensions to 
segregate during transport through transfer lines, special attention is required on 
suspension uniformity during the fi lling process. Appropriate constant mixing of the 
bulk to keep homogeneity during the fi lling process and sampling of fi nished products 
and other critical points are indispensable conditions to assure an acceptable 
quality level during the fi lling and packing process [20] . 
Mixing Low Levels of Bulk Near End of Filling Process Constant mixing during 
the fi lling process includes mixing low levels of bulk near the end of the fi lling 
process. Large - size batches of bulk suspension require the transfer of the residual 
material to a smaller tank in order to assure appropriate mixing of components 
before fi lling and packing the containers [20] . 
Potency Uniformity of Unit - Dose Products Products manufactured have to be of 
quality at least as good as the established acceptable quality level (AQL). The quality 
level should be based on the limits specifi ed by the USP. However, when the bulk 
product is not properly mixed during fi lling and packing processes, liquid dosage 
forms, and specially suspensions, are not homogeneous and unit - dose products 
contain very different amounts of the active component and potency. For these 
reasons, fi nished products have to be tested to assure that the fi nal volume and/or 
weight as well as the amount of active ingredient are within the specifi ed limits [6] . 
Calibration of Provided Measuring Devices Measuring devices consist of droppers, 
spoons for liquid dosage forms, and cups labeled with both tsp and mL. Measuring 
devices have to be properly calibrated in order to assure the right amount 
of ingredients per volume to be administered [6] . 
Container Cleanliness of Marketing Product The previous cleanliness of containers 
fi lled with the product will depend on their transportation exposure, composition, 
and storage conditions. Glass containers usually carry at least mold spores of 
different microorganisms, especially if they are transported in cardboard boxes. 
Other containers and closures made with aluminum, Tefl on, metal, or plastic usually 
have smooth surfaces and are free from microbial contamination but may contain 
fi bers or insects [45] . Some manufacturers receive containers individually wrapped 
to reduce contamination risks and others use compressed air to clean them. However, 
the cleanliness of wrapped containers will depend on the provider ’ s guarantee of 
the manufacturing process and compressed - air equipment may release vapors or 
oils that have to be tested and validated [6] . 

CRITICAL ASPECTS OF LIQUIDS MANUFACTURING PROCESS 337 
4.3.4.9 Stability 
The typical stability problems are color change, loss of active component, and clarity 
changes for solutions; inability to resuspend the particles and loss of signifi cant 
amounts of the active component for suspensions; and creaming and breaking 
(or coalescence) for emulsions [1] . These instabilities are usually related to the 
following: 
Active and Primary Degradant. A liquid dosage form is stable while it remains 
within its product specifi cations. When chemical degradation products are 
known, for stability study and expiration dating, the regulatory requirements 
for the primary degradant of a active component are chemical structure, biological 
effect and signifi cance at the concentrations to be determined, mechanism 
of formation and order of reaction, physical and chemical properties, 
limits and methods for quantitating the active component and its degradant 
molecule at the levels expected to be present, and pharmacological action or 
inaction [45] . Examples of drugs in liquid dosage forms that are easily degraded 
are vitamins and phenothiazines [6] . 
Interactions with Closure Systems. Elastomeric and plastic container and closure 
systems release leachable compounds into the liquid dosage form, such as 
nitrosamines, monomers, plasticizers, accelerators, antioxidants, and vulcanizing 
agents [44] . Each type of container and closure with different composition 
and/or design proposed for marketing the drug or physician ’ s samples has to 
be tested and stability data should be developed. Containers should be stored 
upright, on their side, and inverted in order to determine if container – closure 
interactions affect product stability [6, 45] . 
Moisture Loss. When the containers are inappropriately closed, part of the 
vaporized solvent is released and the concentration and potency of the active 
component may be increased [6] . 
Microbiological Contamination. Inappropriate closure systems also increase the 
possibilities of microbial contamination when opening and closing containers 
[6] . 
4.3.4.10 Process Validation 
Objective Process validation has the objectives of identifying and controlling 
critical points that may vary product specifi cations through the manufacturing 
process [6] . 
Amount of Data To validate the manufacturing process, the manufacturer has to 
design and specify in the protocol the use of data sheets to keep information about 
the control of product specifi cations from each batch in - process as well as fi nished - 
product tests. Some formats are common to different products, though each type of 
product has some specifi c information to be kept on special sheets. Thus, the amount 
of data varies from one type of product to another [6] . 
Scale - Up Process Data obtained using special batches for the validation of the 
scale - up process are compared with data from full - scale batches and batches used 
for clinical essays [6] . 

338 LIQUID DOSAGE FORMS 
Product Specifi cations The most important specifi cations or established limits for 
liquid dosage forms are microbial limits and test methods, medium pH, dissolution 
of components, viscosity, as well as particle size uniformity of suspended components 
and emulsifi ed droplets. Effectiveness of the preservative system depends on 
the dissolution of preservative components and may be affected by the medium pH 
and viscosity. In addition, dissolved oxygen levels are important for components 
sensitive to oxygen and/or light [6] . 
Bioequivalence or Clinical Study In the patient, the general or systemic circulation 
is responsible for carrying molecules to different tissues of the body. To assure 
the expected bioactivity of a product, the amount of drug that reaches the systemic 
circulation per unit of time is analyzed and is known as bioavailability. Bioequivalence 
is the comparison of the bioavailability of a product with a reference product. 
While oral solutions may not always need bioequivalence studies because they are 
considered self - evidente, suspensions usually require bioequivalence or clinical 
studies in order to demonstrate effectiveness. However, OTC suspension products 
such as antacids are exempt from these studies [6] . 
Control of Changes to Approved Protocol The manufacturing process of a specifi c 
product is validated and approved internally by the quality control unit and externally 
by the FDA. Any change in the approved protocol has to be documented to 
explain the purpose and demonstrate that the change will not unfavorably affect 
product safety and effi cacy. Factors include potency and/or bioactivity as well as 
product specifi cations. However, the therapeutic activity and uniformity of the 
product are the main concerns after formulation and process changes [20] . 
4.3.5 LIQUID DOSAGE FORMS * 
Douche A liquid preparation, intended for the irrigative cleansing of the vagina, 
that is prepared from powders, liquid solutions, or liquid concentrates and 
contains one or more chemical substances dissolved in a suitable solvent or 
mutually miscible solvents. 
Elixir A clear, pleasantly fl avored, sweetened hydroalcoholic liquid containing 
dissolved medicinal agents; it is intended for oral use. 
Emulsion A dosage form consisting of a two - phase system comprised of at least 
two immiscible liquids, one of which is dispersed as droplets (internal or dispersed 
phase) within the other liquid (external or continuous phase), generally 
stabilized with one or more emulsifying agents. (Note: Emulsion is used as a 
dosage form term unless a more specifi c term is applicable, e.g. cream, lotion, 
ointment.). 
Enema A rectal preparation for therapeutic, diagnostic, or nutritive purposes. 
Extract A concentrated preparation of vegetable or animal drugs obtained by 
removal of the active constituents of the respective drugs with a suitable menstrua, 
evaporation of all or nearly all of the solvent, and adjustment of the 
residual masses or powders to the prescribed standards. 
* The defi nitions in this section are from ref. 11 . 

For Solution A product, usually a solid, intended for solution prior to 
administration. 
For Suspension A product, usually a solid, intended for suspension prior to 
administration. 
For Suspension, Extended Release A product, usually a solid, intended for suspension 
prior to administration; once the suspension is administered, the drug 
will be released at a constant rate over a specifi ed period. 
Granule, Effervescent A small particle or grain containing a medicinal agent in 
a dry mixture usually composed of sodium bicarbonate, citric acid, and tartaric 
acid which, when in contact with water, has the capability to release gas, resulting 
in effervescence. 
Inhalant A special class of inhalations consisting of a drug or combination of 
drugs, that by virtue of their high vapor pressure can be carried by an air 
current into the nasal passage where they exert their effect; the container from 
which the inhalant generally is administered is known as an inhaler. 
Injection A sterile preparation intended for parenteral use; fi ve distinct classes 
of injections exist as defi ned by the USP. 
Injection, Emulsion An emulsion consisting of a sterile, pyrogen - free preparation 
intended to be administered parenterally. 
Injection, Solution A liquid preparation containing one or more drug substances 
dissolved in a suitable solvent or mixture of mutually miscible solvents that is 
suitable for injection. 
Injection, Solution, Concentrate A sterile preparation for parenteral use which, 
upon the addition of suitable solvents, yields a solution conforming in all 
respects to the requirements for injections. 
Injection, Suspension A liquid preparation, suitable for injection, which consists 
of solid particles dispersed throughout a liquid phase in which the particles 
are not soluble. It can also consist of an oil phase dispersed throughout an 
aqueous phase, or vice - versa. 
Injection, Suspension, Liposomal A liquid preparation, suitable for injection, 
which consists of an oil phase dispersed throughout an aqueous phase in such 
a manner that liposomes (a lipid bilayer vesicle usually composed of phospholipids 
which is used to encapsulate an active drug substance, either within a 
lipid bilayer or in an aqueous space) are formed. 
Injection, Suspension, Sonicated A liquid preparation, suitable for injection, 
which consists of solid particles dispersed throughout a liquid phase in which 
the particles are not soluble. In addition, the product is sonicated while a gas 
is bubbled through the suspension and these result in the formation of microspheres 
by the solid particles. 
Irrigant A sterile solution intended to bathe or fl ush open wounds or body 
cavities; they ’ re used topically, never parenterally. 
Linament A solution or mixture of various substances in oil, alcoholic solutions 
of soap, or emulsions intended for external application. 
Liquid A dosage form consisting of a pure chemical in its liquid state. This 
dosage form term should not be applied to solutions. 
LIQUID DOSAGE FORMS 339

340 LIQUID DOSAGE FORMS 
Liquid, Extended Release A liquid that delivers a drug in such a manner to allow 
a reduction in dosing frequency as compared to that drug (or drugs) presented 
as a conventional dosage form. 
Lotion An emulsion, liquid dosage form. This dosage form is generally for 
external application to the skin. 
Lotion/Shampoo A lotion dosage form which has a soap or detergent that is 
usually used to clean the hair and scalp; it is often used as a vehicle for dermatologic 
agents. 
Mouthwash An aqueous solution which is most often used for its deodorant, 
refreshing, or antiseptic effect. 
Oil An unctuous, combustible substance which is liquid, or easily liquefi able, on 
warming, and is soluble in ether but insoluble in water. Such substances, 
depending on their origin, are classifi ed as animal, mineral, or vegetable oils. 
Rinse A liquid used to cleanse by fl ushing. 
Soap Any compound of one or more fatty acids, or their equivalents, with an 
alkali; soap is detergent and is much employed in liniments, enemas, and in 
making pills. It is also a mild aperient, antacid and antiseptic. 
Solution A clear, homogeneous liquid dosage form that contains one or more 
chemical substances dissolved in a solvent or mixture of mutually miscible 
solvents. 
Solution, Concentrate A liquid preparation (i.e., a substance that fl ows readily 
in its natural state) that contains a drug dissolved in a suitable solvent or 
mixture of mutually miscible solvents; the drug has been strengthened by the 
evaporation of its nonactive parts. 
Solution, for Slush A solution for the preparation of an iced saline slush, which 
is administered by irrigation and used to induce regional hypothermia (in 
conditions such as certain open heart and kidney surgical procedures) by its 
direct application. 
Solution, Gel Forming/Drops A solution, which after usually being administered 
in a drop - wise fashion, forms a gel. 
Solution, Gel Forming, Extended Release A solution that forms a gel when it 
comes in contact with ocular fl uid, and which allows at least a reduction in 
dosing frequency. 
Solution/Drops A solution which is usually administered in a drop - wise 
fashion. 
Spray A liquid minutely divided as by a jet of air or steam. 
Spray, Metered A non - pressurized dosage form consisting of valves which allow 
the dispensing of a specifi ed quantity of spray upon each activation. 
Spray, Suspension A liquid preparation containing solid particles dispersed in 
a liquid vehicle and in the form of coarse droplets or as fi nely divided solids 
to be applied locally, most usually to the nasal - pharyngeal tract, or topically 
to the skin. 
Suspension A liquid dosage form that contains solid particles dispersed in a 
liquid vehicle. 

Suspension, Extended Release A liquid preparation consisting of solid particles 
dispersed throughout a liquid phase in which the particles are not soluble; the 
suspension has been formulated in a manner to allow at least a reduction in 
dosing frequency as compared to that drug presented as a conventional dosage 
form (e.g., as a solution or a prompt drug - releasing, conventional solid dosage 
form). 
Suspension/Drops A suspension which is usually administered in a dropwise 
fashion. 
Syrup An oral solution containing high concentrations of sucrose or other 
sugars; the term has also been used to include any other liquid dosage form 
prepared in a sweet and viscid vehicle, including oral suspensions. 
Tincture An alcoholic or hydroalcoholic solution prepared from vegetable 
materials or from chemical substances. 
Notes : 
1. A liquid is pourable; it fl ows and conforms to its container at room temperature. 
It displays Newtonian or pseudoplastic fl ow behavior. 
2. Previously the defi nition of a lotion was “ The term lotion has been used to 
categorize many topical suspensions, solutions, and emulsions intended for 
application to the skin. ” The current defi nition of a lotion is restricted to an 
emulsion. 
3. A semisolid is not pourable; it does not fl ow or conform to its container at 
room temperature. It does not fl ow at low shear stress and generally exhibits 
plastic fl ow behavior. 
4. A colloidal dispersion is a system in which particles of colloidal dimension 
(i.e., typically between 1 nm and 1 . m) are distributed uniformly throughout a 
liquid. 
REFERENCES 
1. Crowley , M. M. ( 2005 ), Solutions, emulsions, suspensions, and extracts , in USIP , Remington: 
The Science and Practice of Pharmacy , Lippincott Williams & Wilkins , Philadelphia , 
pp. 745 – 774 . 
2. Block , L. H. ( 2002 ), Nonparenteral liquids and semisolids , in Levin , M. , Ed., Pharmaceutical 
Process Scale - Up , Marcel Dekker , New York , pp. 57 – 94 . 
3. Spurgeon , T. ( 2007 ), Quality by design in solid dosage processes: How will QbD impact 
manufacturing? Contract Pharm. , 3 – 1 . 
4. Allen , L. V. The art, Science, and Technology of Pharmaceutical Compounding . American 
Pharmaceutical Association (APhA) . Washington D.C. , 2002 . Pages 93, 95. 
5. Qu , A. , and Maglayo , A. ( 2003 ), Outsourcing stability services. Contract Pharma . 
11 – 12. Available at: http://www.contractpharma.com/articles/2003/11/outsourcing - 
stability - services 
6. USA Department of Health and Human Services: Food and Drug Administration (FDA) . 
Guides to inspections. 

342 LIQUID DOSAGE FORMS 
7. Kourounakis , P. N. ( 1994 ), Advanced Drug Design and Development , Taylor and Francis , 
p. 141 . 
8. Papich , M. G. ( 2005 ), Drug compounding for veterinary patients , AAPS J. , 7 ( 2 ), 
E281 – E287 . 
9. Wiberg , C. C. , Leppik , I. E. , and Cloyd , J. C. ( 2005 ), Lower phenytoin serum levels in 
persons switched from brand to generic phenytoin , Neurol. J. , 64 ( 8 ), 1485 – 1486 . 
10. Brooks , K. ( 2007 ), Pharma IT outsourcing: ERP consolidates IT infrastructure, Contract 
Pharm , March 2007, available: http://www.contractpharma.com/articles/2007/03/pharma - 
it - outsourcing 
11. USA Department of Health and Human Services, Food and Drug Administration (FDA) . 
Center for Drug Evaluation and Research (CDER) Data Standards Manual: Dosage 
Form (4/92) available at: http://www.fda.gov/cder/dsm/DRG/drg00201.htm . 
12. Drugs@FDA glossary of terms, U.S. Department of Health and Human Services, Food 
and Drug Administration (FDA), available: http://www.fda.gov/cder/drugsatfda/glossary. 
htm#L . 
13. DeLuca , P. P. ( 1992 ), Formulation of small volume parenterals , in Avis , K. E. , Lieberman , 
H. A. , and Lachman , L. , Eds., Pharmaceutical Dosage Forms: Parenteral Medications , Vol. 
1 , Marcel Dekker , New York . 
14. Allen , L. V. , Popovich , N. G. , and Ansel , H. C. , Ansel ’ s Pharmaceutical Dosage Forms and 
Drug Delivery Systems , Lippincott William & Wilkins , Baltimore , 2004 , Chapters 2 
and 5. 
15. SAP Information Center, Best practices for automotive: Pharmaceutical industry, Liquid 
dosage forms, available: http://help.sap.com/ . 
16. Sokoll , K. ( 2007 ), Optimizing drug development strategies: Exploring methods to improve 
the process, Contract Pharm . Marzo 2007. Available: http://www.contractpharma.com/ 
articles/2007/03/optimizing - drug - development - strategies 
17. Carleton , F. J. , and Agalloco , J. P. ( 1999 ), Validation of Pharmaceutical Processes: Sterile 
Products , Marcel Dekker , New York . 
18. Stacey , R. D. , Griffi n , D. , and Shaw , P. ( 2000 ), Complexity and Management: Fad or Radical 
Challenge to Systems Thinking? Taylor and Francis Group , Boca Raton, Fl . 
19. Hardman , J. G. , Limbrid , L. E. , Molinoff , P. B. , and Ruddon , R. W. Goodman and Gilman ’ s 
The Pharmacological Basis of Therapeutics , McGraw - Hill , New York , 2006 . 
20. Scott , R. R. ( 1989 ), A practical guide to equipment selection and operating techniques , 
in Lieberman , H. A. , Rieger , M. M. , and Banker , G. S. , Eds., Pharmaceutical Dosage Forms: 
Disperse Systems , Vol. 2 , Marcel Dekker , New York , pp 1 – 71 . 
21. Reepmeyer , J. C. , Revelle , L. K. , and Vidavsky , I. ( 1998 ), Detection of clobetasol propionate 
as an undeclared steroid in zinc pyrithione formulations by high - performance liquid 
chromatography with rapid - scanning ultraviolet spectroscopy and mass spectrometry . 
Journal of Chromatography A , Vol. 828 , Number 1 , 18 December 1998, pp. 239 – 246(8) . 
22. FARR Air Pollution Control (2005), FARR dust collectors, HEPA BIBO 
fi ltration systems, Pharmaceutical Industry Standard On - line, available: http://www. 
pharmaceuticalonline.com/downloads/detail.aspx?docid=74B77B1C - 0ED9 - 4E7B - A317 - 
5DD0D23124B3&VNETCOOKIE=NO . 
23. Biotrace International (2004), Air samplers. LIT055/002/29.06.04 . 
24. 3 - A Sanitary Standards, Technical Resource Center, resource papers, available: http:// 
www.3 - a.org/techresource/papers.htm . 
25. Cook , Jr. , J. L. ( 1998 ), Standard Operating Procedures and Guidelines , PennWell Books , 
Tulsa, OK , p. 318 . 

 
28. Thierauf , R. J. , and Hoctor , J. J. ( 2006 ), Optimal Knowledge Management: Wisdom Management 
Systems Concepts and Applications , Idea Group , Hershey, PA . 
29. Petru s evski , V. M. , and Metodija , Z. N. ( 2001 ), Volume non - additivity of liquid mixtures: 
Modifi cations to classical demonstrations , Chem. Ed. J. , 6 , 3 . 
30. Zhou , Q. , and Milton , R. ( 2003 ), Molecular interactions of surfactants in mixed monolayers 
at the air/aqueous solution interface and in mixed micelles in aqueous media: 
The regular solution approach , Am. Chem. Soc. , 19 ( 11 ), 4555 – 4562 . 
31. CANTY Process Technology ( 2007 ), CANTY Lighting Systems, available: http://www. 
jmcanty.com/images/overview/16pagermaster.pdf . 
32. Wikipedia, Non - Newtonian fl uid defi nition, available: http://en.wikipedia.org/wiki/Non - 
Newtonian_fl uid . 
33. Lieberman , H. A. , Rieger , M. M. , and Banker , G. S. ( 1988 ), Pharmaceutical Dosage Forms: 
Disperse Systems , Vol. 1 , Marcel Dekker , New York . 
34. Gibson , M. , Pharmaceutical Preformulation and Formulation: A Practical Guide from 
Candidate Drug Selection to Commercial Dosage Form , Interpharm , Boca Raton, FL , 
2004 , pp. 581 . 
35. Davidow , L. W. , Davidow , L. , and Thompson , J. E. , A Practical Guide to Contemporary 
Pharmacy Practice , Lippincott Williams & Wilkins , 2003. pp. 704. 
36. Sinko , P. J. Martin ’ s Physical Pharmacy and Pharmaceutical Sciences , Lippincott Williams 
& Wilkins , Baltimore, MD., 2005, pp. 795. 47. 
37. Nusim , S. ( 2005 ), Active Pharmaceutical Ingredients: Development, Manufacturing, and 
Regulation , Taylor and Francis Group , Boca Raton, FL . 
38. Chongprasert , S. , and Nail S. L. ( 1998 ), Infl uence of drug polymorphism on the 
physical chemistry of freeze - drying, Digital dissertations and theses, Purdue e - pubs, ETD 
Collection for Purdue University , available: http://docs.lib.purdue.edu/dissertations/ 
AAI9939331/ . 
39. Borka , L. ( 1971 ), The stability of chloramphenicol palmitate polymorphs. Solid and solution 
phase transformations , Acta Pharm. Suec. , 9 – 8 ( 4 ), 365 – 372 . 
40. Rosenholm , J. B. ( 2007 ), Wetting of surfaces and interfaces: A conceptual equilibrium 
thermodynamic approach , in Colloid Stability: The Role of Surface Forces, Part II , Tharwat 
F. Tadros Colloids and Interface Science Series, Vol. 2, Wiley-VCH, Weinheim. 
41. Lerk , C. F. , Lagas , M. , Lie - a - Huen , L. , Broersma , P. , and Zuurman , K. ( 1979 ), In vitro and 
in vivo availability of hydrophilized phenytoin from capsules , J. Pharm. Sci. , 5 – 68 ( 5 ), 
634 – 638 . 
42. Rosoff , M. ( 1988 ), Specialized pharmaceutical emulsions , in Lieberman , H. A. , Rieger , 
M. M. , and Banker , G. S. , Eds., Pharmaceutical Dosage Forms: Disperse Systems , Vol. 1 , 
Marcel Dekker , New York , pp. 245 – 283 . 
43. Moorthaemer , B. , Sprakel , J. ( 2006 ) Improving the stability of a suspension. Pharmaceutical 
Technology Europe, O1 February 2006. Available: http://www.ptemag.com/ 
pharmtecheurope/article/articleDetail.jsp?id=306687 
44. Allen , T. ( 1997 ), Particle Size Measurement , Chapmann and Hall , London . 
45. Connors , K. A. , Amidon G. L. , and Stella V. J. ( 1986 ), Chemical Stability of Pharmaceuticals: 
A Handbook for Pharmacists , Wiley - Interscience , New York . 
REFERENCES 343

344 LIQUID DOSAGE FORMS 
46. Miller , S. C. , and Drabik , B. R. ( 1984 ), Rheological properties of poloxamer vehicles , Int. 
J. Pharm. , 18 , 269 . 
47. Millard , J. W. , Alvarez - Nunez , F. A. , and Yalkowsky , S. H. ( 2002 ), Solubilization by 
cosolvents — Establishing useful constants for the log - linear model , Int. J. Pharm. , 245 ( 1 ), 
153 – 166 (14). 
48. Nash , R. ( 1996 ), Pharmaceutical suspensions , in Lieberman , H. A. , Rieger , M. M. , and 
Banker , G. S. , Eds., Pharmaceutical Dosage Forms: Disperse Systems , Vol. 2 , Marcel 
Dekker , New York , pp. 1 – 46 . 
49. Adeyeye , C. M. , and Price , J. C. ( 1997 ), Chemical dissolution stability and microscopic 
evaluation of suspensions of ibuprofen and sustained release ibuprofen - wax microspheres , 
J. Microencapsul. , 14 ( 3 ), 357 – 377 . 
50. Fong - Spaven , F. , and Hollenbeck , R. G. ( 1986 ), Thermal rheological analysis of triethanolamine 
- stearate stabilized mineral oil in water emulsions , Drug Dev. Ind. Pharm. , 12 , 
289 . 
51. Moelwyn-Hughes , E. A. (1961), Physical Pharmacy , Pergamon , New York , pp. 297 . 
52. Lan , C. W. ( 2005 ), Flow and segregation control by accelerated rotation for vertical Bridgman 
growth of cadmium zinc telluride: ACRT versus vibration , J. Crystal Growth , 274 
( 3 – 4 ), 379 – 386 . 
53. USP 29: U.S. Pharmacopeia and the National Formulary (USP 29/NF 24), United States 
Pharmacopeial Convention, Rockville, MD. 2006 . 
54. PMF Pharmaceutical Microbiology Forum Newsletters, available: http://www. 
microbiologyforum.org/news.htm . 
55. Sutton , S. ( 2006 ), The harmonization of the microbial limits test — Enumeration , Microbiol. 
Net. — PMF , 12 ( 3 ), 2 – 3 . 
56. Carstensen , J. T. , and Rhodes , C. T. ( 2000 ), Drug Stability: Principles and Practices , Macel 
Dekker , New York , pp. 773 . 
57. Robinson , E. P. ( 2006 ), Pseudomonas aeruginosa contamination of liquid antacids: A 
survey , J. Pharm. Sci. , 60 ( 4 ), 604 – 605 . 
58. Steer , H. W. , and Colin - Jones , D. G. ( 1975 ), Mucosal changes in gastric ulceration and 
their response to carbenoxolone sodium . Gut BMJ , 16 , 590 – 597 . 
59. Dreeszen , P. ( 2003 ), Biofi lm: Key to Understanding and Controlling Bacterial Growth in 
Automated Drinking Water Systems , Edstrom Industries , Waterford, WI , pp. 19 . 

SPECIAL/NEW DOSAGE FORMS 
SECTION 5


347 
5.1 
CONTROLLED - RELEASE 
DOSAGE FORMS 
Anil Kumar Anal 
Living Cell Technologies (Global) Limited, Auckland, New Zealand 
Contents 
5.1.1 Introduction 
5.1.2 Rationale 
5.1.3 General Design Principle for Controlled - Release Drug Delivery Systems 
5.1.4 Physicochemical and Biological Factors Infl uencing Design and Performance of 
Controlled - Release Formulations 
5.1.4.1 Physicochemical Factors 
5.1.4.2 Biological Factors 
5.1.5 Controlled - Release Oral Dosage Forms 
5.1.5.1 Anatomical and Physiological Considerations 
5.1.5.2 Fundamentals of Controlled - Release Oral Dosage Forms 
5.1.5.3 Factors Infl uencing Oral Controlled - Release Dosage Forms 
5.1.6 Design and Fabrication of Controlled - Release Dosage Forms 
5.1.6.1 Microencapsulation 
5.1.6.2 Nanostructure - Mediated Controlled - Release Dosage Forms 
5.1.6.3 Liposomes 
5.1.6.4 Niosomes 
5.1.7 Technologies for Developing Transdermal Dosage Forms 
5.1.8 Ocular Controlled - Release Dosage Forms 
5.1.9 Vaginal and Uterine Controlled - Release Dosage Forms 
5.1.10 Release of Drugs from Controlled - Release Dosage Forms 
5.1.10.1 Time - Controlled - Release Dosage Forms 
5.1.10.2 Stimuli - Induced Controlled - Release Systems 
5.1.11 Summary 
References 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

348 CONTROLLED-RELEASE DOSAGE FORMS 
5.1.1 INTRODUCTION 
Therapeutic value and pharmaeconomic value have in recent years become major 
issues in defi ning health care priorities under the pressure of cost containment [1] . 
The improvement in drug therapy is a consequence of not only the development of 
new chemical entities but also the combination of active substances and a suitable 
delivery system. The treatment of an acute disease or chronic illness is mostly 
accomplished by delivery of one or more drugs to the patient using various pharmaceutical 
dosage forms. Tablets, pills, capsules, suppositories, creams, ointments, 
liquids, aerosols, and injections are in use as drug carriers for many decades. These 
conventional types of drug delivery systems are known to provide a prompt release 
of the drug. Therefore, to achieve as well as to maintain the drug concentration 
within the therapeutically effective range needed for treatment, it is often necessary 
to take this type of drug several times a day, resulting in the signifi cant fl uctuation 
in drug levels [2] . For all categories of treatment, a major challenge is to defi ne the 
optimal dose, time, rate, and site of delivery. Recent developments in drug delivery 
techniques make it possible to control the rate of drug delivery to sustain the duration 
of therapeutic activity and/or target the delivery of drug to a specifi c organ or 
tissue. Many investigations are still going on to apply the concepts of controlled 
delivery for a wide variety of drugs [3] . 
5.1.2 RATIONALE 
The basic rationale for controlled drug delivery is to alter the pharmacokinetics and 
pharmacodynamics of pharmacologically active moieties by using novel drug delivery 
systems or by modifying the molecular structure and/or physiological parameters 
inherent in a selected route of administration. It is desirable that the duration 
of drug action become more a design property of a rate - controlled dosage form and 
less, or not at all, a property of the drug molecules ’ inherent kinetic properties. 
The rationale for development and use of controlled dosage forms may include 
one or more of the following arguments [4] : 
• Decrease the toxicity and occurrence of adverse drug reactions by controlling 
the level of drug and/or metabolites in the blood at the target sites. 
• Improve drug utilization by applying a smaller drug dose in a controlled - release 
form to produce the same clinical effect as a larger dose in a conventional 
dosage form. 
• Control the rate and site of release of a drug that acts locally so that the drug 
is released where the activity is needed rather than at other sites where it may 
cause adverse reactions. 
• Provide a uniform blood concentration and/or provide a more predictable drug 
delivery. 
• Provide greater patient convenience and better patient compliance by signifi - 
cantly prolonging the interval between administrations. 
However, there are also disadvantages attached to the use of controlled - release 
dosage forms. These include higher cost of manufacturing, unpredictability, poor in 

vitro/in vivo correlation, reduced potential, and poor systemic availability in general 
and the effective release period is infl uenced and limited by the gastrointestinal 
(GI) residence time [5] . The transit time of a dosage form through the GI tract is 
dependent on the physical characteristics of the formulation as well as on physiological 
factors such as stomach emptying time and effect of food on the absorption 
process. 
Only drugs with certain properties are suitable for controlled - release dosing. 
Characteristics that may make a drug unsuitable for controlled - release dosing 
include a long or short elimination half - life, a narrow therapeutic index, a large dose, 
low/slow solubility, extensive fi rst - pass clearance, and time course of circulating drug 
levels different from that of the pharmacological effect. The ideal drug delivery 
should be inert, biocompatible, mechanically strong, comfortable for the patient, 
capable of achieving high drug loading, simple to administer, and easy to fabricate 
and sterilize [6] . A range of materials have been employed to control the release of 
drugs and other active substances. Controlled - release dosage forms have been 
developed for over four decades. One of the fi rst practically used controlled - release 
oral dosage forms was the Spansule capsule, which was introduced in the 1950s. 
Spansule capsules were manufactured by coating a drug onto nonpareil particles 
and further coating with glyceryl stearate and wax. Subsequently, ion exchange 
resins were proposed for application as sustained - release delivery systems of 
ac cessible drug. Since then numerous products have been introduced and 
commercialized. 
5.1.3 GENERAL DESIGN PRINCIPLE FOR CONTROLLED - RELEASE 
DRUG DELIVERY SYSTEMS 
In the drug delivery system, the pharmacodynamics of active molecules becomes 
more a function of design and less one of inherent kinetic properties. Therefore, a 
deep understanding of the design of controlled - release systems of the pharmacokinetics 
and pharmacodynamics of the drug is required [7] . The conventional tablet 
or capsule provides only a single and transient burst of drug. A modifi cation introduced 
to the molecular structure of the drug (often used to decrease the elimination 
rate) or a system for modifi ed release rate is the common approaches used to 
increase the interval between two doses. The objective of both these approaches is 
to decrease the fl uctuations in plasma levels during multiple dosing. This allows the 
dosing interval to increase without compromising the required dosage levels. If the 
half - life of a drug is less than 6 h or the passage time in the smaller intestinal track 
is decreased, there might not be enough time to allow proper absorption, thus 
making frequent dosing compulsory. For other routes, where the residence time is 
not a constraint, dosing intervals can be as long as months or even years. 
A controlled - release drug delivery system serves primarily two functions [8] . 
First, it involves the transport of the drug to a particular part of the body. This may 
be accomplished in two ways, parenterally and nonparenterally. Second, the release 
of active ingredients occurs in a controlled manner, depending on the preparation 
of dosage forms. This determines the rate at which a drug is made available to the 
body once it has been delivered. Controlled drug delivery occurs when a biomaterial, 
either natural or synthetic, is judiciously combined with a drug or other active 
CONTROLLED-RELEASE DRUG DELIVERY SYSTEMS 349

350 CONTROLLED-RELEASE DOSAGE FORMS 
agent in such a way that the active agent is released from the material in a predesigned 
manner. To be successfully used in controlled drug delivery formulations, a 
material must be chemically inert and free of leachable impurities. It must also have 
an appropriate physical structure, with minimal undesired aging, and be readily 
processable. 
Controlled - release systems provide numerous benefi ts over conventional dosage 
forms. Conventional dosage forms are not able to control either the rate of drug 
delivery or the target area of administration and provide an immediate or rapid 
drug release. This necessitates frequent administration in order to maintain a therapeutic 
level. As a result, as shown in Figure 1 , drug concentrations in the blood 
fl uctuate widely. The concentrations of drug remain at a maximum value, which may 
represent a toxic level, or a level at which undersized side effects might occur, and 
a minimum value, below which the drug is no longer effective. The duration of 
therapeutic effi cacy is dependent upon the frequency of administration, the half - life 
of the drug, and the release rate of dosage forms. In contrast, controlled - release 
dosage forms not only are able to maintain therapeutic levels of drug with narrow 
fl uctuations but also make it possible to reduce the frequency of drug administration. 
The drug concentrations, as shown in Figure 1 , released from controlled - release 
dosage forms fl uctuate within the therapeutic range over a longer period of time. 
The plasma concentration profi le depends on the preparation technology, which 
may generate different release kinetics, resulting in different pharmacological and 
pharmacokinetic responses in the blood or tissues. 
The primary objectives of controlled drug delivery are to ensure safety and to 
improve effi cacy of drugs as well as patient compliance. This is achieved by better 
control of plasma drug levels and less frequent dosing. For conventional dosage 
forms, only the dose ( D ) and dosing interval ( . ) can vary above which undesirable 
or side effects are elicited. As an index of this window, the therapeutic index (TI) 
can be used. This is often defi ned as the ratio of lethal dose (LD 50 ) to median effective 
dose (ED 50 ). Alternatively, it can be defi ned as the ratio of maximum drug 
concentration ( C max ) in blood that can be tolerated to the minimum concentration 
( C min ) needed to produce an acceptable therapeutic response. 
FIGURE 1 Theoretical plasma concentration after administration of various dosage forms: 
( a ) standard oral dose; ( b ) oral overdose; ( c ) IV injection; ( d ) controlled - release system. 
Toxic level 
Minimum 
effective level 
c 
a 
b 
d 
Drug concentration in blood 
Time

Different types of modifi ed release systems can be defi ned [4, 8] : 
• Sustained release (extended release) that permits a reduction in dosing frequency 
as compared to the situation in which the drug is presented as a conventional 
form 
• Delayed release when the release of the active ingredient comes sometimes 
other than promptly after administration 
• Pulsatile release when the device actively controls the dosage released following 
predefi ned parameters 
In general, the sustained - release dosage form is designed to maintain therapeutic 
blood or tissue levels of the drug for an extended period of time. This is accomplished 
by attempting to obtain zero - order release from the designed dosage form. 
Zero - order release constitutes drug release from the dosage form that is independent 
of the amount of drug in the delivery system at a constant release rate. Systems 
that are designed for prolonged release can also be attributed as achieving 
sustained - release delivery systems. Repeat - action tablets are an alternative method 
of sustained release in which multiple doses of drugs are contained within a dosage 
form and each dose is released at a periodic interval, while delayed - release systems 
may not be sustaining, since often the function of these dosage forms is to maintain 
the drug within the dosage form. 
5.1.4 PHYSICOCHEMICAL AND BIOLOGICAL FACTORS 
INFLUENCING DESIGN AND PERFORMANCE OF 
CONTROLLED - RELEASE FORMULATIONS 
A number of variables, such as drug properties including stability, solubility, partitioning 
characteristics. charge and protein binding behavior, routes of drug delivery, 
target sites, acute or chronic therapy, the disease, and the patient, must be considered 
to establish the criteria for designing controlled - release products [9] . The performance 
of a drug in its release pattern from the dosage form as well as in the body 
proper is a function of its properties. These properties can at times prohibit placement 
if the drug is in a controlled - release form, restrict the route of drug administration, 
and signifi cantly modify performance for one reason or another. There is 
no clear distinction between physicochemical and biological factors since the biological 
properties of a drug are a function of its physicochemical properties while 
biological properties result from typical pharmacokinetic studies on the absorption, 
distribution, metabolism, and excretion (ADME) characteristics of a drug as well 
as those resulting from pharmacological studies. 
5.1.4.1 Physicochemical Factors 
Physicochemcial properties, such as aqueous solubility, partition coeffi cient and 
molecular size, drug stability, and protein binding, are those that can be determined 
from in vitro experiments. 
CONTROLLED-RELEASE FORMULATIONS 351

352 CONTROLLED-RELEASE DOSAGE FORMS 
Ionization, p K a 
, and Aqueous Solubility Most drugs are weak acids or bases. It 
is important to note the relationship between the p K a of the compound and the 
absorptive environment. Delivery systems that are dependent on diffusion or dissolution 
will likewise be dependent on the solubility of drug in the aqueous media. 
Since drugs must be in solution before they can be absorbed, compounds with very 
low aqueous solubility usually have the oral bioavailability problems because of 
limited GI transit time of the undissolved drug particles and they are limited at the 
absorption site. Unfortunately, for many of the drugs and bioactive compounds, the 
site of maximum absorption occurs at the site where solubility of these compounds 
is least. 
The drug (e.g., tetracycline) for which the maximum solubility is in the stomach 
but high absorption takes place in the intestinal region may be poor candidates for 
controlled - release systems, unless the system is capable of retaining the drug in the 
stomach and gradually releasing it to the small intestine or unless the solubility is 
made higher and independent of the external environment by encapsulating those 
compounds in a membrane system. Other compounds, such as digoxin [10] , with 
very low solubility, are inherently sustained, since their release over the time course 
of a dosage form in the gastrointestinal tract is limited by dissolution of the drug. 
Although the action of a drug can be prolonged by making it less soluble, this may 
occur at the expense of consistent and incomplete bioavailability. 
The choice of mechanism for oral sustained/controlled - release systems is limited 
by the aqueous solubility of the drug. Thus, diffusional systems are poor choices for 
low aqueous - soluble drugs since the driving force for diffusion, the concentration 
in aqueous solution, will be low. The lower limit for the solubility of a drug to be 
formulated in a controlled - release system has been reported to be 0.1 mg/mL [11] . 
Partition Coeffi cient and Molecular Size Following administration, drugs and 
other bioactive compounds must traverse a variety of membranes to gain access to 
the target area. The partition coeffi cient and molecular size infl uence not only the 
permeation of drug across biological membranes but also diffusion across or through 
a rate - controlling membrane or matrix. The partition coeffi cient is generally defi ned 
as the ratio of the fraction of drug in an oil phase to that of an adjacent aqueous 
phase. Drugs with extremely high partition coeffi cient (i.e., those that are highly oil 
soluble) readily penetrate the membranes but are unable to proceed further, while 
the excessive high aqueous - soluble compounds, having low oil/water partition coef- 
fi cients, cannot penetrate the membranes. A balance in the partition coeffi cient is 
needed to give an optimum fl ux for permeation through the biological and rate - 
controlling membranes. The ability of drugs to diffuse through membranes, also 
known as diffusivity, is related to its molecular size by the following equation: 
log log log D s V k s M k V V = . + = . + M M 
where D is the diffusivity, M is the molecular weight, V is the molecular volume, 
and s V , s M , k V , and k M are constants in a particular medium. Generally, there is smaller 
diffusivity with the denser medium. 
Drug Stability The stability of drug in the environment where it is to be exposed 
is an essential physicochemical factor to be considered before designing controlled 

dosage forms [12] . For example, orally administered drugs are subjected to both 
acid – base hydrolysis and enzymatic degradation [13] . For drugs that are unstable in 
the stomach, the dosage forms can be designed in so that they can be placed in a 
slowly soluble form or have their release delayed until they reach the intestine. This 
type of approach can be ineffective and the drug may be unstable in the small 
intestine or undergo extensive gut - wall metabolism. To obtain better bioavailability 
for such types of drugs, which are unstable even in the intestine, a different route 
of administration (e.g., transdermal with controlled - release dosage forms) can be a 
better option [14] . A transdermal patch of nitroglycerin is a good example. The 
details for transdermal dosage forms will be described later in this chapter. 
5.1.4.2 Biological Factors 
A drug, being a chemical/biological agent or a mixture of chemical and biological 
agents, is recognized as a xenobiotic by the human body. Subsequently, the drug will 
be prevented from entering the body and/or eliminated after its entry. As a result, 
the defense mechanisms of the human body become barriers to the delivery of 
drugs. A drug may encounter physical, physiological, enzymatic, or immunological 
barriers on its way to the site of action. Hence, the design of controlled - release 
product should be based on a comprehensive picture of drug disposition. This would 
entail a complex examination of the ADME characteristics of the drug. The details 
of these effects on various controlled - release dosage systems will be given in the 
following sections of this chapter. 
5.1.5 CONTROLLED - RELEASE ORAL DOSAGE FORMS 
Oral drug delivery is the preferred route for drug administration because of its 
convenience, economy, and high patient compliance compared with several other 
routes. About 90% of the drugs are administered via the oral route [15, 16] . For the 
oral controlled administration of drugs, several research and development activities 
have shown encouraging signs of progress in the development of programmable 
controlled - release dosage forms as well as in the search for new approaches to 
overcome the potential problems associated with oral drug administration. Many 
oral drugs are perceived as “ patient friendly ” for compliance, often requiring that 
the medication only needs to be taken once a day. The most prescribed drugs in the 
United States that use oral drug delivery technologies include Lipitor (atorvastatin 
calcium), manufactured by Pfi zer, and AstraZeneca ’ s Toprol - XL (metoprolo succinate). 
These potential developments and recently developed approaches are discussed 
here along with an overview of GI physiology. 
5.1.5.1 Anatomical and Physiological Considerations 
Anatomically, the alimentary canal can be divided into a conduit region and 
digestive and absorptive regions. The conduit region includes the mouth, pharynx, 
esophagus, and lower rectum. The digestive and absorptive regions include the 
stomach, small intestine, and all parts of the large intestine except the very distal 
region. 
CONTROLLED-RELEASE ORAL DOSAGE FORMS 353

354 CONTROLLED-RELEASE DOSAGE FORMS 
The role of the stomach in drug and nutrition absorption is very limited, and it 
acts primarily as a reception area for oral dosage forms. Nonionic, lipophilic molecules 
of moderate size can be absorbed through the stomach only to a limited extent 
owing to the small epithelial surface area and the short duration of contact with the 
stomach epithelium in comparison with the intestine [17] . The transit time in the GI 
tract varies from one person to another and also depends upon the physical properties 
of the object ingested and the physiological conditions of the alimentary canal 
(Table 1 ). After passing through the stomach, the next organ that a drug or bioactive 
compound encounters is the small intestine. The intestinal epithelium is composed 
of absorptive cells (enterocytes) interspersed with goblet cells (specialized for mucus 
secretion) and a few enteroendocrine cells (that release hormones). The enterocytes 
of intestinal epithelium are the most important cells in view of the absorption of 
drugs and nutrients [18] . Histologically, colonic mucosa resembles the small intestinal 
mucosa, the absence of villi being the major difference [19] . The microvilli of 
the large intestine enterocytes are less organized than those of the small intestine. 
The resulting decrease in the surface area of the colon leads to a low absorption 
potential in comparison with the small intestine. However, the colonic residence 
time is longer than that for the small intestine, providing extended periods of time 
for the slow absorption of drugs and nutrients [20] . Figure 2 shows the various 
physiological processes encountered by an orally administered drug during the 
course of GI transit. 
5.1.5.2 Fundamentals of Controlled - Release Oral Dosage Forms 
Oral controlled drug delivery is a system that provides the continuous delivery of 
drugs at predictable and reproducible kinetics for a predetermined period throughout 
the course of GI transit [21] . Also included are systems that target the delivery 
of a drug to a specifi c region within the gastrointestinal tract (GIT) for either local 
or a systemic action. All the oral controlled drug delivery systems have limited utilization 
in the GI controlled administration of drugs if the systems cannot remain 
in the vicinity of the absorption site for the lifetime of the drug delivery. In the 
exploration of oral controlled - release dosage forms, one encounters three areas of 
potential challenges [22] : 
1. Drug Delivery System To develop a viable oral controlled - release drug delivery 
system capable of delivering a drug at a therapeutically effective rate to a 
desirable site for the duration required for optimal effi cacy. 
2. Modulation of GI Transit Time To modulate the GI transit time so that the 
drug delivery system developed can be transported to a target site or to the 
TABLE 1 Gastrointestinal Tract: Physical Dimensions and 
Dynamics 
Region Surface area (m 2 ) pH 
Transit Time 
Fluid Solid 
Stomach 0.1 – 0.2 1.2 50 min 8 h 
Small intestine 100 6.8 2 – 6 h 4 – 9 h 
Large intestine 0.5 – 1 7.5 2 – 6 h 3 – 72 h 

vicinity of an absorption site and reside there for a prolonged period of time 
to maximize the delivery of a drug dose. 
3. Minimization of Hepatic First - Pass Elimination If the drug to be delivered 
is subjected to extensive hepatic fi rst - pass elimination, preventive measures 
should be devised to either bypass or minimize the extent of hepatic metabolic 
effect. 
With most orally administered drugs, targeting is not a primary concern, and it 
is usually intended for drugs to permeate to the general circulation and perfuse to 
other body tissues, except it is medicated intentionally for localized effects in the 
GIT. There is a general assumption that increasing concentration at the absorption 
site will increase the rate of absorption and, therefore, increase circulating blood 
levels, which in turn promotes greater concentrations of drug at the site of action. 
If toxicity is not an issue, therapeutic levels can thus be extended as shown in Figure 
3 . In essence, “ drug delivery ” by these systems usually depends on release from 
specifi c types of dosage forms and permeation through an epithelial membrane to 
the blood. Still, the biological and physicochemical factors that come across play 
important roles in the design of such systems. The physicochemical properties have 
been described earlier in this chapter while biological factors involved with oral 
dosage forms will be described below. 
FIGURE 2 Physical model illustrating various physiological processes during gastrointestinal 
transit. 
Stomach 
Liver 
Jejunum 
Pportal vein 
Ileum 
Colon 
Rectum 
Complexation 
Absorption 
S
y
s
t
e
m
i 
c
c
i 
r
c
u
l
a
t
i 
o
n 
Adsorption 
Hydrolytic 
Enzymatic 
Degradation within 
wall 
Hydrolytic 
Enzymatic 
Microbial 
Enterohepatic 
recycling 
Absorption 
Absorption 
Absorption 
Absorption 
Degradation 
CONTROLLED-RELEASE ORAL DOSAGE FORMS 355

356 CONTROLLED-RELEASE DOSAGE FORMS 
The degree to which a delivery system can achieve standard release profi les for 
a variety of chemically and physically diverse, pharmaceutically active molecules is 
a measure of a delivery system ’ s effi cacy and fl exibility (Figure 3 ). Among the most 
challenging profi les, linear, zero - order release of highly soluble actives over a 12 – 
24 - h period could be considered a reasonable performance standard against which 
delivery systems may be judged. 
5.1.5.3 Factors Infl uencing Oral Controlled - Release Dosage Forms 
Biological Half - Life The usual goal of an oral controlled - release dosage form is 
to maintain therapeutic blood levels, as shown in Figure 3 , over an extended period 
of time. A drug must be absorbed and enter the circulation at approximately the 
same rate at which it is eliminated. The elimination rate is quantitatively described 
by the half - life ( t 1/2 ). Each drug has its own characteristic elimination rate, which is 
the sum of all elimination process, including metabolism, urinary excretion, and all 
other processes that permanently remove drug from the bloodstream. 
Therapeutic compounds with short half - lives are excellent candidates for controlled/
sustained - release preparations, since this can reduce dosing frequency [23] . 
In general, drugs with half - lives shorter than 2 h, such as furosemide or levodopa, 
are poor candidates for controlled - release preparations. Compounds with longer 
half - lives, such more than 8 h, also do not need to be in the form of controlled 
release, since their effect is already sustained. Digoxin, warfarin and phenytoin are 
some examples [24 – 26] . However, drugs having even longer half - lives can be used 
in other forms of modifi ed release, such as pulsatile release. 
Gastrointestinal Tract and Absorption The design of a controlled - release dosage 
form should be based on a comprehensive picture of drug disposition. Both the 
pharmacokinetic property and biological response parameter have a useful range 
for the design of sustained - and controlled - release products. The potential problems 
inherent in oral controlled - release oral dosage forms generally relate to (i) interac- 
FIGURE 3 Profi le of drug level in blood: ( a ) traditional dosing of tablets; ( b ) controlled 
drug delivery dose. 
(a) 
Maximum desired level 
Minimum effective level 
Dose Dose Dose Dose 
Time 
(b) 
Maximum desired level 
Minimum effective level 
Time 
Drug level 
Drug level 

tions between the rate, extent, and location that the dosage form releases the drug 
and (ii) the regional differences in GI physiology [27] . 
Total GI transit time in the normal population varies from 5 to 36 h, with an 
average total transit time of approximately 24 h [28] . There is still much more to 
explore about the physiological processes involved and factors that infl uence gastric 
emptying, intestinal transit, and colon residence. One of the major factors is food 
administration, which delays the “ housekeeper wave, ” causing delay in gastric emptying. 
A high - fat meal may delay gastric emptying from 3 to 5 h, and the total GI 
transit delay is largely a function of delay in gastric emptying in this case. If we 
presume that the transit time of most drugs and devices in the absorptive areas of 
the GI tract is about 8 – 12 h, the maximum half - life for absorption should be approximately 
3 – 4 h; otherwise, the device will pass out of the potential absorptive regions 
before drug release is complete. That many controlled - release products are having 
somewhat lesser bioavailability than their conventional dosage forms may be due 
to incomplete release of the dosage form or release at such a slow rate that the drug 
has passed the actual site of absorption. Compounds that demonstrate a true lower 
absorption rate constant will probably be poor candidates for controlled - release 
dosage forms [28] . 
An understanding of the behavior of dosage forms in the stomach has been 
gained largely from scintographic studies in which phases of a meal and formulations 
are labeled with different nucleotides, particularly technetium - 99 and indium - 
111 [29] . Such studies have demonstrated that retention times of formulations in 
the stomach are dependent on the size of formulations and whether or not the formulation 
is taken with a meal. Enteric - coated or enteric matrix tablets may be 
retained for a considerable time if dosed with heavy meals or breakfast. Multiparticulate 
dosage forms will empty more slowly in the presence of food, and because 
the dosage forms mix evenly with the food, the entry into the small intestine will 
be strongly infl uenced by the caloric density and bulk of the ingested meal. The 
rate of gastric emptying, therefore, predicts the absorption behavior [29, 30] . However, 
the absorption of drugs from small, soft gelatin capsules is sometimes less 
predictable [31] . 
In the small intestine, contact time with the absorptive epithelium is limited, and 
the small intestinel transit time is 3.5 – 4.5 h [32] . The scintographic data show that 
many prolonged release products, particularly those intended for twice - or once - 
daily administration, actually release some of their drug contents in the colon where 
it may be absorbed into the systemic circulation for higher bioavailability. It is 
anticipated that conditions of dissolution, absorption, and metabolism in the distal 
portions of the intestine are different than in the proximal regions, due to differences 
in pH, lumen fl uid, mucosal morphology, and motility. 
For most formulations, colonic absorption represents the only real opportunity to 
increase the interval between doses. Transit through the lower part of the gut is 
quoted at about 24 h, but in reality only the ascending colonic environment has suffi - 
cient fl uid to facilitate dissolution. In the cecum, the fermentation of soluble fi ber 
produces fatty acids and gas [33] . The gas rises into the transverse colon and can form 
temporary pockets, restricting access of water to the formulation. Consequently, 
distal release of drug is associated with poor spreading, reduced surface area, and 
restricted absorption. In the colon, water availability is also low past the hepatic 
fl exure, as the ascending colon is extremely effi cient at water absorption [34] . 
CONTROLLED-RELEASE ORAL DOSAGE FORMS 357

358 CONTROLLED-RELEASE DOSAGE FORMS 
5.1.6 DESIGN AND FABRICATION OF CONTROLLED - RELEASE 
DOSAGE FORMS 
5.1.6.1 Microencapsulation 
Microencapsulation has been the subject of massive research efforts since its inception 
around 1950. Today, it is the mechanism utilized by approximately 65% of all 
sustained - release systems [35] . The technique ’ s popularity can be attributed mainly 
to its wide variety of applications. Hundreds of drugs have been microencapsulated 
and used as controlled - release systems. Some examples are Arthritis Bayer, 
Dexatrim Capsules, and Dimetapp Elixir. 
Microencapsulation provides more effi cient drug delivery because it increases 
the ability of the drug to interact with the body. The active ingredient of a drug is 
encapsulated into a particle that may be as small as 1 . m. The greatest feature of 
microencapsulation is the control provided by the choice of coating [36 – 38] . This 
control allows microencapsulation to be a controlled - release device. Microcapsules 
can be engineered to gradually release drugs to the body. To achieve this type of 
delivery, equilibrium is established which will monitor the liberation of medicine 
from those microcapsules. A microcapsule may be opened by many different 
means. Release mechanisms include fracture by heat, solvation, diffusion, and pressure 
[39] . A coating may also be designed to open specifi c areas of the body. A 
microcapsule containing drugs that will be consumed by GI fl uids must not be fractured 
until after it passes through the stomach [36 – 39] . A coating can therefore be 
used that is able to withstand stomach acids and allow the drug to pass through the 
stomach. 
Methods of Microencapsulation 
1. Air Suspension This method, known as the Wurster process or fl uidized - bed 
coating, involves dispersing solid particulate core materials in a supporting air 
stream and the spray coating of the suspended materials [40] . The design of the 
chamber and its operating parameters effect a recirculating fl ow of the particles 
through the coating zone of the chamber, where a coating material, usually a polymer 
solution, is sprayed onto the fl uidized particles. The cyclic process is repeated until 
the desired coat thickness is obtained. 
2. Pan Coating This process has been around for many decades and is commonly 
associated with sugar coating. It is essential that the particles be greater than 
600 . m for effective coating. The process has been extensively utilized for the preparation 
of controlled - release beads. 
The coating by this method is applied as a solution or as an atomized spray to the 
desired solid core material in the coating pan [41] . Warm air is passed over the coated 
materials as the coatings are applied in the coating pans to remove the coating 
solvent. Sometimes the fi nal solvent removal is carried out in a drying oven. 
3. Multiorifi ce Centrifugal Process This is a mechanical process involving the 
use of centrifugal forces to hurl a material particle through an enveloping microencapsulation 
membrane to effect mechanical encapsulation [42] . The microcapsules 
are then hardened and congealed. This method is capable of microencapsulating 
liquids and solids dispersed in a liquid. 

DESIGN AND FABRICATION OF CONTROLLED-RELEASE DOSAGE FORMS 359 
4. Coacervation Phase Encapsulation by coacervation is the one of the more 
popular methods commonly studied. The process consists of three steps carried out 
under continuous agitation [43] : 
Step 1 The core material is dispersed in a solution of coating polymer, the solvent 
for the polymer being the liquid manufacturing vehicle phase. 
Step 2 Deposition of the coating, accomplished by controlled, physical mixing of 
the coating material and the core material in the manufacturing vehicle. 
Step 3 Rigidization of the coating by thermal, cross - linking, or desolvation techniques 
to form self - sustaining microcapsules. 
Since the core materials are microencapsulated while being dispersed in 
some liquid manufacturing vehicle, subsequent drying operations are usually 
required. 
5. Solvent Evaporation Technique [39, 44, 45] The process is carried out in a 
liquid manufacturing vehicle. The core material to be encapsulated is dissolved 
or dispersed in the coating polymer solution. With agitation, the core coating 
material mixture is dispersed in the liquid manufacturing vehicle phase to obtain 
the appropriate microcapsule size. The microcapsules can be used in suspension 
form, coated onto substrates or isolated as powders. A schematic of the emulsifi cation/
solvent evaporation technique to prepare drug - loaded microparticles is shown 
in Figure 4 . 
6. Spray Drying Spray drying, by defi nition, is the transformation of feed from 
a fl uid state to a dried particulate form by spraying the feed into a hot drying 
medium [44, 46] . The feed can be a solution, suspension, or paste. A schematic is 
shown in Figure 5 . 
Spray drying consists of four process stages: 
(i) Atomization of feed into spray 
(ii) Spray – air contact (mixing and fl ow) 
(iii) Drying of spray (moisture evaporation) 
(iv) Separation of dried product from the air 
FIGURE 4 Schematic of microspheres prepared by emulsifi cation/solvent evaporation 
method. 
Polymer 
(e.g., chitosan) 
solution + drug 
14 h 
37°C 
1000 
rpm 
Cross linking 
agent, e.g, 
tripolyphosphate 
Filter wash 
n-Hexane 
Microsphere 
Span 85 
Cottonseed oil

360 CONTROLLED-RELEASE DOSAGE FORMS 
7. Spray Congealing [47] Spray congealing is similar to spray drying in that it 
involves dispersing the core material in a liquefi ed coating substance. Coat solidifi cation 
is accomplished by congealing the molten coating material or by solidifying a 
dissolved coating material by introducing the coat – core material mixture into a 
nonsolvent. Removal of the nonsolvent is then achieved by sorption, extraction, or 
evaporation techniques. Waxes, fatty acids and alcohol, and polymers and sugars, 
which are solids at room temperature but melt at high temperatures, are applicable 
to spray congealing methods. 
5.1.6.2 Nanostructure - Mediated Controlled - Release Dosage Forms 
The effi ciency of drug delivery to various parts of the body is directly affected by 
particle size. Nanostructure - mediated drug delivery, a key technology for the realization 
of nanomedicine, has the potential to enhance drug bioavailability, improve 
the timed release of drug molecules, and enable precision drug targeting [48] . 
Nanoscale drug delivery systems can be implemented within pulmonary therapies, 
as gene delivery vectors, and in stabilization of drug molecules that would otherwise 
degrade too rapidly. Additional benefi ts of using targeted nanoscale drug carriers 
are reduced drug toxicity and effi cient drug distribution. 
Anatomic features such as the blood – brain barrier, the branching pathways of 
the pulmonary system, and the tight epithelial junctions of the skin make it diffi cult 
for drugs to reach many desired physiological targets. Nanostructured drug carriers 
will help to penetrate or overcome these barriers to drug delivery [49] . Greater 
uptake effi ciency has also been shown for GI absorption and transcutaneous per- 
FIGURE 5 Schematic of spray drying method. 
Exhaust tube 
Smell 
clamp 
Cyclone 
Chamber head 
To compressor (atomizer) 
Sample tube 
To compressor (de-blocker) 
Main chamber 
Collection tube 
Jet nozzle 
Pump 
Sample 
Collection bottle

DESIGN AND FABRICATION OF CONTROLLED-RELEASE DOSAGE FORMS 361 
meation, with particles around 100 and 50 nm in size, respectively. However, such 
small particles traveling in the pulmonary tract may help with delivery to the pulmonary 
extremities. For instance, the outer layers of the carrier architecture may 
be formulated to biodegradable as the carrier travels through the pulmonary tract. 
As the drug carrier penetrates further into the lung, additional shedding will allow 
the encapsulated drug to be released. Biodegradable nanoparticles of gelatin and 
human serum albumin (HSA) show promise as drug carriers. 
Advantages of nanostructure - mediated drug delivery include the ability to deliver 
drug molecules directly into cells and the capacity to target tumors within healthy 
tissue [50] . The mechanisms of cellular uptake of external particulates include 
calthrin - and caveoli - mediated endocytosis, pinocytosis, and phagocytosis. However, 
phagocytosis may not play a role in the uptake of nanoscale particles because of 
the small size of such particles. 
Nanoscale drug delivery architectures are able to penetrate tumors due to the 
discontinuous, or “ leaky, ” nature of the tumor microvasculature, which typically 
contains pores ranging from 100 to 1000 nm in diameter. The microvasculature of 
healthy tissue varies by tissue type, but in most tissues, including the heart, brain, 
and lung, there are tight intercellular junctions less than 10 nm. Therefore, tumors 
within these tissue types can be selectively targeted by creating drug delivery nanostructures 
greater than the intercellular gap of the healthy tissue but smaller than 
the pores found within the tumor vasculature. 
Various nanoscale architecture can be designed, including solid spheres, hollow 
spheres, tubes, porous particles, solid particles, and branched structures (Table 2 ). To 
achieve such nanostructures, different fabrication methods are used depending on 
the types of material. The methods used for nanoscale assembly include molecular 
self - assembly, bioaggregation, nanomanipulation, photochemical patterning, molecular 
imprinting, layer - by - layer electrsostatic deposition, and vapor deposition. 
TABLE 2 Nanoscale Drug Delivery Technologies 
Drug 
Delivery 
Technology Materials Nanostructure Forms 
Biologicals Lipids, peptides, nucleic acids, 
polysaccharides, viruses 
Vesicles, nanotubes, rings, 
nanoparticles, nanocapsules, 
nanospheres 
Polymeric poly(lactic acid), poly(glycolic acid), 
poly(alkylcyanoacrylate), 
poly(3 - hydroxybutanoic acid), 
Poly(organophosphazene), 
poly(ethylene glycol), 
poly(caprolactone), poly(ethylene 
oxide), poly(amidoamine), 
poly(l - glutamic acid), 
poly(propylene imine) 
Vesicles, nanospheres, 
nanoparticles, micelles, 
dendrimers 
Silicon based Silicon, silicon dioxide Porous nanoparticles, nanoneedles 
Carbon based Carbon Nanotubes 
Metallic Gold, silver, palladium, platinum Nanoparticles, nanoshells 

362 CONTROLLED-RELEASE DOSAGE FORMS 
Manufacturing and Characterization of Nanoparticles/Nanocapsules/ 
Nanospheres Production of nanoparticles of soft materials is much more diffi cult 
and challenging than that of hard materials because of the high stickiness of the 
former. The bulk pharmaceuticals are available in solids of large sizes, which often 
can be easily solubilized in solvent to obtain particular sizes. Hence, there are two 
extremes of sizes: molecular size (each particle containing one molecule) and larger 
sizes (e.g., each particle containing on the order of 10 18 molecules). To obtain 
nanoparticles in the range of 50 – 300 nm of drug delivery, one requires on the order 
of 10 4 – 10 8 molecules in each particle. This size has to be achieved from either solution 
- phase (single - molecule) or millimeter - size particles (10 18 molecules). 
Pearl/Ball Milling Technology for Production of Drug Nanocrystals There are two 
different drug nanocrystal products, prepared by using pearl/ball milling technology. 
The Rapamune coated tablet is the more convenient formulation, introduced by 
Wyeth Pharmaceuticals in 2002. The Emend, introduced in 2003 by MSD, Sharp and 
Dohme Gmbh, is a capsule composed of sucrose, microcrystalline cellulose, hyperlose, 
and sodium dodecylsulfate [51] . 
Traditional equipment used for micronization of drug powders such as rotor - 
stator colloid mills or jet mills are of limited use for the production of nanocrystals. 
For example, jet milling leads to a drug powder with a size range of roughly 0.1 – 
20 . m, containing only a very small fraction of about 10% in the nanometer range. 
However, it has been shown when running a pearl mill over a suffi ciently long 
milling time that drug nanosuspensions can be obtained [52 – 54] . These mills consist 
of a milling container fi lled with fi ne milling pearls or larger sized balls. The container 
can be static and the milling material is moved by a stirrer; alternatively, the 
complete container is moved in a complex movement leading consequently to 
movement of the milling pearls. 
The different milling materials available include traditional steel, glass, and zircon 
dioxide as well as new special polymers such as hard polystyrene. A general problem 
associated with this technology is the erosion from the milling materials during 
manufacturing [55] . Surfactants and stabilizers have to be added for the physical 
stability of the produced nanosuspensions. In the production process, the coarse 
drug powder is dispersed by high - speed stirring or homogenization in a surfactant/ 
stabilizer solution to yield macro - and nanosuspensions. The choice of surfactant or 
stabilizers depends not only on the properties of the particles to be suspended 
(e.g., affi nity of surfactant/stabilizer to the crystal surface) but also on the physical 
principles (e.g., electrostatic or steric stabilization) and the route of administration. 
In general, steric stabilization is recommended as it is less susceptible to electrolytes 
in the gut and blood. 
There are number of pearl mills available on the market, ranging from 
laboratory - scale to industrial - scale volumes. The ability of large - scale production is 
an essential prerequisite for the introduction of a product to the market. One advantage 
of pearl mills, apart from their low cost, is their ability in scaling up. 
Nanoparticles/Nanoemulsions/Nanospheres Prepared by High - pressure Homogenization 
High - pressure homogenization is a technology that has been applied for 
many years in various areas of the production of emulsions and suspensions. A distinct 
advantage of this technology is its ease in scaling up, even to very large volumes. 

DESIGN AND FABRICATION OF CONTROLLED-RELEASE DOSAGE FORMS 363 
In the pharmaceutical industry, parenteral emulsions such as Intralipid and Lipofundin 
(mean droplet diameter 200 – 400 nm) are generally produced by this technology 
[56] . Typical pressures for the production of drug nanosuspensions are 1000 – 1500 
bars (corresponding to 100 – 150 MPa); the number of required homogenization 
cycles varies from 10 to 20 depending on the properties of the drug. Most of the 
homogenizers used are based on the piston gap principle; an alternative can be jet 
stream technology [57] . 
In the piston gap homogenizer, the liquid is forced through a tiny homogenization 
gap, typically in the range of 5 – 20 . m (depending on the pressure applied and the 
viscosity of the dispersion medium). Using a Micron Lab 40, the suspension is supplied 
from a metal cylinder by a piston, and the cylinder diameter is approximately 
3 cm. The suspension is moved by the piston having an applied pressure between 
100 and 1500 bars. The piston gap homogenizer corresponds to a tube system in 
which the tube diameter narrows from 3 to 20 . m. The Microfl uidizer (Microfl uidics 
Inc.) is based on the jet stream principle [58] . Two streams of liquid collide, diminution 
of droplets or crystals is achieved mainly by particle collision, but occurrence 
of cavitation is achieved mainly by particle collision. 
Nanoparticles/Nanocapsules Obtained by Interfacial Polymerization Nanoparticles/ 
nanocapsules can be obtained by fast polymerization of a monomer at the interface 
between the organic and the aqueous phase of an emulsion. Alkylcyanoacrylates 
have been proposed for the preparation of both oil - and water - containing nanocapsules 
[59] . These monomers polymerize within a few seconds, initiated by hydroxyl 
ions from equilibrium dissociation of water or by nucleophilic groups of any compound 
of the polymerization medium. 
1. Formation of Nanocapsules/Nanospheres Containing Oil Core This type of 
nanocapsule is preferred for the encapsulation of lipophilic and oil - soluble compounds. 
The general procedure for the preparation of oil - containing nanocapsules 
by interfacial polymerization of alkylcyanoacrylates consists of preparing a very fi ne 
oil - in - water (O/W) emulsion with an additional water - miscible organic solvent such 
as ethanol or acetone [60, 61] . These solvents are used to disperse the oil as very 
small droplets in the aqueous phase, which contains a hydrophilic surfactant. The 
solvent also serves as a vehicle for the monomer. Gallardo et al. [62] proposed a 
mechanism to explain nanocapsule formation. An important factor is that the 
organic solvent must be completely water miscible so that the formation of 
small enough oil droplets occurs spontaneously while the solvent is diffusing toward 
the aqueous phase and the water is diffusing toward the organic phase. The polymerization 
of monomer is also induced by contact with hydroxyl ions from the water 
phase, which should be very fast to allow effi cient formation of a thin layer of coating 
around the oil droplet and thus achieve effective encapsulation of drugs. 
The organic phase containing the oil, the monomer, and the bioactive compounds, 
dissolved in the water - miscible organic solvent, is injected into the aqueous phase 
containing water and a hydrophilic surfactant under strong magnetic stirring. The 
nanocapsules/nanospheres are formed immediately to give a milky suspension. The 
organic phase is then removed under reduced pressure using a rotary evaporator. 
The most commonly used materials for the preparation of oil - loaded nanocapsules 
are given in Table 3 . 

364 CONTROLLED-RELEASE DOSAGE FORMS 
Nanocapsule/nanosphere size ranges between 200 and 350 nm were observed to 
be affected by both the oil – ethanol ratio and the oil – monomer ratio [63, 64] . It is 
also infl uenced by the particular oil, water - miscible organic solvent, and nonionic 
surfactant in the aqueous phase. The pH of the aqueous phase and the temperature 
also affect the size distribution. 
2. Nanocapsules/Nanospheres Containing Aqueous Core Obtained by Interfacial 
Polymerization Nanocapsules/nanospheres with an aqueous core are a recent 
technology developed for the effi cient encapsulation of water - soluble compounds, 
which are generally very diffi cult to include within carrier systems. This type of 
nanocapsule/nanosphere may also be obtained by interfacial polymerization, but in 
this case monomers are added to a water - in - oil (W/O) emulsion. Anionic polymerization 
of the cyanoacrylates in the water phase is initiated at the interface by 
nucleophiles such as hydroxyl ions in the aqueous phase, leading to the formation 
of nanocapsules/nanospheres with an aqueous core. A typical procedure as described 
by Lambert et al. [65] consists of preparing an aqueous phase composed of ethanol 
(20% v/v) in water (pH 7.4) which is emulsifi ed in an organic phase containing 
Miglyol oil and Montane 80. Slow addition of cyanoacrylic monomer under mechanical 
stirring (about 4 h) allows the polymerization to occur. Thus the water droplets 
are surrounded by the polymer - forming nanocapsules with an aqueous core dispersed 
in an oily phase. 
Polymeric Nanocapsules/Nanospheres/Nanoparticles 
1. Manufacturing by Interfacial Nanodeposition/Nanoprecipitation The nanoprecipitation 
procedure generally consists of a water - miscible organic phase such as 
an alcohol or a ketone containing oil (with or without lipophilic surfactant) with an 
aqueous phase containing a hydrophilic surfactant. The polymer, which may be 
preformed synthetic, semisynthetic, or natural, is solubilized in the organic phase 
(or in a phase in which the polymer is soluble). After addition of the organic phase 
to the aqueous phase, the polymer diffuses with the organic solvent and is stranded 
at the interface between oil and water. The driving force for the formation of 
nanocapsules/nanospheres is the rapid diffusion of the organic solvent in the 
aqueous phase inducing interfacial nanoprecipitation of the polymer around droplets 
of the oily phase. The polymers which may be used to manufacture nanoparticles 
by this method include natural polymers such as gum arabic, chitosan, alginate, 
gelatin, ethylcellulose, hydroxypropyl methylcellulose (HPMC), and hydroxypropyl 
methylcellulose phthalate (HPMCP); semisynthetic polymers such as diacyl . - 
cyclodextrin; and synthetic polymers such as poly( d , l - lactide), poly( . - caprolactone), 
TABLE 3 Main Components Used to Prepare Oil - Containing Nanocapsules/ 
Nanospheres by Interfacial Polymerization 
Components Examples 
Oil Miglyol, Lipiodol, benzylbenzoate 
Monomers Ethylcyanoacrylate, isobutylcyanoacrylate, n - butylcyanoacrylate, 
isohexylcyanoacrylate 
Organic solvents Ethanol, acetone, acetonitrile, n - butanol, isopropanol 
Surfactants Poloxamer 188, poloxamer 238, poloxamer 407, Triton X100, Tween 80 

DESIGN AND FABRICATION OF CONTROLLED-RELEASE DOSAGE FORMS 365 
and poly(alkylcyanoacrylate), which are most commonly employed [65] . Similarly, 
a broad range of oils is suitable for the preparation of nanocapsules/nanospheres, 
including vegetable or mineral oils and pure compounds such as ethyl oleate and 
benzyl benzoate. The criteria for the selection of these compounds are the nontoxicity 
and the low solubility of the oil in the polymers and vice versa. 
Both hydrophilic and lipophilic surfactants can be used to stabilize the polymeric 
nanoparticles. Generally the lipophilic surfactant is a natural lecithin of relatively 
low phosphotidylcholine content, whereas the hydrophilic one is synthetic: anionic 
(lauryl sulfate), cationic (quaternary ammonium), or more commonly nonionic 
[poly(oxyethylene) - poly(propylene)glycol]. Nanoparticles can be prepared in the 
absence of surfactants, but there are lots more chances to get aggregated during 
storage. 
2. Nanoparticles Obtained by Multiple Emulsion/Solvent Evaporation Method 
The multiple emulsion/solvent evaporation method was initially developed for the 
preparation of microcapsules. This method consists in fi rst dissolving the drug in an 
aqueous solution with or without a surfactant and the polymer in a volatile organic 
solvent that is not miscible to water. Polymers used for the formation of such types 
of particles have been mainly poly(lactide - co - glycolide) and poly( . - caprolactone) 
[66, 67] . The inner water phase is then poured into the organic phase. This mixture 
is generally emulsifi ed forming the fi rst inner emulsion or the primary W/O emulsion, 
which is then mixed vigorously into an aqueous phase (outer water phase) that 
contains an emulsifi er forming the water - in - oil - in - water (W/O/W) multiple emulsion. 
The resulting multiple nanoemulsion is continuously stirred and the solvent is 
allowed to evaporate, inducing precipitation of polymer and, thereby, the formation 
of solid drug loaded nanoparticles. 
5.1.6.3 Liposomes 
Liposomes were discovered in the mid - 1960s [68] and were originally studied as cell 
membrane models. They have since gained recognition in the fi eld of drug delivery. 
Liposomes are formed by the self - assembly of phospholipid molecules in an aqueous 
environment. The amphophilic phospholipid molecules form a closed bilayer sphere 
in an attempt to shield their hydrophobic groups from the aqueous environment 
while still maintaining contact with aqueous phase via the hydrophilic head group. 
The resulting closed sphere may encapsulate aqueous soluble drugs within the 
central aqueous compartment or lipid - soluble drugs within the bilayer membrane. 
Alternatively, lipid - soluble drugs may be complexed with other polymers (e.g., 
cyclodextrin) and subsequently encapsulated within the liposome aqueous compartment. 
The encapsulation within/association of drugs with liposomes alters the drug 
pharmacokinetics. 
Attractive Biological Properties of Liposomes [69] 
• Liposomes are biocompatible. 
• Liposomes can entrap hydrophilic bioactive compounds in their internal compartment 
and hydrophobic into the membrane. 
• Liposome - incorporated bioactives are protected from the inactivating effect of 
external conditions yet do not cause undesirable side reactions. 

366 CONTROLLED-RELEASE DOSAGE FORMS 
• Liposomes provide a unique opportunity to deliver pharmaceuticals into cells 
or even inside individual cellular compartments. 
• The size, charge, and surface properties of liposomes can be easily changed by 
adding new ingredients to the lipid mixture before liposome preparation and/or 
by variation of preparation methods. 
The clinical applications of liposomes are well known (Table 4 ). The initial 
success achieved with many liposome - based drugs has fueled further clinical investigations. 
One of the drawbacks of the use of liposomes is the fast elimination from 
the blood and capture of liposomal preparations by the cells of the reticuloendothelial 
system (RES), primarily in the liver. 
There are a number of different types of liposomal vesicles [69] : 
• Multilamellar Vesicles These range in size from 500 to 5000 nm and consist of 
several concentric bilayers. 
• Small Unilamellar Vesicles These are around 100 nm in size and are formed 
by a single bilayer. 
• Large Unilamellar Vesicles These range in size from 200 to 800 nm. 
• Long Circulating Liposomes Different methods have been suggested to 
achieve long circulation of liposomes in vivo, including coating the liposome 
surface with inert, biocompatible polymers, such as polyethylene glycol (PEG), 
which form a protective layer over the liposome surface and slow down its 
recognition by opsonins and therefore subsequent clearance of liposomes. An 
important feature of protective polymers is their fl exibility, which allows a relatively 
smaller number of surface - grafted polymer molecules to create an impermeable 
layer over the liposome surface. These types of modifi ed liposomes 
demonstrate dose - dependent, nonsaturable , long - linear kinetics, and increased 
bioavailability. 
• Immunoliposomes To increase liposomal drug accumulation in the desired 
tissues and organs, the use of targeted liposomes with surface - attached ligands 
capable of recognizing and binding to cells of interest has been suggested [70] . 
Immunoglobulins (Ig) of the IgG class and their fragments are the most widely 
TABLE 4 Liposomal Drugs Approved for Clinical Application or Undergoing Clinical 
Evaluation 
Active Drug Product Name Applications 
Daunorubicin DaunoXome Sarcoma 
Doxorubicin Mycet Breast cancer 
Doxil/Caelyx Sarcoma, ovarian cancer, breast cancer 
Amphotericin B AmBisome Fungal infections 
Cytarabine DepoCyt Lymphomatous meningitis 
Vincristine Onco TCS Non - Hodgkin ’ s lymphoma 
Lurtotecan NX211 Ovarian cancer 
Nystatin Nyotran Topical antifungal agent 
All - trans retinoic acid Altragen Leukemia, carcinomas 
DNA plasmid encoding HLA - B7 
and .2 - microglobulin 
Allovectin - 7 Metastatic melanoma 

used targeting moieties for liposomes, which can be attached, without affecting 
liposomal integrity or the antibody properties, by covalent bonding to the liposome 
surface or by hydrophobic insertion into the liposomal membrane after 
modifi cation with hydrophobic residues. 
5.1.6.4 Niosomes 
The success achieved with liposomal formulations stimulated the search for other 
vesicle - forming amphiphiles. Nonionic surfactants were among the fi rst alternative 
materials studied and a large number of surfactants have since been found to self - 
assemble into closed bilayer vesicles which may be used for drug delivery [71] . 
Anticancer niosomes are expected to accumulate within tumors. The niosomal 
encapsulation of methotrexate and doxorubicin increases drug delivery to the tumor 
and tumoricidal activity. Unlike nonstealth liposomes, doxorubicin niosomes (size 
800 nm) possessing a triglycerol or doxorubicin niosomes (size 200 nm) possessing 
a muramic acid surface are not taken up signifi cantly by the liver. As such, these 
triglycerol niosomes accumulate in the tumor. However, muramic acid vesicles do 
accumulate in the spleen. Uptake by the liver and spleen make niosomes ideal for 
targeting diseases manifesting in these organs. One such condition is leishmaniasis, 
and number of studies have shown that niosomal formulations of sodium stibogluconate 
improve parasite suppression in the liver, spleen, and bone marrow. 
Niosomes may also be used as depot systems for short - acting peptide drugs on 
intramuscular administration [72] . 
Niosomal antigens are potent stimulators of the cellular and humoral immune 
response. The formulation of antigens as a niosome in W/O emulsions further 
increases the activity of antigens. The controlled - release property of these types 
of emulsion formulations is responsible for enhancing the immunological 
responses. 
5.1.7 TECHNOLOGIES FOR DEVELOPING TRANSDERMAL 
DOSAGE FORMS 
Continuous intravenous infusion at a programmed rate has been recognized as a 
superior model of drug delivery not only to bypass the hepatic fi rst - pass elimination 
but also to maintain a constant, prolonged, and therapeutically effective drug level 
in the body. A closely monitored intravenous infusion can provide both the advantages 
of direct entry of drugs into the systemic circulation and control of circulating 
drug levels. However, such a mode of drug delivery entails certain risks and therefore 
necessitates hospitalization of patients and close medical supervision of the 
medication. Recently there has been an increasing awareness that the benefi ts of 
intravenous drug infusion can be easily duplicated, without its potential hazards, by 
continuos transdermal drug administration through intact skin [73] . 
Advances in transdermal delivery systems (TDSs) and the technology involved 
have been rapid because of the sophistication of polymer science, which now allows 
incorporation of polymeric additives in TDSs in adequate quantity. Drugs with 
which transdermal therapy was pioneered include scopolamine, nitroglycerine, isosorbide 
dinitrite, clonidine, estradiol, nicotine, and testosterone [74] . 
TECHNOLOGIES FOR DEVELOPING TRANSDERMAL DOSAGE FORMS 367

368 CONTROLLED-RELEASE DOSAGE FORMS 
Advantages of Transdermal Drug Delivery System [75] 
• Avoids GI absorption (pH effects, enzymatic activity, drug interactions) 
• Substitute for oral route 
• Avoids fi rst - pass effect (drug deactivation by digestive and liver enzymes) 
• Multiday therapy with a single application 
• Extends the activity of the drugs with short half - lives 
• Provides capacity to terminate drug effects rapidly 
• Rapid identifi cation of medication in emergency 
Limitations of Transdermal Drug Delivery [75] 
• Not for all drugs 
• Limited time that the patch can remain affi xed 
• Variable intra - and interindividual percutaneous effi ciency absorption 
effi ciency 
• Variable adhesion to different skin types 
• Skin rashes and sensitization 
• Bacterial and enzymatic drug metabolism under the patch 
• Complex technology/high cost 
Skin Site for Transdermal Drug Administration The skin is one of the most 
extensive and readily accessible organs of the human body. The skin of an average 
adult body covers a surface area of approximately 2 m 2 and receives about one - third 
of the blood circulating through the body. It is elastic, rugged, and, under normal 
physiological conditions, self - regenerating. It serves as a barrier against physical and 
chemical attacks and shields the body from invasion by microorganisms. 
Microscopically the skin is a multilayered organ composed of, anatomically, many 
histological layers, but it is generally described in terms of three tissue layers: the 
epidermis, the dermis, and the subcutaneous fat tissue. 
Microscopic sections of the epidermis show two main parts: the stratum corneum 
and the stratum germinativum. The stratum corneum forms the outermost layer of 
the epidermis and consists of many layers of compacted, fl attened, dehydrated, 
keratinized cells in stratifi ed layers. In normal stratum corneum, the cells have a 
water content of only approximately 20% compared to the normal physiological 
level of 70% in the physiologically active 10% (w/w) to maintain fl exibility and 
softness. It becomes rough and brittle, resulting in so - called dry skin, when its moisture 
content decreases at a rate faster than can be resupplied from the underlying 
tissues. The stratum corneum is responsible for the barrier function of the skin. It 
also behaves as the primary barrier to percutaneous absorption. The thickness of 
this layer is mainly determined by the extent of stimulation of the skin surface by 
abrasion and weight bearing; hence thick palms and soles develop. 
Several technologies have been successfully developed to provide rate control 
over the release and skin permeation of drugs. These technologies can be classifi ed 
into four basic approaches which are described below [73, 75] . 

Polymer Matrix Diffusion - Controlled Transdermal Drug Delivery ( TDD ) 
System In this approach, the drug reservoir is formed by homogeneously dispersing 
the drug solids in a hydrophilic or lipophilic polymer matrix, and the medicated 
polymer formed is then molded into medicated disks with a defi ned surface area 
and controlled thickness. This drug - reservoir - containing polymer disk is then 
mounted onto an occlusive baseplate in a compartment fabricated from a drug - 
impermeable plastic baking (Figure 6 a ). 
FIGURE 6 Cross - sectional view of several TDSs: ( a ) poly(sebacic anhydride) (PSA) matrix 
device; ( b ) membrane - moderated TDS; ( c ) adhesive - controlled TDS; ( d ) microreservoir - type 
TDS; ( e ) matrix dispersion – type TDS. 
(a) 
Impermeable 
backing 
Adhesive matrix reservoir containing 
drug Drug reservoir 
Drug-impermeable metallic plastic 
laminate 
(b) 
Rate-controlling polymeric membrane 
Adhesive layer 
(c) 
Drugimpermeable metallic 
plastic laminate 
Adhesive layer 
Drug reservoir layer 
Rate-controlling adhesive 
membrane 
Metallic 
laminate 
Occlusive baseplate 
(aluminum foil disc) Adhesive foam pad 
(flexible polyurethane) 
(d) 
Adhesive rim 
Microscopic drug reservoirs 
Polymer matrix 
Absorbent pad Drug-impermeable 
plastic backing 
(e) Occlusive baseplate 
(aluminum foil disc) 
Adhesive rim 
r i o v r e s e r g u r D 
TECHNOLOGIES FOR DEVELOPING TRANSDERMAL DOSAGE FORMS 369

370 CONTROLLED-RELEASE DOSAGE FORMS 
Polymer Membrane Permeation - Controlled TDD System In this system, the drug 
reservoir is sandwiched between a drug - impermeable backing laminate and a rate - 
controlling polymeric membrane (Figure 6 b ). The drug molecules are permitted to 
release only through the rate - controlling polymeric membrane. 
Drug Reservoir Gradient - Controlled TDD System The rate of drug release from 
this type of drug reservoir type is gradient controlled. In this system the thickness 
of the diffusion path through which the drug molecule diffuses increases with time 
(Figure 6 c ). 
Microreservoir Dissolution - Controlled TDD System This type of drug delivery 
system can be considered a hybrid of the reservoir and matrix dispersion - type drug 
delivery systems. In this approach, the drug reservoir is formed by fi rst suspending 
the drug solids in an aqueous solution of water – miscible solubilizer (e.g., PEG), and 
then homogeneously dispersing the drug suspension with controlled aqueous solubility, 
in a lipophilic polymer, by high - shear mechanical force, to form thousands of 
unleachable microscopic drug reservoirs. This thermodynamically unstable dispersion 
is quickly stabilized by immediately cross - linking the polymer chains in situ, 
which produces a medicated polymer disk with a constant surface area and a fi xed 
thickness. Mounting the medicated disk at the center of an adhesive pad then produces 
a TDD system (Figure 6 d and e ). 
5.1.8 OCULAR CONTROLLED - RELEASE DOSAGE FORMS 
The eye is unique in its therapeutic challenges. The eye drop dosage form is easy to 
instill but suffers from the inherent drawback that the majority of the medication 
it contains is immediately diluted in the tear as soon as the eye drop solution is 
instilled. Usually less than 10% of a topically applied dose is absorbed into the eye, 
leaving the rest of the dose to potentially absorb into the bloodstream, resulting in 
unwanted side effects [76] . The objectives of most of the controlled delivery system 
are to maintain the drug in the precorneal area and allow its diffusion across the 
cornea. Polymeric matrices seem to reduce the drainage signifi cantly, but other 
newer methods of controlled - release dosage forms can also be used. 
The sustained release of artifi cial tears has been achieved by a hydroxypropylcellulose 
polymer insert [77] . However, the best known application of diffusional 
therapy in the eye, Ocusert - Pilo, as shown in Figure 7 , is a relatively simple structure 
with two rate - controlling membranes surrounding the drug reservoir containing 
FIGURE 7 Schematic of Ocusert intraocular device for controlled release of pilocarpine. 
White annular 
rings 
Pilocarpine reservoir 
Rate-controlling 
ethylene–vinyl 
acetate copolymer 
membranes

pilocarpine. The unit is placed in the eye and resides in the lowe cul - de - sac, just 
below the cornea. Since the device itself remains in the eye, the drug is released into 
the tear fi lm. The advantage of such a device is that it can control intraocular pressure 
for up to a week. Controlled release is achieved with less drug and fewer side 
effects, since the release of drug is zero order. However, it is diffi cult to keep it in 
the eye for a longer time and can cause discomfort. 
The prodrug administration is also getting attention as ocular controlled - release 
dosage forms. Since the corneal surface presents an effective lipoidal barrier, especially 
to hydrophilic compounds, it seems reasonable that a prodrug that is more 
lipophilic than the parent drug will be more successful in penetrating this barrier. 
Recently, dipivalyl epinephrine (Dividephrine), a dipivalyl ester of epinephrine, has 
been formulated [78] . Epinephrine itself is poorly absorbed owing to its polar characteristics 
and is highly metabolized. The prodrug form is 10 times as effective at 
crossing the cornea and produces substantially higher aqueous humor levels. 
New sustained technologies are also gaining much interest in ocular delivery, as 
in other routes. Liposomes as drug carriers have achieved enhanced ocular delivery 
of certain drugs, antibiotics, and peptides. Prolonged delivery of pilocarpine can be 
achieved with a polymeric dispersion or submicrometer emulsions [79] . 
5.1.9 VAGINAL AND UTERINE CONTROLLED - RELEASE 
DOSAGE FORMS 
Controlled - release devices for vaginal and uterine areas are most often for the 
delivery of contraceptive steroid hormones. The advantages are prolonged release, 
minimal side effects, and increased bioavailability. First - pass metabolism that inactivates 
many steroid hormones can also be avoided. 
Therapeutic levels of the medroxyprogestrone vaginal ring have been achieved 
at a total dose that is one - sixth the required oral dose [80] . The sustained release of 
progesterone from various polymers given vaginally has also been found useful in 
cervical ripening and the induction of labor. A possible new use of the vaginal route 
is for long - term delivery of antibodies. When various antibodies, including monoclonal 
IgG, were administered from polymer vaginal rings in test animals, antibody 
concentrations remained high over a month in vaginal secretions and detectable in 
blood serum [81] . 
The hormone - releasing devices in uterus have a closer resemblance to controlled 
release because they involve the release of a steroid compound by diffusion [82, 83] . 
Progesterone, the active ingredient, is dispersed in the inner reservoir, surrounded 
by ethlene/vinyl acetate copolymer membrane. The release of progesterone from 
this system is maintained almost constant for about a year [84 – 86] . 
5.1.10 RELEASE OF DRUGS FROM CONTROLLED - RELEASE 
DOSAGE FORMS 
There are three primary mechanisms by which active agents can be released 
from a delivery system: diffusion, degradation, and swelling followed by diffusion. 
Any or all of these mechanisms may occur in a given release system. Probable 
RELEASE OF DRUGS FROM CONTROLLED-RELEASE DOSAGE FORMS 371

372 CONTROLLED-RELEASE DOSAGE FORMS 
mechanisms of drug release from controlled - release dosage forms are briefl y 
described in Table 5 . Diffusion occurs when a drug or other active agent passes 
through the polymer that forms the controlled - release device. The diffusion can 
occur on a macroscopic scale — as through pores in the polymer matrix — or on a 
molecular level — by passing between polymer chains [4, 87] . 
Table 5 describes the probable mechanisms of drug delivery from controlled - 
release dosage forms under contain environmental conditions. A polymer and active 
agent have generally been mixed to form a homogeneous system, also referred to 
as a matrix system. Diffusion occurs when the drug passes from the polymer matrix 
into the external environment. As the release continues, its rate normally decrease 
with this type of system, since the active agent has a progressively longer distance 
to travel and therefore requires a longer diffusion time to release. The drug release 
is accomplished only when the polymer swells. Because many of the potentially most 
useful pH - sensitive polymers swell at high pH values and collapse at low pH values, 
the triggered drug delivery occurs upon an increase in the pH of the environment. 
Such materials are ideal for systems such as oral delivery, in which the drug is not 
released at low pH values in the stomach but rather at high pH in the upper small 
intestine. 
In reservoir systems, a reservoir — whether solid drug, dilute solution, or highly 
concentrated drug solution with in polymer matrix — is surrouned by a fi lm or membrane 
of a rate - controlling material [5] . The only structure effectively limiting the 
TABLE 5 Probable Mechanism of Drug Delivery from Hydrogels with Certain 
Environmental Conditions 
Stimulus Hydrogel Mechanism 
pH Acidic or basic 
hydrogel 
Change in pH, swelling – diffusion, erosion or 
burst release of drug 
Ionic strength Ionic hydrogel Change in ionic strength, change in 
concentration of ions inside gel, change in 
swelling, release of drug 
Chemical species Hydrogel containing 
electron - accepting 
groups 
Electron - donating compounds, formation of 
charge transfer complex, change in swelling, 
release of drug 
Enzyme – substrate Hydrogel containing 
immobilized 
enzymes 
Substrate present, enzymatic conversion, 
product changes, swelling of gel, release of 
drug 
Magnetic Magnetic particles 
dispersed in 
alginate 
microspheres 
Applied magnetic fi eld, change in pores in gel, 
change in swelling, release of drug 
Thermal Thermoresponsive 
hydrogel 
Change in temperature, change in polymer – 
polymer and water – polymer interactions, 
change in swelling, release of drug 
Electrical Polyelectrolyte 
hydrogel 
Apply electric fi eld, membrane charging, 
electrophoresis of charged drug, change in 
swelling, release of drug 
Ultrasound 
irradiation 
Ethylene – vinyl 
alcohol hydrogel 
Ultrasound irradiation, temperature increase, 
release of drug 

release of the drug is the polymer layer surrounding the reservoir. Since this polymer 
coating is essentially uniform and of a nonchanging thickness, the diffusion rate of 
the active agent can be kept fairly stable throughout the lifetime of the delivery 
system. 
For the diffusion - controlled systems ddescribed thus far, the drug delivery device 
is fundamentally stable in the biological environment and does not change its size 
through either swelling or degradation [4] . It is also possible for a drug delivery 
system to be designed so that it is incapable of releasing its agent or agents until it 
is placed in an appropriate biological environment. Swelling - controlled - release 
systems are initially dry and, when placed in the body, will absorb water or other 
body fl uids and swell. The swelling increases the aquenous solvent content with the 
formulation as well as the polymer mesh size, enabling the drug to diffuse through 
the swollen network into the external environment. Most of the materials used in 
swelling - controlled - release systems are based on hydrogels, which are polymers that 
well without dissolving when placed in water or other biological fl uids. 
5.1.10.1 Time - Controlled - Release Dosage Forms 
To achieve a drug release which is independent of the environment (e.g., pH, enzymatic 
activity, intestinal motility), the lag time prior to the drug release has to be 
controlled primarily by the delivery system. The release mechanisms employed 
include bulk erosion of polymers in which drug release by diffusion is restricted, 
surface erosion of layered devices composed of alternation drug - containing and 
drug - free layers, osmotically controlled rupture, and enzymatic degradation of liposomes. 
The device environment may modulate the release profi le of any of these 
systems and may depend on factors such as the amount of free moisture, regional 
blood fl ow, and various cellular activities at the site [88, 89] . 
Systmes with Eroding or Soluble Barrier Coatings These types of delivery systems 
comprise reservoir devices coated with a barrier layer. the barrier dissolves or 
erodes after a specifi ed lag period, after which the drug is released rapidly from the 
reservoir core. In general, the lag time prior to drug release from a reservoir - type 
device can be controlled by the thickness of the coating layer, for example, the 
Chronotropic systems, which consists of a drug - containing core layered with hydroxy 
propyl methyl cellulose (HPMC), optionally coated with an outer enteric coating. 
The lag time prior to drug release is controlled by the thickness and the viscosity 
grade of the HPMC layer. After erosion or dissolution of the HPMC layer, a distinct 
pulse was observed. The Chronotropic system [90, 91] is an oral dosage form designed 
to achieve time - controlled delivery. This system has been developed keeping in mind 
the interaction between GI fl uid and coating polymer, which causes time - or site - 
controlled release. The reaction causes the liberation of drugs by the mechanism of 
swelling of polymer, increased permeability, and dissolution/erosion phenomena. 
This system probably works better for poorly water soluble drug because highly 
water soluble drugs could diffuse through the swollen HPMC layer prior to complete 
erosion. 
The TIME CLOCK system for the oral dosage should enable fast and complete 
release of drug after a predetermined lag time [92] . A tablet was made containing 
the drug molecule and bulking agents (lactose, polyvinlpyrrolidine, corn starch, and 
RELEASE OF DRUGS FROM CONTROLLED-RELEASE DOSAGE FORMS 373

374 CONTROLLED-RELEASE DOSAGE FORMS 
magnesium stearate). This core was coated with a hydrophobic dispersion of carnauba 
wax, bees ’ wax, poly(oxyethylene) sorgitan monooleate, and HPMC in water. 
By altering the coating thickness, the lag time could be proportionally modulated. 
In vitro results indicated that the drug core was dissolved immediately after direct 
immersion in water and release was completed within 30 min, while a rapid release 
was observed after a certain lag time for the TIME CLOCK system with the hydrophobic 
coating. In vivo results revealed that drug disintergration was modulated by 
the coating thickness of the drug core as well as the food intake before drug administration. 
This approach may also be used to control the release onset time. Since 
the drug core is formulated with soluble ingredients, shell dissolution/distintegration 
becomes the key factor to control the lag time. Furthermore, drug release is independent 
of normal physiological conditions, such as pH, digestive state, and anatomical 
position at the time of release. This approach could be applicable for oral 
as well as for implant systems. Figure 8 illustrates the theoretical description of drug 
release from surface - eroding polymeric controlled - release dosage forms [93] . 
Systems with Rupturable Coatings This class of reservoir - type pulsatile release 
system is based on rupturable coatings. The drug is released from a core after rupturing 
of a surrounding polymer layer, caused by a pressure buildup within the 
system, as shown in Figure 9 [93] . The pressure necessary to rupture the coating can 
be achieved with gas - producing effervescent excipients, inner osmotic pressure, or 
swelling agents. 
An effervescent mixture of citric acid and sodium bicarbonate was incorporated 
in a tablet core coated with ethyl cellulose. The carbon dioxide development after 
water penetration into the core resulted in a pulsatile release after rupture of the 
coating, which was strongly dependent on the mechanical properties of the coating 
layer: The weak and nonfl exible ethyl cellulose fi lm ruptured suffi ciently when 
FIGURE 8 Theoretical controlled release from a surface - eroding polymeric system. 
( Adapted from ref. 93 with permission of Elsevier Copyright 1999 .) 
Polymer degradation 
with MW decrease 
until critical MW 
High-molecular-weight drug in 
bulk-eroding polymer 
(limited release by diffusion) 
Surface release 
Porous polymer matrix 
Release of remaining entrapped drug 
(booster release) 
Completion of polymer 
degradation by fragmentation 
and macrophage uptake

compared with more fl exible fi lms. The lag time before release increased with 
increasing coating level and increasing hardness of the core tablet. The effectiveness 
of so - called superdistintegrants, which are highly swellable agents, was demonstrated 
for a capsule - based system consisting of a drug containing a core capsule, 
and swelling layer, and a rupturable polymeric layer. Croscarmellose, sodium starch 
glycolate, or low - substituted hydroxypropyl cellulose (L - HPC) were used as swelling 
substances, which resulted in a complete fi lm rupture followed by a rapid drug 
release. The lag time is controlled by the composition of the outer polymer layer: 
Water - soluble polymers such as HPMC increase the permeability and therefore 
reduce the lag time. The swelling energy of several excipients decreased in the following 
order: croscamellose sodium > L - HPC > crospovidone > HPMC. Bothe solid 
and liquid drug formulations could be delivered with this system [94 – 96] . 
A novel capsule was made from ethyl cellulose for the time - controlled release 
of drugs in the colon [97] . Initially the ethyl cellulose capsule was prepared using a 
gelatin capsule with ethyl cellulose, followed by dissolution of the gelatin in water. 
The thickness of the ethyl cellulose capsule body was varied and the effect of wall 
thickness on the release of drugs in the capsules was investigated. Ethyl cellulose 
capsules contained a large number of mechanically made micropores (400 . m) at 
the bottom. Also located in the bottom of the capsule body was a swellable layer 
consisting of L - HPC. Above the swellable layer was the drug reservoir, which contained 
a mixture of the model drug, fl uorescein, and a bulking agent, such as lactose 
or starch. The capsule was thus capped and sealed with a concentrated ethyl cellulose 
solution. After administration of drug - containing capsule, water molecules 
penetrated the capsule through the micropores in the bottom of the capsule body. 
Hydration and swelling of HPC induced an increase in the internal osmotic pressure, 
which resulted in the “ explosion ” of the capsule and a burstlike drug release was 
observed. By altering the thickness of the capsule, the lag time of the drug release 
could be altered. A similar approach for the pulsatile release of drug was reported 
in which a hydrostatic pressure was generated inside the capsules. 
FIGURE 9 Theoretical controlled release from an osmotically driven system. ( Adapted 
from ref. 93 with permission of Elsevier Copyright 1999 .) 
Surface erosion of outer 
drug-containing layer 
Drug release from outer layer 
as surface erodes 
No drug release 
Drug-free layer 
Surface erosion of 
drug-free layer 
Drug release from core 
(booster release) 
RELEASE OF DRUGS FROM CONTROLLED-RELEASE DOSAGE FORMS 375

376 CONTROLLED-RELEASE DOSAGE FORMS 
Systems with Capsular Structure Several single - unit pulsatile dosage forms with 
a capsular design have been developed. Most of them consist of an insoluble capsular 
body which contains the drug and a plug which is removed after a predetermined 
lag time because of swelling, erosion, or dissolution. 
The Pulsincap system consists of a water - insoluble capsule body fi lled with the 
drug formulation. The capsule half is closed at the open end with a swellable hydrogel 
plug. The dimension and the position of the plug can control the lag time prior 
to the release. In order to assure a rapid release of the drug content, effervescent 
agents or disintegrants can be included in the drug formulation, in particular with 
water - insoluble drugs. The system is coated with an enteric layer which dissolves 
upon reaching the higher pH region of the small intestine. This system comprises 
insoluble capsules and plugs. The plugs consist either of swellable materials, which 
are coated with insoluble but permeable polymers (e.g., polymethacrylates), or of 
erodible substances, which are compressed (e.g., HPMC, polyvinyl alcohol, polyethylene 
oxide) or prepared by congealing of melted polymers (saturated polyglycolated 
glycerides of glyceryl monooleate). The erosion of the plug can also be 
controlled enzymatically: A pectin plug can be degraded by pectinolytic enzymes 
being directly incorporated into the plug [98 – 100] . 
Linkwitz et al. [101] described the delivery of agents from osmotic systems based 
on the technology of an expandable orifi ce. The system is in the form of a capsule 
from which the drug is delivered by the capsule ’ s osmotic infusion of moisture from 
the body. The delivery orifi ce opens intermittently to achieve a pulsatile delivery 
effect. The orifi ce forms in the capsule wall, which is constructed of an elastic material, 
preferably elastomer (e.g., styrene – butadiene copolymer), which strectches 
under apressure dufferential caused by the pressure rise inside the capsule as the 
osmotic infusion progessses. The orifi ce is small enough that when the elastic wall 
is relaxed, the fl ow rate of drug through the orifi ce is substantially zero, but when 
the elastic wall is stretched due to the pressure differential across the wall exceeding 
a threshold, the orifi ce expands suffi ciently to allow the release of the drug at a 
phsiologically required rate. This osmotically driven delivery device as an implant 
can used in the anal – rectal passageway, in the cervical canal, as an artifi cial gland, 
in the vagina, as ruminal bolus, and the like. 
A core - shelled cylindrical dosage form is available comprising a hydrophobic 
polycarbonate coating and a cylindrical core of alternating polyanhydride isolating 
layer and drug - loaded poly[ethyl glycinate) (benzyl amino acethydroxamate) phosphazene] 
(PEBP) layer for a programmable drug delivery system for single - dose 
vaccine and other related applications [102] . The pulsatile release of model compounds 
[fl uorescein isothiocyanate (FITC) – dextran and myoglobin] whith a certain 
lag time (18 – 118 h) was achieved on the basis of the pH - sensitive degradation of 
PEBP and its cooperative interaction with polyanhydrides. In another experiment, 
Jiang and Zhu [103, 104] designed laminated devices comprising of polyanhydrides 
as isolating layers and pH - sensitive complexes of poly(sebacic anhydride) - b - 
polyethylene glycol (PSA - b - PEG) and poly(trimellitytylimdoglycine - co - sebacic 
anhydride) -b - polyethylene glycol [P(TMA - gly - co - SA) - b - PEG] as protein - loaded 
layers. The release of model proteins [bovine serum albumin (BSA) and myoglobin] 
showed a typical pulsatile fashion. The lag time prior to the release correlated with 
the hydrolytic druation of polyanhydrides, which varied from 30 to 165 h depending 
on polymer type and isolating layer thickness. 

5.1.10.2 Stimuli - Induced Controlled - Release Systems 
Several polymeric delivery systems undergo phase transitions and demonstrate 
marked swelling – deswelling change in reponse to environmental changes, including 
solvent composition ionic strength, temperature, electric fi elds, and light [105] . 
Responsive drug release from those systems results from the stimuli - induced 
changes in the gels or micelles, which maydeswell, swell, or erode in response to 
the respecive stimuli. The mechanisms of drug release include ejection of the drug 
from the gel as the fl uid phase synerses out and drug diffusion along a concentration 
gradient. 
pH-Responsive Drug Release Dosage Forms pH - sensitive enteric coatings 
have been used routinely to deliver drug to the small intestine. These polymer 
coatings are insensitive to the acidic conditions of the stomach yet dissolve at 
the higher pH environment of the small intestine. This pH differential has 
also been attempted for colonic delivery purposes, although the polymers 
used for colonic targeting tend to have a threshold pH for dissolution that is 
higher than for those used in conventional enteric coating applications 
[104 – 106] . 
The synthesis and characterization of series of novel azo hydrogels for colon - 
targeting drug delivery have been described. The colon specifi city is achieved dure 
to the presence of pH - sensitive monomers and azo cross - linking agents in the 
hydrogel structures. Most commonly, copolymers of methacrylic acid and methylmethacrylate 
that dissolve at pH 6 (Eudragit L) and pH 7 (Eudragit S) have been 
extensively investigated [106, 107] . This approach is based on the assumption that 
gastrointestinal pH increases progressively from the small intestine to the colon. 
The pH in the distal small intestine is usually around 7.5, while the pH in the proximal 
colon is closer to 6. 
To overcome the premature release of drugs, a copolymer of methacrylic acid, 
methylmethacrylate, and ethyl acetate (Eudragit ES), which dissolves at a slower 
rate and at a higher threshold pH (7 – 7.5), has been developed [108] . The trn A series 
of in vitro dissolution studies with this polymer have highlighted clear benefi ts over 
the Eudragit S polymer for colon targeting. A gamma scintigraphy study comparing 
the in vivo performance of these various polymers revealed that Eudragit S (coating 
over the tablets) was superior to the older polymers in terms of retarding drug 
release in the small intestine, although, in some cases, the coated tablets did not 
break up at all. pH - sensitive delivery systems are commercially available for mesalazine 
(5 - iminosalicylic cid) (Asacol and Salofalk) and budesonide (Budenofalk and 
Entocort) for the treatment of ulcerative colitis and Crohn ’ s disease, respectively 
[109] . 
Natural polysaccharides are being used for the development of solid dosage 
forms for pH - dependent delivery and for targeting the release of drugs in colon 
[110] . Various major approaches utilizing polysaccharides are fermentable coating 
of the drug core, embedding of the drug in biodegradable matrix, and formation of 
drug – saccharide conjugate (prodrugs). A large number of polysaccharides have 
already been studied for their potential in these types of delivery systems, such as 
chitosan, alginate, pectin, chondroitin sulfate, cyclodextrin, dextrans, guar gum, 
inulin, amylose, and locust bean gum [111] . 
RELEASE OF DRUGS FROM CONTROLLED-RELEASE DOSAGE FORMS 377

378 CONTROLLED-RELEASE DOSAGE FORMS 
A pectin - and - galactomannan coating was developed by Lee et al. [112] . It consists 
of a conventional tablet coated with pectin and galactomannan. The coating 
from aqueous solutions of pectin and galactomannan was shown to be strong, elastic, 
and insoluble in gastric fl uid. Figure 10 shows the plasma concentration profi le of 
nifedifi ne from pectin – galactomannan - coated tablets and associated in vivo transit 
and disintegration characteristics. The mean plasma concentration of nifedifi ne was 
negligible for more than 5 h postdose and then increased rapidly. 
CODES Technology CODES is a unique colon - specifi c drug delivery technology 
that was designed to avoid the inherent problems associated with pH - or time - 
dependent systems [113, 114] . The design of CODES exploited the advantages of 
certain polysaccharides that are only degraded by microorganisms available in the 
colon [115] . This is coupled with a pH - sensitive polymer coating. Since the degradation 
of polysaccharides occurred only in the colon, this system exhibited the 
capability to achieve colon delivery consistently and reliably. As schematically presented 
in Figure 11 , one typical confi guration consists of a core table coated with 
three layers of polymer coatings. The fi rst coating (next to the core tablet) is an 
acid - soluble polymer (e.g., Eudragit E) and an outer coating is enteric with a 
HPMC barrier layer in between the oppositely charged polymers. The polysaccharides, 
degradable by enteroorganisms, generate organic acid, including mannitol, 
maltose, lactulose, and fructooligosaccharides. During the transit through the 
GI tract, CODES remains intact in the stomach, but the enteric and barrier coatings 
disolve in the intestines. In vivo performance of CODES in beagle dogs was 
studied using acetaminophen as the model drug and lactulose as the matrix - forming 
excipient in the core tablet. Compared with enteric - coated tablet, the onset of 
acetaminophen release form CODES was delayed more than 3 h, as shown in 
Figure 12 . 
FIGURE 10 Nifedipine plasma concentration – time profi le from pectin – galactomannan - 
coated tablets. ( Adapted from ref. 110 with permission of Elsevier Copyright 2002 .) 
0
1
2
3
4
5
6
7
8
9 
10 
Nifedipine concentration (ng/mL) 
0 3 6 9 12 15 16 24 21 
Time (h) 
Initial disintegration 
Complete disintegration 
Colon arrival time

COOES 
Enteric-coated core 
1400 
1200 
1000 
800 
600 
400 
200
0 
Time (h) 
APAP plasma concentration (ng/mL) 
0 2 4 6 8 10 12 14 16 
FIGURE 11 Conceptual design of CODES technology. ( Adapted from ref. 114 with permission 
of Elsevier Copyright 2002 .) 
In stomach 
In small intestime 
In colon 
Enter coating 
Acid-soluble 
polymer coating 
Lactulose 
Microflora 
Organic acid 
FIGURE 12 Percentage of acetaminophen released from CODES and enteric - coated core 
tablets in beagle dogs. ( Adapted from ref. 114 with permission of Elsevier Copyright 2002 .) 
Thermoresponsive Drug Release Dosage Forms Temperature is the most widely 
utilized triggering signal for a variety of modulated or pulsatile drug delivery systems. 
The use of temperature as a signal has been justifi ed by the fact that the body temperature 
often deviates from the physiological temperature (37 ° C) in the presence 
of pathogens or pyrogens. This deviation sometimes can be a useful stimulus that 
RELEASE OF DRUGS FROM CONTROLLED-RELEASE DOSAGE FORMS 379

380 CONTROLLED-RELEASE DOSAGE FORMS 
achivates the release of therapeutic agents from various temperature - responsive 
drug delivery systems for diseases accompanying fever. The drug delivery systems 
that are responsive to temperature utilize various polymer properties, including the 
thermally reversible coil/globule transition of polymer molecules, swelling change 
of networks, glass transition, and crystalline melting. 
Thermoresponsive hydrogels have been investigated as possible drug delivery 
carriers for stimuli - responsive drug delivery systems [116 – 118] . The common characteristics 
of temperature - sensitive polymers are the presence of hydrophobic 
groups, such as methyl, ethyl, and propyl groups. Of the many temperature - sensitive 
polymers, poly( N - isopropylacrylamide) (PIPPAm) is probably the most extensively 
used. PIPPAm cross - linked gels have shown thermoresponsive, discontinuous swelling/
deswelling phases — swelling, for example, at temperatures below 32 ° C while 
shrinking above this temperature. A sudden temperature increase above the transition 
temperature of these gels resulted in the formation of a dense, shrunken layer 
on the gel surface, which hindered water permeation from inside the gel into the 
environment. Drug release from the PIPPAm hydrogels at temperatures below 32 ° C 
was governed by diffusion, while above this temperature drug release was stopped 
completely, due to the “ skin layer ” formation on the gel surface (on – off drug release 
regulation). 
Swelling – deswelling kinetics of conventional cross - linked hydrogels are normally 
the reciprocal of the square of the gel dimension. This mobility of the cross - 
linked chains in the gel is affected by the surrounding chains and the 
swelling – deswelling phases of the gel are governed by the collective diffusions of 
the network chains. Thus, to accelerate structural changes of the gel in response 
to external stimuli, several approaches have been developed which form porous 
structure within the gel and decrease gel size. Kaneko et al. [119, 120] introduced 
a method to accelerate gel swelling – deswelling kinetics based on the molecular 
design of the gel structure by grafting the free mobile linear PIPPAm chains within 
the cross - linked PIPPAm hydrogels. These novel graft - type PIPPAm gels had the 
same transition temperature as conventional cross - linked PIPPAm gels and existed 
in the swollen state below the transition temperature, while above this temperature, 
they shrank. Adense skin layer formed on the conventional PIPPAm gels 
upon temperature change above the transition temperature, which limited the 
complete shrinkage of the gel. In contrast, the PIPPAm - grafted gels showed rapid 
deswelling kinetics without the formation of a skin layer on the gel surface. This 
is probably due to the rapid dehydration of the graft chains formed by hydrophobic 
aggregation on the three - dimensional cross - linked gel chains. The low - molecular 
- weight compounds released immediately from conventional PIPPAm gels after 
a temperature increase, after which the release was teminated due to the formation 
of a dense impermeable skin layer on the surface. In comparison, 65% of the drug 
was released in one burst from free PIPPAm - grafted hydrogels with a graft molecular 
weight (MW) of 9000 following the temperature increase. Graft - type gels with 
a molecular weight of 4000 showed oscillating drug release profi les. The release of 
high - molecular - weight compound (e.g., dextran, MW 9300) from PIPPAm graft - 
type gels was shown to burst after a temperature increase of 40 ° C. The difference 
in drug release profi les for two graft - type gels is probably due to the different 
strengths of aggregation forces between the formed hydrophobic cores within the 
graft - type gels. That is, the high - molecular - weight graft chains formed more hydro

phobic cores within the gels upon the temperature increase, which induced rapid 
gel deswelling. 
Temperature - sensitive hydrogels can also be placed inside a rigid capsule containing 
holes or apertures. The on – off release is achieved by the reversible volume 
change of temperature - sensitive hydrogels. Such a device is called a squeezing 
hydrogel device because the drug release is affected by the hydrogel dimension. In 
addition to temperature, hydrogels can be made to respond to other stimuli, such 
as pH. In this type of system, the drug release rate was found to be proportional to 
the rate of squeezing of the drug - loaded polymer. 
Clinical applications of thermosensitive hydrogels based on NIPAAm and its 
derivatives have limitations [121] . The monomers and cross - linkers used in the synthesis 
of the hydrogels are still not known to be biocompatible and biodegradable. 
The observation that acrylamide - based polymers activate platelets upon contact 
with blood, together with the unclear metabolism of poly(NIPAAm), requires 
extensive toxicity studies before clinical applications can merge. 
Recently some studies have been conducted on anocomposite hydrogels for 
photothermally modulated drug delivery. Gold nanoshells can be designed to absorb 
light strongly at desired wavelengths, in particular, in the near infrared between 800 
and 1200 nm, where tissue is relatively transparent [122] . When optically absorbing 
gold nanoshells are embedded in a matrix material, illuminating them at their resonance 
wavelength causes the nanoshells to transfer heat to their local environment. 
This photothermal effect can be used to optically modulate drug release from a 
nonshell polymer composite drug delivery system. To accomplish photothermally 
modulated release, the matrix polymer material must be thermally responsive. 
The authors observed the pulsatile release of insulin and other proteins in response 
to near - infrared irradiation when gold nanoshells were embedded in NIPAAm - co - 
acrylamide hydrogels [122] . 
Electroresponsive Release An electric fi eld as an external stimulus has advantages, 
such as the availability of equipment, which allow precise control with regard 
to the magnitude of current, duration of electric pulses, interval between pulses, and 
so on. Electrically responsive delivery systems are prepared from polyelectrolytes 
(polymers which contain relatively high concentration of ionizable groups along the 
backbone chain) and are thus pH responsive as well as electroresponsive. Under 
the infl uence of an electric fi eld, electroresponsive hydrogels generally deswell or 
bend, depending on the shape of the gel that lies parallel to the electrodes, whereas 
deswelling occurs when the hydrogel lies perpendicular to the electrodes. Synthetic 
(e.g., acrylate and methacrylae derivatives) as well as naturally occurring polymers 
(including hyaluronic acid, chondroitin sulfate, agarose, carbomer, xanthan gum, 
and calcium alginate separately or in combination) have been used. Complex multicomponent 
gels or interpenetrating networks have been prepared in order to 
enhance the gels or interpenetrating networks have been prepared in order to 
enhance the gel ’ s electroresponsiveness [123] . Electrically enhanced transdermal 
delivery provides the time - dependent delivery. Ionotophoresis, the electromigrational 
movement of charged molecules through the skin under a low - voltage 
and continuous driving force, has been reported for a number of bioactive compounds, 
such as leutinizaing hormone relesing hormone (LHRH), testosterone, and 
buserelin. 
RELEASE OF DRUGS FROM CONTROLLED-RELEASE DOSAGE FORMS 381

382 CONTROLLED-RELEASE DOSAGE FORMS 
Electronic Microelectromechanical System for Controlled Release Electronic 
microelectromechanical devices are manufactured using standard microfabrication 
techniques that are used to create silicon chips for computers, and they often have 
moving parts or components that enable some physical or analytical function to be 
performed by the device. Microfabrication techniques, the same processing techniques 
used to make microprocessors for computers and other microelectronic 
devices, have been used increasingly to produce microscale devices whose primary 
functions are mechanical, chemical and optical in nature. Such devices are commonly 
referred to as microelectromechanical systems (MEMS) and are found in 
ink - jet printers, automotive applications, and microtube engines in the aerospace 
industry. MEMS for biological applications are classifi ed as either microfl uidic 
devices or nonmicrofl uidic devices. The ultimate goal of MEMS is to develop a 
microfabricated device with the ability to store and release multiple chemical substances 
on demand by a mechanism devoid of moving its parts [124, 125] . A wide 
variety of microreservoirs, micropumps, cantilevers, rotors, channels, valves, sensors, 
and other structures have been fabricated, typically from the materials that have 
been demonstrated to be biocompatible and can be sterilely fabricated and hermetically 
sealed. The digital capabilities of MEMS may allow greater temporal control 
over drug release compared to traditional polymer - based systems, while the batch - 
processing techniques used in the microelectronics industry can lead to greater 
device uniformity and reproducibility than is currently available to the pharmaceutical 
industry. The use of MEMS for drug delivery necessitates the existence of drug 
depot or supply within or on the device. One straightforward approach to achieve 
this drug reservoir is the fabrication of silicon microparticles that contain an internal 
reservoir loaded with drug. These devices could be used for oral drug delivery, with 
release of the drug triggered by binding of a surface - functionalized molecule to cells 
in the digestive tract. 
The completely implantable minipump made by Minimed has a pulsatile, radio - 
controlled injection rate through a catheter into the intraperitoneal region [126] . 
One study found that patients with the implantable pump did not differ from control 
subjects on any meansure of psychosocial function but that puump users monitored 
their blood glucose levels more frequently and had lower average blood glucose 
levels. Even though this type of device may improve patient ’ s mbility and reduce 
infections by eliminating transcutaneous catheters, they may still be hampered by 
their size, cost, ability to deliver only drugs in solution, and limited stability of some 
drugs in solution at 37 ° C. Ikemoto and Sharpe [127] have developed a stepmotor 
micropump for the injection of nanoliter volumes of d - amphetamine solution into 
discrete brain regions of freely moving rats that was well tolerated. This micropump 
delivered a reliable volume of 50 nL per infusion over an hour at a rate of one infusion 
per minute. 
Another development in MEMS technology is the microchip. The microchip 
consists of an array of reservoirs that extend through an electrolyte - impermeable 
substrate. The prototype microchip is made of silicon and contains a number of drug 
reservoirs. Each reservoir is sealed at one end by a thin gold membrane of material 
that serves as an anode in an electrochemical reaction and dissolves when an electric 
potential is applied to it in an electrolyte solution. The reservoirs are fi lled with any 
combination of drug or drug mixtures in any form (i.e., solid, liquid, or gel) through 
the opening opposite the anode membrane by ink - jet printing or microinjection and 

are then sealed with a waterproof material. A cathode is also required for the electrochemical 
reaction to take place, and the cathode is usually made of the same 
conductive material as the anode to simplify the fabrication procedure. The device 
is submerged in an electrolyte solution containing ions and upon electric stimulation 
forms a soluble complex with the anode in its ionic form. When release is desired, 
an electric potential is applied between an anode membrane and a cathode, and the 
gold membrane anode is dissolved within 10 – 20 s and allows the drug in the reservoir 
to be released. This electric potential causes oxidation of the anode material to 
form a soluble complex with the electrolytes which when dissolves allowing release 
of the drug. Complex release patterns (such as simultaneous constant and pulsatile 
release) can be achieved from the microchips. The microchip has the ability to 
control both release time and release rate. The rate of release from a reservoir is a 
function of the dissolution rate of the materials in the reservoir, the diffusion rate 
of these materials out of the reservoir, or both. Therefore, the release rate from an 
individual reservoir can be tailored to a particular application by proper selection 
of the materials placed inside the reservoir [e.g., pure drug(s), drugs with polymers] 
[124, 125] . 
A microchip with insulin - fi lled reservoirs could eventually provide a better alternative 
for the treatment of insulin - dependent diabetes mellitus (IDDM) [125] . 
Because the microchip is capable of being programmed as well as integrated with 
other electronic devices, it is supposable that the microchip could be incorporated 
into a closed - loop biofeedback system. An electronic apparatus that continuously 
measures the blood glucose levels could provide the stimulus to the microchip and 
result in release of insulin into the bloodstream. Although such a system could still 
not perfectly mimic an endogenous system of healthy person, it could practically 
meet the needs of IDDM patients. Pulsatile release of synthetic gonadotropin – 
releasing hormone (GnRH) can be achieved with a programmed microchip. A subcutaneous 
implanted microchip containing 1000 drug reservoirs would be adequate 
to administer a month ’ s worth of drug therapy. The implanted microchip would be 
a convenient means to achieve the desired pharmacotherapeutic outcome of ovulation 
without interfering with the patient ’ s daily activities or causing phlebitis. 
While microchip drug delivery would be the most technologically advanced 
delivery system, it has itself limited storage capacity for therapeutic drugs [125] . 
Because most applications of this technology require implantation within bodily 
tissues, the question arises, “ What would be done when the chip runs out of drug? ” 
Some sort of procedure would be required to retrieve the empty chip cartridge once 
it has emptied. Due to the limited quantity of drug that can be stored on one 
chip, this technology is only ideal for potent drugs. If a larger dose of a medication 
is required, the chip would not be adequate for dispensing larger quantities 
of drug. 
Magnetically Induced Release Magnetic carriers receive their magnetic response 
to a magnetic fi eld from incorporated materials such as magnetite, iron, nickel, and 
cobalt. For biomedical applications, magnetic carriers must be water based, biocompatible, 
nontoxic, and nonimmunogenic. Earlier, Langer et al. [128] embedded magnetite 
or iron beads into a drug - fi lled polymer matrix and then showed that they 
could activate or increase the release of the drug from the polymer by moving a 
magnet over it or by applying an oscillating magnetic fi eld. When the frequency of 
RELEASE OF DRUGS FROM CONTROLLED-RELEASE DOSAGE FORMS 383

384 CONTROLLED-RELEASE DOSAGE FORMS 
the applied fi eld was increased from 5 to 11 Hz, the release of BSA from ethylenevinylacetate 
copolymer (EVAc) matrices slowed in a linear fashion. The rate of 
release could be modulated by altering the position, orientation, and magnetic 
strength of the embedded materials as well as by changing the amplitude of frequency 
of the magnetic fi eld. The micromovement within the polymer produced 
microcracks in the matrix and thus made the infl ux of liquid, dissolution, and effl ux 
of the drug. Done repeatedly, this would allow the pulsatile delivery of insulin. 
Another mechanistic approach based on magnetic attraction is the slowing down 
of oral drugs in the gastrointestinal system. This is possible by fi lling an additional 
magnetic component into capsules or tablets. The speed of travel through the 
stomach and intestines can then be slowed down at specifi c positions by an external 
magnet, thus changing the timing and/or extent of drug absorption into stomach or 
intestines. Slowing down the passage of magnetic liposomes with a magnet actually 
increased the blood levels of drug. Babincova et al. [129] developed magnetoliposomes 
for triggered release of drug. In their delivery systems, they entrapped 
dextran – megnetite and model drug 6 - carboxyfl uorescein in the liposomes and used 
laser to trigger the release of drug. The magnetite absorbs the laser light energy to 
heat the lipid bilayer above the gel – liquid crystal - phase transsition temperature Tc , 
which is 41 ° C for dipalmitoyl - phosphatidylcholine. Liposomes made from this lipid 
release their content as soon as the temperature reaches this level. They have also 
suggested that the absorption of laser energy by magnetite particles provides a 
means for localized heating and controlled release of liposome with a single laser 
pulse. This may have potential applications for selective drug delivery, especially to 
the eyes and skin. Even though the magnetic - modulated therapeutic approach 
is promising, it still needs very careful attention for a number of physical and 
magnetism - related properties. The magnetic force, which is defi ned by its fi eld 
and fi eld gradient, needs to be large and carefully shaped to activate the delivery 
system within the target area. The magnetic materials should be tissue stable 
and compatible. 
Chemically Induced Release 
Gluose - Responsive Insulin Release Device A decrease in or the absence of insulin 
secretion from pancreatic islets is the cause of diabetes mellitus. An effective glucose - 
responsive insulin delivery system should be composed of a glucose - sensing component 
and an insulin - releasing component. The sensing component detects a change 
in the glucose level and produces a signal that affects the releasing component. The 
magnitude of the signal increases with increasing glucose concentration, and so does 
the rate of insulin release. Based on this principle, various polymer - based glucose - 
responsive delivery systems have been designed, most of which are hydrogels that 
can alter their volume and degree of hydration in response to glucose concentration. 
Several systems have already been developed which are able to respond to glucose 
concentration changes, such as glucose oxidase (GOD), which catalyzes glucose 
oxidation [130] . Glucosylated insulin bound to concanavalin (Con) A was released 
through exchange with external glucose, due to the difference in their binding constants. 
This system needs direct injection of microcapsules into the peritoneal cavity 
of patients, which may cause undesirable side effects arising from the immune 
response to Con A if Con A was directly exposed to immune systems after breakage 

of microcapsules. Obaidat and Park [131] prepared a copolymer of acrylamide and 
allyl glucose. The side - chain glucose units in the copolymer were bound to Con A. 
These hydrogels showed a glucose - responsive, sol – gel phase transition dependent 
upon the external glucose concentration. The nonlinear dependence of this sol – gel 
phase transition with regard to the glucose concentration was due not only to the 
increased binding affi nity of allyl glucose to Con A compared to native glucose but 
also to the cooperative interaction between glucose - containing copolymer and Con 
A. Kataoka et al. [132] developed glucose and thermoresponsive hydrogels using 
acrylamidophenylboronic acid and N - isopropylacrylamide (IPAAm). The obtained 
gels, containing 10 mol % phenylboronic acid moieties, showed a transition temperature 
of 22 ° C in the absence of glucose. Below this temperature, the gels existed in 
a swollen state. The introduction of glucose to the medium altered the transition 
temperature of the gels in such a way that the transition temperature increased with 
increasing glucose concentration to reach 36 ° C at 5 g/L glucose concentration. 
Boronic acid was in equilibrium between the undissociated and dissociated forms. 
With increasing glucose concentration, the equilibrium shifted to increase the 
amount of dissociated boronate groups and gels became more hydrophilic. Although 
all of the glucose - sensitive insulin delivery systems are elegant and highly promising, 
many improvements need to be made for them to become clinically useful. First of 
all, the response of these systems upon changes in the environment occurs too 
slowly. In clinical situations, these systems need to respond to ever - changing glucose 
concentrations at all times, requiring hydrogels that can respond reproducibility and 
with rapid - response onset times on a long - term basis. An additional constant is that 
all the components used in the systems must be biocompatible. 
Chemotactic Factor - Induced Controlled - Release Systems With physical or chemical 
stress such as injury and broken bones, an infl ammation reaction takes place at 
the injured site. At the infl ammatory sites, infl ammation-responsive phagocytic cells 
such as macrophages and polymorphonuclear cells play a role in healing the injury. 
During infl ammation, hydroxyl radicals (OH • ) are produced from the cells. Yui and 
co - workers [133, 134] developed infl ammatory - induced hydroxyl radicals and 
designed drug delivery system which responded to the hydroxyl radicals 
and degraded in a limited manner. They used hyaluronic acid (HA), a linear aminopolysaccharide 
composed of repeating disaccharide subunits of N - acetyl - d - 
glucosamine and d - guluronic acid. In the body, HA is mainly degraded by 
hyaluronidase, or hydroxyl radicals. Degradation of HA via the enzyme is very low 
in a normal state of health. Degradation via hydroxyl radicals, however, is usually 
dominant and rapid when HA is injected at infl ammatory sites. These authors prepared 
cross - linked HA with ethyleneglycol diglydylether or polyglycerol polygluycidalether 
. These HA gels degraded only when the hydroxyl radicals were generated 
through the reaction between the iron (Fe 2+ ) ions and the hydrogen peroxide in 
vitro. Thus, a surface erosion type of degradation was achieved. When microspheres 
were incorporated in the HA hydrogels as a model drug, these microspheres were 
released when hydroxyl radicals induced HA gel degradation. Furthermore, 
degradation of HA in vivo tests showed that HA gels are degraded only when 
infl ammation was induced by surgical incision. Control HA gels were stable over 
100 days. It is possible to treat locally in infl ammatory diseases such as rheumatoid 
arthritis using anti - infl ammatory drug incorporated in HA gels [135] . 
RELEASE OF DRUGS FROM CONTROLLED-RELEASE DOSAGE FORMS 385

386 CONTROLLED-RELEASE DOSAGE FORMS 
5.1.11 SUMMARY 
As pharmaceutical scientists have increased their knowledge of pharmacokinetics 
and pharmacodynamics, it has become apparent that these factors can result in more 
effi cacious drugs. The number of new drug entities appearing on the market yearly 
has declined and pharmaceutical manufacturers have shown a renewed interest in 
improving existing dosage forms and developing more sophisticated drug delivery 
systems, including those employing the principles of controlled drug release. 
Current research in this area involves numerous new and novel systems, many 
of which have strong therapeutic potential. In this chapter, we have tried to emphasize 
the importance of oral routes as well as others, such as ocular, transdermal, 
intrauterine, and vaginal. The various microencapsulation, nanoencapsulation, and 
liposome technologies and the release of drugs and bioactive compounds from such 
products have been described. 
REFERENCES 
1. Bhalla , H. L. ( 1999 ), Drug delivery research in India: A challenge and an opportunity , 
J. Controlled Release , 62 , 65 – 68 . 
2. Breimer , D. D. ( 1999 ), Future challenges for drug delivery , J. Controlled Release , 62 , 
3 – 6 . 
3. Chien, Y. W. (1992), Novel Drug Delivery Systems , Marcel Dekker , New York , pp. 1 , 2 . 
4. Brannon - Peppas , L. ( 1997 ), Polymers in controlled drug delivery , Biomaterials , 11 , 
1 – 14 . 
5. Kydoneieus , A. F. ( 1980 ), Controlled Release Technologies: Methods, Theory, and Applications 
, CRC Press , Boca Raton, FL . 
6. Murano , E. ( 1998 ), Use of natural polysaccharides in the microencapsulation techniques , 
J. Appl. Ichthyol. 14 , 245 – 249 . 
7. Junginger , H. E. , and Verhoef , J. C. ( 1998 ), Macromolecules as safe penetration enhancers 
for hydrophilic drugs: A fi ction , Pharm. Sci. Today , 1 , 375 – 376 . 
8. Robinson , J. R. , and Lee , V. H. L. ( 1987 ), Controlled Drug Delivery: Fundamentals and 
Applications , Marcel Dekker , New York . 
9. Hofman , A. F. , Pressman , J. H. , Code , C. F. , and Witztum , K. F. ( 1983 ), Controlled entry of 
orally - administered drugs: Physiological considerations . Drug Dev. Ind. Pharm. , 9 , 1077 . 
10. Manninen , V. , Melin , J. , and Reissel , P. ( 1972 ), Tablet disintegration: Possible link with 
biological availability of digoxin , Lancet , 26 , 490 – 491 . 
11. Fincher , J. H. ( 1968 ), Particle size of drugs and its relationship to absorption and activity , 
J. Pharm. Sci. , 57 , 1825 – 1835 . 
12. Lee , V. H. , and Robinson , J. R. ( 1987 ), Sustained and Controlled Release Drug Delivery 
Systems , Marcel Dekker , New York , p. 71 . 
13. Fix , J. A. ( 1996 ), Oral controlled release technology for peptides: Status and future prospectus 
. Pharm. Res. , 13 , 1760 – 1763 . 
14. Chien , Y. W. ( 1983 ), Logics for transdermal controlled drug administration , Drug Dev. 
Ind. Pharm. , 9 , 497 – 520 . 
15. Gandhi , R. , Kaul , C. L. , and Panchangula , R. ( 1999 ), Extrusion and spheronization in 
the development of oral controlled - release dosage forms , Pharmaceutical Science and 
Technology Today , 2 , 160 – 170 . 

16. Anal , A. K. ( 2007 ), Time - controlled pulsatile delivery systems for bioactive compounds, 
Recent Patents Drug Deliv . Formulat. , 1 , 73 – 79 . 
17. Chan , O. H. , and Stewart , B. H. ( 1996 ), Physico - chemical and drug delivery considerations 
for oral drug bioavaibility . Drug Discovery Today , 1 , 461 – 472 . 
18. Wilson , C. G. ( 2000 ), Gastrointestinal transit and drug absorption , in Oral Drug Absorption: 
Prediction and Assessment , Marcel Dekker , New York , pp. 1 – 9 . 
19. Knutson , L. , Knutson , F. , and Knutson , T. ( 2000 ), Permeability in the gastrointestinal 
tract , in Oral Drug Absorption: Prediction and Assessment , Marcel Dekker , New York , 
pp. 11 – 16 . 
20. Waterbeemd , H. V. ( 2000 ), Intestinal permeability: Prediction from theory , in Oral Drug 
Absorption: Prediction and Assessment , Marcel Dekker , New York , pp. 31 – 49 . 
21. Chien , Y. W. ( 1992 ), Novel Drug Delivery Systems , Vol. I, Marcel Dekker , New York , 
pp. 139 – 196 . 
22. Jantzen , G. M. , and Robinson , J. R. ( 2000 ), Sustained and controlled release drug delivery 
systems , in Modern Pharmaceutics , Marcel Dekker , New York , pp. 501 – 528 . 
23. Welling , P. G. ( 1983 ), Oral controlled drug adminstration: Pharmacokinetic considerations 
. Drug Dev. Ind. Pharm. , 9 , 1185 – 1125 . 
24. Hofman , A. F. , Dressman , J. H. , Code , C. F. , and Witztum , K. F. ( 1983 ), Controlled entry 
of oral administered drugs: Physiological considerations , Drug Dev. Ind. Pharm. , 9 , 
1077 – 1083 . 
25. Fara , J. W. ( 1983 ), Gastrointestinal transit of solid dosage forms . Pharm. Technol. , 7 , 
23 – 26 . 
26. Meyer , J. H. , Ohashi , H. , Jehn , D. , and Thomson , J. B. ( 1981 ), Size of particles emptied 
from the human stomach , Gastroenterology , 80 , 1489 – 1496 . 
27. Welling , P. G. , and Dobrinska , M. R. ( 1987 ), Dosing considerations and bioavailability 
assessment of controlled drug delivery systems , in Controlled Drug Delivery: Fundamentals 
and Applications , Marcel Dekker , New York , pp. 253 – 289 . 
28. Hinton , J. M. , Lennard - Jones , J. E. , and Young , A. C. ( 1969 ), Alimentary canal transit 
time for an inert object in humans , Gut , 10 , 842 . 
29. Frier , M. , and Perkins , A. C. ( 1994 ), Radiopharmaceuticals and the gastrointestinal tract , 
Eur. J. Nucl. Med. Mol. Imaging , 21 , 1234 – 1242 . 
30. Basit , A. W. , Podczeck , F. , Newton , M. , Waddington , W. A. , Ell , P. J. , and Lacey , L. F. 
( 2004 ), The use of formulation technology to assess regional gastrointestinal drug 
absorption in humans , Eur. J. Pharm. Sci. , 21 , 179 – 189 . 
31. Delie , F. , and Blanco - Pr i eto , M. J. ( 2005 ), Polymeric particulares to improve oral bioavailability 
of peptide drugs , Molecules , 10 , 65 – 80 . 
32. Walter , E. , Kissel , T. , and Amidon , G. E. ( 1996 ), The intestinal peptide carrier: A potential 
transport system for small peptide - derived drugs , Adv Drug Deliv. Rev. , 20 , 33 – 58 . 
33. Scheline , R. R. ( 1973 ), Metabolism of foreign compounds by gastrointestinal microorganisms 
, Pharmacol. Rev. , 25 , 451 – 523 . 
34. Edwards , C. ( 1997 ), Physiology of colorectal barrier . Adv. Drug Deliv. Rev. , 28 , 173 – 190 . 
35. Whateley , T. L. ( 1992 ), Microencapsulation of Drugs , Harwood Academic , Geneva, 
Switzerland . 
36. Anal , A. K. , and Stevens , W. F. ( 2005 ), Chitosan - alginate multilayer beads for controlled 
release of ampicillin , Int. J. Pharm. , 290 , 45 – 54 . 
37. Anal , A. K. , Bhopatkar , D. , Tokura , S. , Tamura , H. , and Stevens , W. F. ( 2003 ), Chitosan - 
alginate multilayer beads for gastric passage and controlled intestinal release of protein , 
Drug Dev. Ind. Pharm. , 29 , 713 – 724 . 
REFERENCES 387

388 CONTROLLED-RELEASE DOSAGE FORMS 
38. Hui , H. W. , Lee , V. H. L. , and Robinson , J. R. ( 1987 ), Design and fabrication of oral 
controlled release drug delivery systems , in Controlled Drug Delivery: Fundamentals and 
Applications , Marcel Dekker , New York , pp. 373 – 421 . 
39. Anal , A. K. , and Singh , H. ( 2007 ), Recent advances in microencapsulation of probioitcs 
for industrial applications and targeted delivery , Trends Food Sci. Technol. , 18 , 240 – 251 . 
40. Mathir , Z. M. , Dangor , C. M. , Govender , T. , and Chetty , D. J. ( 1997 ), In vitro characterization 
of a controlled - release chlorpheniramine maleate delivery system prepared by 
the air - suspension technique , J. Microencapsul. , 14 , 743 – 751 . 
41. Deasy , P. B. ( 1991 ), Microencapsulation of drugs by pan and air suspension techniques , 
Crit. Rev. Ther. Drug Carrier Syst. , 8 , 39 – 89 . 
42. O ’ Connor , R. E. , and Schwartz , J. B. ( 1989 ), Extrusion and spheronization technology , 
in Pharmaceutical Pelletization Technology , Marcel Dekker , New York , pp. 187 – 215 . 
43. Gopferich , A. , Alonso , M. J. , and Langer , R. ( 1994 ), Development and characterization 
of microencapsulated microspheres , Pharm. Res. , 11 , 1568 – 1574 . 
44. Anal , A. K. , Stevens , W. F. , and L o pez , C. R. ( 2006 ), Ionotropic cross - linked chitosan 
microspheres for controlled release of ampicillin , Int. J. Pharm. , 312 , 166 – 173 . 
45. Weidenauer , U. , Bodmeier , D. , and Kissel , T. ( 2003 ), Microencapsulation of hydrophilic 
drug substances using biodegradable polyesters. Part I: Evaluation of different techniques 
for the encapsulation of pamidronate di - sodium salt , J. Microencapsul. , 20 , 
509 – 524 . 
46. Cui , J. H. , Goh , J. S. , Park , S. Y. , Kim , P. H. , and Le , B. J. ( 2001 ), Preparation and physical 
characterization of alginate microparticles using air atomization method , Drug Dev. Ind. 
Pharm. , 27 , 309 – 319 . 
47. Purvis , T. , Vaughan , J. M. , Rogers , T. L. , Chen X. , Overhoff , K. A. , Sinswat , P. , Hu , J. , 
McConville , J. T. , Johnston , K. P. , and Williams , R. O. ( 2006 ), Cryogenic liquids, nanoparticles, 
and microencapsulation , Int. J. Pharm. , 4 , 43 – 50 . 
48. Charman , W. N. , Chan , H. K. , Finnin , B. C. , and Charman , S. A. ( 1999 ), Drug delivery: 
A key factor in realizing the full therapeutic potential of drugs , Drug Dev. Res. , 46 , 
316 – 327 . 
49. Kayser , O. , Lemke , A. , and Hern a ndez - Trejo , N. ( 2005 ), The impact of nanobiotechnolgy 
on the development of new drug delivery systems , Curr. Pharm. Biotechnol. , 6 , 3 – 5 . 
50. Roco , M. C. ( 2003 ), Nanotechnology: Convergence with modern biology and medicine , 
Curr. Opin. Biotechnol. , 14 , 337 – 346 . 
51. Merck & Co. ( 2004 ), Drug Information: Emend, capsules, available: www.merck.com. 
52. Liversidge , G. G. , Cundy , K. C. , Bishop , J. F. , and Czekai , D. A. ( 1992 ), Surface modifi ed 
drug nanoparticles, Un. St. Patent 5,145,684 , Sterling Drug, New York. 
53. Merisko - Liversidge , E. , Liversidge , G. G. , and Cooper , E. R. ( 2003 ), Nanosizing: A formulation 
approach for poorly water - soluble compounds , Eur. J. Pharm. Sci. , 18 , 
113 – 120 . 
54. Merisko - Liversidge , E. , Sparpotdar , P. , and Bruno , J. ( 1996 ), Formulation and antitumor 
activity evaluation of nanocrystalline suspension of poorly soluble anticancer drugs , 
Pharm. Res. , 13 , 272 – 278 . 
55. Buchmann , S. , Fischli , W. , Thiel , F. P. , and Alex , R. ( 1996 ), Aqueous microsuspension, an 
alternative intravenous formulation for animal studies, paper presented at the 42nd 
Annual Congress of the International Association for Pharmaceutical Technology, 
Mainz, p. 124 . 
56. M u ller , R. H. , and Heinemann , S. ( 1989 ), Surface modeling of microparticles as parenteral 
systems with high tissue affi nity , in Bioadhesion - Possibilities and Future Trends , 
Wissenschaftliche Verlagsgesellschaft , Stuttgart , pp. 202 – 214 . 

57. M u ller , R. H. , and Peters , K. ( 1998 ), Nanosuspensions for the formulation of poorly 
soluble drugs: I: Preparation by a size - reduction technique , Int. J. Pharm. , 160 , 229 – 
237 . 
58. Maa , Y. F. , and Hsu , C. C. ( 1998 ), Performance of sonication and microfl uidisation for 
liquid - liquid emulsifi cation , Pharm. Dev. Technol. , 4 , 233 – 240 . 
59. Roller , J. M. , Covereur , P. , Robolt - Treupel , L. , and Puisieux , F. ( 1986 ), Physicochemical 
and morphological characterization of polyisobutyl cyanoacrylates nanocapsules , 
J. Pharm. Sci. , 75 , 361 . 
60. Chouinard , F. , Kan , F. W. , Leroux , J. C. , Foucher , C. , and Lenaerts , V. ( 1991 ), Preparation 
and purifi cation of polyisohexylcyanoacrylate nanocapsules , Int. J. Pharm. , 72 , 211 . 
61. Ammoury , N. , Fessi , H. , Devissaguet , J. P. , Ouisieux , F. , and Benita , S. ( 1989 ), Physicochemical 
characterization of polymeric nanocapsules and in vitro release evaluation of 
indomethacin as a drug model , STP Pharma , 5 , 642 . 
62. Gallardo , M. M. , Courraze , G. , Denizor , B. , Treupel , L. , Couvreur , P. , and Puisieux , F. 
( 1993 ), Preparation and purifi cation of isohexylcyanoacrylate nanocapsules , Int. J. 
Pharm. , 100 , 55 – 64 . 
63. Chouinard , F. , Buczkowski , S. , and Lenaerts , V. ( 1994 ), Poly(alkylcyanoacrylate) nanocapsules: 
Physico - chemical characterization and mechanism of formation , Pharm. Res. , 
11 , 869 . 
64. Wohlgemuth , M. , Machtle , W. , and Mayer , C. ( 2000 ), Improved preparation and physical 
studies of polybutylcyanoacrylate nanocapsules , J. Microencapsul. , 17 , 437 . 
65. Lambert , G. , Fattal , E. , Pinto - Alphandary , H. , Gulik , A. , and Couvereur , P. ( 2000 ), Polyisobutylcyanoacrylate 
nanocapsules containing an aqueous core the delivery of oligonucleotides 
, Int. J. Pharm. , 214 , 13 . 
66. Couvreur , P. , Barratt , G. , Fattal , E. , Legrand , P. , and Vauthier , C. ( 2002 ), Nanocapsule 
technology: A review . Crit. Rev. Therap. Drug Carrier Syst. , 19 , 99 – 134 . 
67. Quintanar - Guerrero , D. , Allemann , E. , Doelker , E. , and Fessi , H. ( 1998 ), Preparation 
and characterization of nanocapsules from performed polymers by a new process based 
on emulsifi cation - diffusion technique , Pharm. Res. , 15 , 1056 . 
68. Ravi , K. M. N. ( 2000 ), Nano and microparticles as controlled drug delivery devices , 
J. Pharm. Sci. , 3 , 234 – 258 . 
69. Torchilin , V. P. ( 2005 ), Recent advances with liposomes as pharmaceutical carriers , 
Nature Rev.: Drug Discovery , 4 , 145 – 159 . 
70. Leserman, L. (2004), Liposomes as protein carriers in immunology , J. Liposome Res. , 14 , 
175 – 189 . 
71. Uchegbu , I. F. , and Vyas , S. P. ( 1998 ), Non - ionic surfactant based vesicles (niosomes) in 
drug delivery , Int. J. Pharm. , 172 , 33 – 70 . 
72. Uchegbu , I. F. ( 2000 ), Synthetic surfactant vesicles: Niosomes and other non - 
phospholipid vesicular systems , in Drug Targeting and Delivery , Vol. 11, Harwood 
Academic , Amsterdam . 
73. Bodor , N. , and Loftsson , T. ( 1987 ), Novel chemical approaches for sustained drug delivery 
, in Controlled Drug Delivery: Fundamentals and Applications , Marcel Dekker , New 
York , pp. 337 – 369 . 
74. Sugibayashi , K. , and Morimoto , Y. ( 1994 ), Polymers for transdermal drug delivery 
systems , J. Controlled Release , 29 , 177 – 185 . 
75. Chien , Y. W. ( 1987 ), Transdermal therapeutic systems , in Controlled Drug Delivery: 
Fundamentals and Applications , Marcel Dekker , New York , pp. 524 – 549 . 
76. Chien , Y. W. ( 1982 ), Ocular controlled release drug administration , in Novel Drug Delivery 
Systems , Marcel Dekker , New York , pp. 13 – 48 . 
REFERENCES 389

390 CONTROLLED-RELEASE DOSAGE FORMS 
77. Gelatt , K. N. , Gum , G. G. , williams , L. W. , and Peiffer , R. L. ( 1979 ), Evaluation of a soluble 
sustained - release ophthalmic delivery unit in the dog , Am. J. Vet. Res. , 40 , 702 – 704 . 
78. Chien , D. S. , and Schoenwald , R. D. ( 1990 ), Ocular pharmacokinetics and pharmacodynamics 
of phenylephrine and phenylephrine oxazolidine in rabbit eyes , Pharm. Res. , 7 , 
476 – 483 . 
79. Vyas , S. P. , Ramchandraiah , S. , Jain , C. P. , and Jain , S. K. ( 1992 ), Polymeric pseudolatices 
bearing pilocarpine for controlled ocular delivery , J. Microencapsul. , 9 , 347 – 355 . 
80. Lyren a s , S. , Clason , I. , and Ulmsten , U. ( 2001 ), In vivo controlled release of PGE2 from 
a vaginal insert (0.8 mm, 10 mg) during induction of labor , Br. J. Obstet. Gynaecol. , 108 , 
169 – 178 . 
81. Sherwood , J. K. , Zeitlin , L. , Whaley , K. J. , Richard , A. C. , and Saltzman , M. ( 1996 ), Controlled 
release of antibodies for long - term topical passive immunoprotection of female 
mice against genital herpes , Nat. Biotechnol. , 14 , 468 – 471 . 
82. Nilsson , C. , Lachteenmaki , P. , and Luukkainen , T. ( 1980 ), Patterns of ovulation and 
bleeding with a low levonregesterol - releasing device , Contraception , 21 , 225 – 233 . 
83. van Laarhoven , J. , Kruft , M. , and Vromans , H. ( 2002 ), In vitro release properties of 
etonogestrel and ethynylestradiol from a contraceptive vaginal ring , Int. J. Pharm. , 232 , 
163 – 173 . 
84. Rathbone , M. J. , Macmillan , K. L. , J o Chle , W. , Boland , M. , and Inskeep , E. K. ( 1998 ), 
Controlled release products for the control of the estrous cycle in cattle, sheep, goats, 
deer, pigs, and horses , Crit. Rev. Ther. Drug Carrier Syst. , 15 , 285 – 380 . 
85. Strix , J. ( 1999 ), German Patent Application DE 98 - 19809243 . 
86. Duncan , R. , and Seymour , L. ( 1989 ), Controlled Release Technolgies, Elsevier Advanced 
Technology, Amsterdam, 11. 
87. Lee , E. S. , Kim , S. W. , Kim , S. H. , Cardinal , J. R. , and Jacobs , H. ( 1980 ), Drug release from 
hydrogel devices with rate - controlling barriers , J. Membr. Sci. , 7 , 293 – 303 . 
88. El - Nokaly , M. A. , Piatt , D. M. , and Charpentier , B. A. ( 1993 ), Polymeric Delivery Systems: 
Properties and Applications , ACS Symposium Series , Washington, DC . 
89. Hui , H. W. , Lee , V. H. L. , and Robinson , J. R. ( 1987 ), Design and fabrication of oral 
controlled release drug delivery systems , in Controlled Drug Delivery: Fundamentals and 
Applications , Marcel Dekker , New York , pp. 373 – 421 . 
90. Sangalli , M. E. , Maroni , A. , Zema , L. , Busseli , C. , Giordano , F. , and Gazzaniga , A. ( 2001 ), 
In vitro and in vivo evaluation of oral system for time and/or site specifi c drug delivery , 
J. Controlled Release , 73 , 103 – 110 . 
91. Sangalli , M. E. , Maroni , A. , Foppoli , A. , Zema , L. , Giordano , F. , and Gazzaniga , A. ( 2004 ), 
Different HPMC viscosity grades as coating agents for an oral time and/or site - 
controlled delivery systems: A study on process parameters and in vitro performances , 
Eur. J. Pharm. Sci. , 22 , 469 – 476 . 
92. Pozzi , F. , Furlani , P. , Gazzaniga , A. , Davis , S. S. , and Wilding , I. R. ( 1994 ), The TIME 
CLOCK ® system: A new oral dosage form for fast and complete of drug after a predetermined 
lag time , J. Controlled Release , 31 , 99 – 108 . 
93. Medlicott , N. J. , and Tucker , I. J. ( 1999 ), Pulsatile release from subcutaneous implants , 
Adv. Drug Deliv. Rev. , 38 , 139 – 149 . 
94. Bussemer , T. , Peppas , N. A. , and Bodmeier , R. ( 2003 ), Evaluation of the swelling, hydration 
and rupturing properties of the swelling layer of a rupturable pulsatile drug delivery 
system , Eur. J. Pharm. Biopharm. , 56 , 261 – 270 . 
95. Bussemer , T. , Peppas , N. A. , and Bodmeier , R. ( 2003 ), Time - dependent mechanical properties 
of polymers coating used in rupturable pulsatile release dosage forms , Drug Dev. 
Ind. Pharm. , 29 , 623 – 630 . 

96. Sungthongjeen , S. , Puttipipatkhachorn , S. , Paeratakul , O. , Dashevsky , A. , and Bodmeier , 
R. ( 2004 ), Development of pulsatile release tablets with swelling and rupturable layers , 
J. Controlled Release , 95 , 147 – 159 . 
97. Niwa , K. , Takaya , T. , Morimoto , T. , and Takada , K. ( 1995 ), Preparation and evaluation 
of a time - controlled release capsule made of ethylcellulose for colon delivery of drugs , 
J. Drug Target. , 3 , 83 – 89 . 
98. Krogel , I. , and Bodmeier , R. ( 1997 ), Pulsatile drug release from an insoluble capsule 
body controlled by an erodible plug , Pharm. Res. , 15 , 474 – 481 . 
99. Krogel , I. , and Bodmeier , R. ( 1999 ), Evaluation of an enzyme - containing capsular shaped 
pulsatile drug delivery system , Pharm. Res. , 16 , 1424 – 1429 . 
100. Gohel , M. C. , and Sumitra , M. ( 2002 ), Modulation of active pharmaceutical material 
release from a novel “ tablet in capsule system ” containing an effervescent blend , 
J. Controlled Release , 79 , 157 – 164 . 
101. Linkwitz , A. , Magruder , J. A. , and Merrill , S. ( 1994 ), Osmotically driven delivery device 
with expandable orifi ce for pulsatile delivery effect, U.S. Patent 5,318,558 . 
102. Qui , L. Y. , and Zhu , K. J. ( 2001 ), Design of core - shelled polymer cylinder for potential 
programmable drug delivery , Int. J. Pharm. , 219 , 151 – 160 . 
103. Jiang , H. L. , and Zhu , K. J. ( 1999 ), Preparation, characterization and degradation characteristics 
of polyanhydrides containing polyethlene glycol , Poly. Int. , 48 , 47 – 52 . 
104. Jiang , H. L. , and Zhu , K. J. ( 2000 ), Pulsatile protein release from a laminated device 
comprising of polyanhydride and pH - sensitive complexes , Int. J. Pharm. , 194 , 51 – 60 . 
105. Anal , A. K. ( 2007 ), Stimuli - induced pulsatile or triggered release of bioactive compounds, 
Recent Patents Endocrine , Metabolic Immune Drug Discovery , 1 , 83 – 90 . 
106. Rubinstein , A. ( 2005 ), Colonic drug delivery , Drug Discovery Today: Technol. , 2 , 33 – 37 . 
107. Deshpande , A. A. , Rhodes , C. T. , Shah , N. H. , and Mallick , A. W. ( 1996 ), Controlled - 
release drug delivery systems for prolonged gastric residence: An overview , Drug Dev. 
Ind. Pharm. , 22 , 531 – 539 . 
108. Rubinstein , A. ( 1995 ), Approaches and opportunities in colon - specifi c drug delivery . 
Curr. Rev. Ther. Drug Carrier Syst. , 12 , 101 – 149 . 
109. Basit , A. , and Bloor , J. ( 2003 ), Perspectives on colonic drug delivery , Pharmatech , 
185 – 190 . 
110. Vandamme , T. F. , Lenoury , A. , Charrueau , C. , and Chaumeil , J. - C. ( 2002 ), The use of 
polysaccharides to target drugs to the colon . Carbohydr. Poly. , 48 , 219 – 231 . 
111. Sinha , V. R. , and Kumria , R. ( 2001 ), Polysaccharides in colon - specifi c drug delivery , Int. 
J. Pharm. , 224 , 19 – 38 . 
112. Lee , S. , Lim , C. B. , Lee , S. , and Pai , C. M. ( 1999 ), Colon selective drug delivery composition, 
WO 99/01115 . 
113. Watanabe , S. , Kawai , H. , Katsuma , M. , and Fukul , M. ( 2002 ), Colon - specifi c drug release 
system, U.S. Patent 6,368,629 . 
114. Takemura , S. , Watanabe , S. , Katsuma , M. , and Fukul , M. ( 2002 ), Gastrointestinal transit 
study of a novel colon delivery system (CODES ™ ) using gamma scintigraphy , Proc. Int. 
Symp. Controlled Release Bioactive Mater. , 27 , 445 – 446 . 
115. Li , J. , Yang , L. , Ferguson , S. M. , Hudson , T. J. , Watanabe , S. , Katsuma , M. , and Fix , J. A. 
( 2002 ), In vitro evaluation of dissolution behaviour for a colon - specifc drug delivery 
system (CODESTM) in multi - pH media using USP apparatus II and III, AAPS Pharm. 
Sci. Technol. , 3, article 33, available: www.aapspharmascitech.org . 
116. Bae , Y. H. , Okano , T. , and Kim , S. W. ( 1991 ), “ On - off ” thermocontrol of solute transport. 
I. Temperature dependence of swelling of N - isopropylacrylamide networks modifi ed 
with hydrophobic components in water , Pharm. Res. , 8 , 531 – 537 . 
REFERENCES 391

392 CONTROLLED-RELEASE DOSAGE FORMS 
117. Bae , Y. H. , Okano , T. , and Kim , S. W. ( 1991 ), “ On - off ” thermocontrol of solute transport. 
II. Temperature dependence of swelling of N - isopropylacrylamide networks modifi ed 
with hydrophobic components in water , Pharm. Res. , 8 , 624 – 628 . 
118. Okano , T. , Bae , Y. H. , Jacobs , H. , and Kim , S. W. ( 1990 ), Thermally on - off switching 
polymers for drug permeation and release , J. Controlled Release , 11 , 255 – 265 . 
119. Kaneko , Y. , Sakai , K. , Kikuchi , A. , Yoshida , R. , Sakurai , Y. , and Okano , T. ( 1995 ), Infl uence 
of freely mobile grafted chain length on dynamic properties of comb - type grafted 
poly(N - isopropylacrylamide) hydrogels , Macromolecules , 28 , 7717 – 7723 . 
120. Kaneko , Y. , Sakai , K. , Kikuchi , A. , Sakurai , Y. , and Okano , T. ( 1996 ), Fast swelling/ 
deswelling kinetics of comb - type grafted poly( N - isopropylacrylamide hydrogels) . 
Macromol. Chem. , 109 , 41 – 53 . 
121. Bromberg , L. E. , and Ren , E. S. ( 1998 ), Thepertaure - responsive gels and thermo gelling 
polymer matrices for protein and peptide delivery , Adv. Drug Deliv. Rev. , 31 , 197 – 221 . 
122. Sershen, S. R. , Westcott, S. L. , Halas , N. J. , and West, N. J. (2000), Temperature-sensitive 
polymer - nanoshell composites for photothermally modulated drug delivery , J. Biomed. 
Mater. Res. , 51 , 293 – 298 . 
123. Yuk , S. H. , Cho , S. H. , and Lee , H. B. ( 1992 ), Electric current - sensitive drug delivery 
systems using sodium - alginate/polyacrylic acid composites , Pharm. Res. , 9 , 955 – 957 . 
124. Santini , J. T. , Cima , M. J. , and Langer , R. ( 1999 ), A controlled - release microchip , Nature , 
397 , 335 – 338 . 
125. Santini , J. T. , Richards , A. C. , Schiedt , R. , Cima , M. J. , and Langer , R. ( 2000 ), Microchips 
as controlled - drug delivery devices , Chem. Int. Ed. , 39 , 2396 – 2407 . 
126. Barrett , D. H. , Davidson , P. C. , Steed , L. J. , Abel , G. G. , Loman , K. E. , and Saudek , C. D. 
( 1995 ), Evaluation of the psychosocial impact of the minimed variable - rate implanatable 
insulin pump , Med. J. , 88 , 1226 – 1230 . 
127. Ikemoto , S. , and Sharpe , L. G. ( 2001 ), A head - attachable device for injecting anolitre 
volumes of drug solutions into brain sites of freely moving rats , J. Neurosci. Meth. , 110 , 
135 – 140 . 
128. Edelman , E. , Kost , J. , Bobeck , H. , and Langer , R. ( 1985 ), Regulation of drug release 
from polymer matrices by oscillating magnetic fi elds , J. Biomed. Mater. Res. , 19 , 67 . 
129. Babincov a , M. , Sourivong , P. , Chorv a t , D. , and Babinec , P. ( 1999 ), Laser triggered drug 
release from magnetoliposomes , J. Magnetism Magnetic Mater. , 194 , 163 – 166 . 
130. Kim , S. W. , Pai , C. M. , Makino , K. , Seminoff , L. A. , Holmberg , D. L. , Gleeson , J. M. , 
Wilson , D. E. , Mack , E. J. ( 1990 ), Self - regulated glycosylated insulin delivery , J. Controlled 
Release , 11 , 193 – 201 . 
131. Obaidat , A. A. , and Park K. ( 1997 ), Characterization of protein release through glucose - 
sensitive hydrogel membranes , Biomaterials , 18 , 801 – 806 . 
132. Kataoka , K. , Miyazaki , H. , Bunya , M. , Okano , T. , and Sakurai , Y. ( 1998 ), Totally synthetic 
polymer gels responding to external glucose concentration: Their preparation and 
application to on - off regulation of insulin release , J. Am. Chem. Soc. , 120 , 12694 – 
12695 . 
133. Yui , N. , Okano , T. , and Sakurai , Y. ( 1992 ), Infl ammation responsive degradation of crosslinked 
hyaluronic acid gels , J. Controlled Release , 22 , 105 – 116 . 
134. Yui , N. , Nihira , J. , Okano , T. , and Sakurai , Y. ( 1993 ), Regulated release of drug microspheres 
from infl ammation responsive degradable matrices of crosslinked hyaluronic 
acid , J. Controlled Release , 25 , 133 – 143 . 
135. Kikuchi , A. , and Okano , T. ( 2002 ), Pulsatile drug release control using hydrogels , Adv. 
Drug Deliv. Rev. , 54 , 53 – 77 . 

393 
5.2 
PROGRESS IN DESIGN OF 
BIODEGRADABLE POLYMER - BASED 
MICROSPHERES FOR PARENTERAL 
CONTROLLED DELIVERY OF 
THERAPEUTIC PEPTIDE/PROTEIN 
Shunmugaperumal Tamilvanan * 
University of Antwerp, Antwerp, Belgium 
Contents 
5.2.1 Introduction 
5.2.2 Peptide/Protein - Loaded Microsphere Production Methods 
5.2.2.1 Phase Separation (A Traditional Technique) 
5.2.2.2 Double Emulsion (A Hydrous Technique) 
5.2.2.3 Spray Drying (An Anhydrous Technique) 
5.2.2.4 New Trends in Production Methods 
5.2.3 Analytical Characterization of Peptide/Protein - loaded Microspheres 
5.2.4 Immune System Interaction with Injectable Microspheres 
5.2.5 Excipient Inclusion: Injectable Peptide/Protein - Loaded Microspheres 
5.2.5.1 Solubility - and Stability - Increasing Excipients 
5.2.5.2 Preservation - Imparting Excipients 
5.2.6 Peptide/Protein Encapsulated into Biodegradable Microspheres: Case Study 
5.2.6.1 Vaccines 
5.2.6.2 Proteins 
5.2.7 Conclusion 
References 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc. 
* Current address: Department of Pharmaceutics, Arulmigu Kalasalingam College of Pharmacy, Anand 
Nagar, Krishnankoil, India

394 BIODEGRADABLE POLYMER-BASED MICROSPHERES 
5.2.1 INTRODUCTION 
At the cellular level, deoxyribonucleic and ribonucleic acids (DNA and RNA, respectively) 
serve as an endogenious vehicle not only to store genetic information but 
also to transfer genetic information from one generation to their offsprings of all 
known living organisms. In addition, utilizing the rule of complementary base 
pairing, the DNA undergoes replication and transcription processes to produce 
respectively a new double - stranded DNA molecule and a complementary single - 
stranded RNA molecule. Following the translation process, peptide and protein are 
synthesized/constructed in ribosomal subunits through peptidic linkages between 
available 20 amino acids. The peptide and protein thus constructed perform a wide 
variety of functions and each cell contains several thousands of different proteins. 
Peptide - and protein - mediated, important physiological and biological processes of 
the human body include ligands/hormones for signaling, enzymes for biotransformation 
reactions, receptors for pharmacological response elucidation, antibodies in 
immune system interactions, transcription, and translation. Hence these molecules 
play a vital role to ensure proper development and functioning of entire organs of 
the human body. 
Webster ’ s New World Dictionary defi nes a drug as “ any substances used as a medicine 
or as an ingredient in a medicine. ” Indeed, peptides and recombinant proteins 
are highly potent, relatively macromolecular and promising therapeutic agents that 
emerged out from the signifi cant development of biotechnic and biogenetic engineering 
technologies. Peptide and protein therapeutics include semisynthetic vaccines, 
monoclonal antibodies, growth factors, cytokines, soluble receptors, hormones, and 
enzymes. The advent of recombinant DNA technology allowed the possibility of the 
commercial production of proteins for pharmaceutical applications from the early 
1980s and, in fact, manufacture of therapeutic proteins represented the fi rst true 
industrial application of this technology [1] . During the 1980s the term biopharmaceutical 
became synonymous with therapeutic protein produced by recombinant DNA 
technology (or, in the case of a small number of therapeutic monoclonal antibodies, 
by hybridoma technology ). Clinical evaluation of nucleic acid – based drugs used for 
the purposes of gene therapy and antisense technology commenced in the 1990s, and 
today the term biopharmaceutical also encompasses such (as - yet - experimental) 
drugs [2] . The fi rst such recombinant therapeutic protein (insulin) was approved for 
general medical use only 24 years ago. Today there are in excess of 100 such products 
approved in some world regions at least, with 88 having re ceived approval within 
the European Union (EU). This represents 36% of all new drug approvals since the 
introduction of the new centralized European drug approval system in 1995 [3] . Over 
the coming decade, therefore, in the region of a dozen new therapeutic proteins 
should, on average, gain regulatory approval each year. While EU fi gures are diffi cult 
to locate, the American Association of Pharmaceutical Researchers and Manufacturers 
(PhRMA) estimates that there are currently some 371 biotechnology medicines 
in development [4] . Out of these 371 biotechnology medicines, as estimated by 
PhRMA, more than 300 are protein based, with recombinant vaccines and monoclonal/
engineered antibodies representing the two most promising categories. Incidentally, 
all 88 biopharmaceu tical products currently approved within the EU are 
protein based. Of the proteins thus far approved, hormones and cytokines represent 
the largest product categories (23 and 18 products, respectively). Hormones approved 
include several recombinant insulins, displaying both native and modifi ed amino acid 

sequences. In addition, several recombinant gonadotrophins [follicle - stimulating 
hormone (FSH), luteinizing hormone (LH), and human chorionic gonadotrophins 
(hCG)] have been approved for the treatment of various forms of subfertility/infertility. 
Cytokines approved include a range of recombinant hematopoietic factors, 
including multiple erythropoietin - based products used for the treatment of anemia 
as well as a colony - stimulating factor aimed at treating neutropenia. Additional 
approved cytokines include a range of interferon - based products, mainly used to 
treat cancer and various viral infections, most notably hepatitis B and C, and a 
recombinant tumor necrosis factor (TNF) used as an adjuvant therapy in the treatment 
of some soft tissue cancers. Blood - related approved therapeutic proteins 
include a range of recombinant blood coagulation factors used to treat hemophilia, 
recombinant thrombolytics, and recombinant anticoagulants. Additional product 
categories include a range of subunit vaccines containing at least one recombinant 
component [mainly hepatitis B surface antigens (HBsAg)] and a variety of monoclonal 
antibody – based products indicated for the treatment/detection of various 
cancers or the prevention of organ transplant rejection. In summary, ailments that 
can be treated more effectively by this new class of therapeutic agents include cancers, 
autoimmune disease, memory impairment, mental disorders, hypertension, and 
certain cardiovascular and metabolic diseases [5, 6] . 
Poor absorption and easy degradation by endogenous proteolytic enzymes 
present in eye tissues, nasal mucosa, and gastro intestinal tract and low transdermal 
bioavailabilities due to relatively large size make the peptide/protein molecules to 
be administered only through parenteral routes either by multiple injections or 
infusion therapy in order to achieve desired therapeutic plasma levels for prolonged 
periods of time. Nevertheless, because of remarkably short half - lives within the in 
vivo arena, the therapeutic usuage of most of the peptide/protein is practically possible 
only through daily multiple injections under close medical supervision. Hence, 
the commercial success of peptides/proteins as therapeutic agents depends mainly 
on development of novel drug delivery systems which could potentially reduce 
the injection frequencies and thus eliminate the accompanying serious problem of 
patient compliance. 
Among the several technologies that have been suggested for reducing injection 
frequencies of therapeutic peptide/protein, microspheres prepared from biodegradable 
polymers are widely recognized for controlled drug delivery following parenteral 
administration. Polyester polymers such as poly(lactic acid) (PLA), poly(glycolic 
acid) (PGA), and their copolymer poly(lactic acid - co - glycolic acid) (PLGA) are 
used routinely for the preparation of injectable microspheres after taking into consideration 
their well - known biocompatibility, controlled biodegradability, absorbability, 
and no toxicity of degradation products [7] . Furthermore, the PLGA types 
and related poly(hydroxyalkanonates) have a long history of medical and pharmaceutical 
use in fi elds as diverse as sutures, bone fi xatives, artifi cial skins and cartilages, 
dental materials, materials for bone generation, drug delivery, and many 
others, as reviewed by Ueda and Tabata [8] . In conjunction with a long safety record 
of PLGA polymers, at least 12 different peptide/protein - loaded PLGA microsphere 
products are available in the market from nine different companies worldwide 
for the treatment of some life - threatening diseases (Table 1 ). In recent years, 
poly(. - caprolactone) (PCL) has been investigated as an alternative to PLGA to 
make microspheres [9, 10] . A glimpse of ongoing research activities utilizing biodegradable 
polymer - based microspheres for various peptide/protein is shown in 
INTRODUCTION 395

396 BIODEGRADABLE POLYMER-BASED MICROSPHERES 
Table 2 (incorporating refs. 12 – 35). However, overcoming the propensity for peptides/
proteins to undergo degradation processes during incorporation into the biodegradable 
microspheres or after injection into the body awaiting release is one of 
the key hurdles in bringing microencapsulated systems for these drugs to market. 
This partially explains the limited and only a countable number of formulations 
available on the market. Furthermore, irrespective of the various microencapsulation 
techniques adopted to prepare peptide/protein - loaded microspheres, several 
transfer - required processes such as fi ltration, centrifugation, and vacuum or freeze 
drying are necessary to obtain a fi nal product, and these processes might be obstacles 
when scaling up the manufacturing technique to produce suffi cient quantities 
of sterile material for clinical trial and, ultimately, commercialization [11] . 
This chapter encompasses investigations made progressively on the design of 
injectable peptide/protein - loaded PLGA microspheres. It covers an update on the 
state of art of the manufacturing of peptide/protein - loaded microspheres through 
both conventional and newer microencapsulation techniques, different analytical 
methods used for microsphere characterization, immune system interaction with 
microspheres following parenteral administration, and potential application of 
microspheres having therapeutic peptides/proteins. Special emphasis is given particularly 
on various instability problems and investigated mechanistic ways to 
obviate the possible instability problems of peptide/protein drug during microsphere 
preparation as well as its release from the microspheres. It should be added 
that although the chapter focuses mainly on PLGA microspheres, many of the 
destabilization mechanisms and stabilization approaches described herein can be 
valid to some extent for other polymeric delivery systems, too. 
TABLE 1 Currently Marketed Preparations (Injectable Microspheres) Containing 
Peptide/Protein Molecules 
Commercial 
Name API Polymer Company Indication 
Lupron Depot Leuprolide PLGA or PLA TAP Prostate cancer, 
endometriosis 
Enantone Depot Leuprolide PLGA or PLA Takeda Prostate cancer, 
endometriosis 
Trenantone Leuprolide PLGA or PLA Takeda Prostate cancer, 
endometriosis 
Enantone Gyn Leuprolide PLGA or PLA Takeda Prostate cancer, 
endometriosis 
Sandostatin LAR Octreotide PLGA - glucose Novartis Acromegaly 
Nutropin Somatropin PLGA Genentech Growth 
defi ciencies 
Trelstar Depot Triptorelin PLGA Pfi zer Prostate cancer 
Decapeptyl SR Triptorelin PLGA or PLA Ipsen - Beaufour Prostate cancer 
Decapeptyl Triptorelin PLGA Ferring Prostate cancer 
Suprecur MP Buserelin PLGA Aventis Endometriosis 
Somatuline LA Lanreotide PLGA Ipsen - Beaufour Acromegaly 
Parlodel LAR Bromocriptine PLGA - Glu Novartis Parkinsonism 
Abbreviations: PLA: polylactide; PLGA: poly(lactide - co - glycolide); API: active pharmaceutical ingredient; 
PLGA - Glu: poly( d , l - lactide - co - glycolide - d - glucose). 

TABLE 2 Injectable Peptide/Proteins/Vaccines Encapsulated in Biodegradable 
Microspheres 
Peptides, Protein, Vaccine Technique Polymer Reference 
Vaccine 
SPf 66 malaria vaccine Double emulsion PLGA 28 
Multivalent vaccines of 
Haemophilus infl uenzae 
type b (Hib), diphtheria 
toxoid (DT), tetanus 
toxoid (TT), pertussis 
toxin (PT) 
Spray drying PLGA 29 
Rotavirus Double emulsion PLG 30 
Polypeptides and Proteins 
Insulin Double emulsion PLA polyethylene 
glycol (PEG) 
12 
Recombinant human 
epidermal growth factor 
(rhEGF) 
Double emulsion PLA 13 
Ribozyme Double emulsion PLA, PLGA 14 
Vapreotide (somatostatin 
analogue) 
Spray drying PLGA 15 
Insulinlike growth factor - 1 
(IGF - 1) 
Double emulsion PLGA - PEG 16 
Ornitide acetate 
leuteinizing hormone 
releasing hormone 
[(LHRH) antagonist] 
Dispersion/solvent 
extraction/evaporation 
PLA, PLGA 17 
Vascular endothelial 
growth factor (VEGF) 
Single emulsion PLGA/PEG 18 
Human chorionic 
gonadotropin (hCG) 
Double emulsion PLA, PLGA 19 
Calcitonin Double emulsion PLGA 20 
FITC - bovine serum 
albumin (BSA) 
Double emulsion Poly( . - caprolactone) 9 
Levonorgestrel and 
ethinylestradiol 
Double emulsion Poly( . - caprolactone) 10 
Recombinant human bone 
morphogenetic protein 
Double emulsion PLGA 21 – 24 
Transforming growth factor 
beta 
Double emulsion PLGA or 
PLGA - PEG 
25 – 27 
Recombinant human 
erythropoietin (rhEPO) 
Modifi ed double 
emulsion 
LPLG - PEO - LPLG 31 
Protein - C Double emulsion PLA 32 
Ovalbumin Double emulsion PLGA 33 
Human serum albumin Double emulsion PLA 34 
Bovine serum albumin Nonaqueous oil - in - oil 
(o/o) emulsion 
PLG 35 
Abbreviations: FITC: fl uroscein isothiocyanate; PLG: poly(lactide - co - glycolide); LPLG - PEO - LPLG: 
copoly(l - lactic - co - glycolic acid - b - oxyethylene - b - l - lactic - co - glycolic adic); PEO: polyethylenenoxide. 
INTRODUCTION 397

398 BIODEGRADABLE POLYMER-BASED MICROSPHERES 
5.2.2 PEPTIDE/PROTEIN - LOADED MICROSPHERE 
PRODUCTION METHODS 
The development of delivery systems for therapeutic peptides/proteins depends on 
biophysical, biochemical, and physiological characteristics of these molecules, including 
molecular size, biological half - life, immunogenicity, conformational stability, 
dose requirement, site and rate of administration, pharmacokinetics, and pharmacodynamics 
[36] . Unlike conventional drug molecules, the unique conformational 
structure of peptidic/proteinic therapeutic agents poses a great challenge right from 
the beginning of the selection of suitable microencapsulation techniques to make 
microspheres. Table 3 lists the considerations to be taken before choosing a particular 
encapsulation technique. Apart from the traditional phase separation technique, 
other techniques suitable for peptide/protein - loaded microsphere production can 
be divided into two main categories: during microsphere preparation, those involved 
in utilizing a hydrous environment such as emulsion - based methods and those based 
on an anhydrous environment such as spray freeze drying, spray drying, freeze 
drying, grinding, jet milling, liquid - phase antisolvent precipitation, and supercritical 
CO2 - based methods [37 – 40] . In the following sections, the various production techniques 
to make injectable peptide/protein - loaded microspheres are briefl y introduced; 
however, a detailed discussion is beyond the scope of this chapter. 
5.2.2.1 Phase Separation (A Traditional Technique) 
Polymer phase separation or coacervation is an excellent technique for the encapsulation 
of water - soluble drugs including peptide/protein into a fi nal microsphere 
product [41] . The peptide/protein molecule is dispersed in solid form into solution 
containing dichloromethane and PLGA. Silicone oil is added to this dispersion at 
a defi ned rate, reducing the solubility of polymer in its solvent. The polymer - rich 
liquid phase (coacervate) encapsulates the dispersed peptide/protein molecules and 
embryonic microspheres are subjected to hardening and washing using heptane. 
The process is quite sensitive to polymer properties, and residual solvent is also an 
important issue. Decapeptyl [triptorelin, a luteinizing hormone releasing hormone 
(LHRH) analogue] [42] and Somatuline LA (lanreotide, a somatostatin analogue) 
[43] are microsphere commercial products developed by this technique (Table 1 ). 
TABLE 3 Factors in Selection of Microencapsulation 
Method to Prepare Peptide/Protein-Loaded Microspheres 
Optimal peptide loading 
High microsphere yield 
Batch content uniformity 
Interbatch reproducibility 
Peptide stability during preparation and release 
Size uniformity 
Adjustable release profi le 
Low burst release 
Flowability of fi nal product 
Residual solvent and polymer monomer control 
Sterilization (both aseptic and terminal) 

5.2.2.2 Double Emulsion (A Hydrous Technique) 
Oil - in - water (o/w) and water - in - oil - water (w/o/w) are the two hydrous techniques 
representing respectively the single - and double - emulsion formation during microsphere 
preparation. However, the w/o/w technique is most commonly employed 
[44] . In this process, peptides/proteins in aqueous solution are emulsifi ed with 
nonmiscible organic solution of polymer to form a w/o emulsion. Dichloromethane 
serves as organic solvent and the o/w primary emulsion is formed using either high - 
speed homogenization or ultrasonication. This primary emulsion is then rapidly 
transferred to an excess of aqueous medium containing a stabilizer, usually polyvinyl 
alcohol. Again homogenization or intensive stirring is necessary to initially form a 
double emulsion of w/o/w. Subsequent removal (by evaporation) of organic solvent 
by heat, vacuum, or both results in phase separation of polymer and core to produce 
microspheres. Instead of solvent evaporation, solvent extraction with a large quantity 
of water with or without a stabilizer can also be undertaken to yield mi crospheres 
containing peptide/protein. Although the w/o/w microencapsulation technique 
seems to be conceptually simple to carry out, the particle formation process is quite 
complicated, and a host of process parameters infl uence the properties of peptide/ 
protein - loaded PLGA microspheres [45] . In spite of that, different peptides and 
proteins such as bovine serum albumin (BSA) or ovalbumin (OVA), insulin, recombinant 
human insulinlike growth factor - 1 (rhIGF - 1), recombinant human epidermal 
growth factor (rhEGF), human chorionic gonadotropin (hCG), protein C, recombinant 
human bone morphogenetic protein (rhBMP), and calcitonin, along with antigens 
and other therapeutically relevant proteins such as recombinant human 
erythropoietin (rhEPO), have been successfully encapsulated (see Table 2 ) by the 
w/o/w double - emulsion technique. Lupron Depot/Enantone Depot/Trenantone/ 
Enantone Gyn (all having leuprolide acetate, a LHRH analogue) are very popular 
commercial microsphere products produced by this technique [46, 47] , available 
both in the EU and United States, for the treatment of either prostate cancer of 
man or infertility (endometriosis) of women (Table 1 ). 
5.2.2.3 Spray Drying (An Anhydrous Technique) 
Spray drying is a rapid, convenient technique which can be conducted under aseptic 
conditions. First, a polymer — prevalently PLGA is applied — is dissolved in a volatile 
organic solvent such as dichloromethane or acetone. The protein is suspended as a 
solid or emulsifi ed as aqueous solution in this organic solution by homogenization. 
After that, the resulting dispersion is atomized through a (heated) nozzle into a 
heated airfl ow. The organic solvent evaporates, thereby forming microspheres with 
dimensions of typically 1 – 100 . m. The microspheres are collected in a cyclone separator. 
For the complete removal of the organic solvent, a vacuum drying or lyophilization 
step can follow downstream. 
The internal structure of the resulting polymeric microspheres depends on the 
solubility of the peptide/protein in the polymer before being spray dried leading to 
the formation of reservoir - or matrix - type products (see Figure 1 ). When the initial 
dispersion is solution, the fi nal product obtained following spray drying is matrix or 
monolithic type, that is, polymer particles with a dissolved or dispersed nature 
of the active ingredient (defi ned as microspheres). Conversely, when the initial 
PEPTIDE/PROTEIN-LOADED MICROSPHERE PRODUCTION METHODS 399

400 BIODEGRADABLE POLYMER-BASED MICROSPHERES 
dispersion is in suspension, the product obtained is reservoir type, that is, a distinct 
polymeric envelope/shell encirculating a liquid core of dissolved active ingredient 
(defi ned as microcapsules). Recombinant human erythropoietin [48] and bromocriptine 
mesylate, Parlodel Depot [49] , are examples of microspheres (matrix type) 
obtained by the spray drying technique. 
5.2.2.4 New Trends in Production Methods 
Several issues such as reducing cost, reducing scale - up diffi culties, improving protein 
stability, allowing for terminal sterilization, and eliminating the need for organic 
solvents during addition of the peptide/protein motivate the development of new 
methods to manufacture microspheres. Moreover conventional microencapsulation 
methods involve relatively harsh conditions that are not generally tolerated by 
peptide/protein molecules without stabilization. Therefore, new and improved processes 
shielding the peptide/protein from deleterious conditions have been proposed 
and evaluated. 
Modifi ed Conventional Methods The w/o/w solvent evaporation or extraction is 
probably one of the most widely used methods for peptide and protein microencapsulation 
[44] , despite its many drawbacks. Improvements and alternatives have 
therefore been proposed such as oil in water (o/w), * o/w (the asterisk including 
cosolvent) and oil in oil (o/o) [50] . 
Utilising a modifi ed w/o/w method, the rhIGF - 1 was encapsulated into PLGA 
microspheres after increasing the pH of the protein solution from 4.5 to 5.5 – 6.0, 
where rhIGF - 1 formed a viscous gel [51] . High entrapment effi ciency of fully bioactive 
protein was achieved, and 92 – 100% of pure, monomeric, and bioactive rhIGF - 1 
was released in vitro over 21 days. The lowering of the rhIGF - 1 solubility at pH 
5.5 – 6.0 probably restricted its conformational fl exibility and changes upon exposure 
to the polymer solvent. Without pH adjustment, approximately 10 – 32% of rhIGF - 1 
was lost upon solvent exposure, due to degradation and aggregation. Elsewhere, a 
w/o 1 /o 2 system was investigated for encapsulating different proteins and peptides, 
with the o 1 and o 2 phases consisting of acetonitrile/dichloromethane and liquid 
paraffi n/Span 80, respectively [52] . The acetonitrile mediated the partial mixing of 
the w and o 1 phases and subsequent protein/peptide precipitation, which was a 
FIGURE 1 Polymeric delivery systems: (a) reservoir systems (microcapsules); (b) matrix 
systems (microspheres). 
Drug 
Drug 
(b) (a) 
Polymer 
Polymer

prerequisite for microencapsulation. The proteins BSA, tetanus toxoid (TT), and 
lysozyme did precipitate at low acetonitrile concentration, resulting in effi cient 
microencapsulation (more than 90%), while a decapeptide and a linear gelatine did 
not precipitate so rapidly, resulting in poor entrapment. TT and lysozyme released 
during the burst phase (15%) maintained their bioactivity, although lack of further 
release suggested aggregation within the microspheres. 
Another approach consisted of dispersing the protein antigen in a mineral oil 
before encapsulation into PLGA microspheres by a o 1 /o 2 /w method [53] . The mineral 
oil (o 1 ) was intended as a barrier to protect the antigen during emulsifi cation with 
the polymer solution and from exposure to moisture during release. Over 92% 
of enzyme - linked imunosorbert assay (ELISA) reactive TT was released from the 
reservoir - type microspheres in a pulsatile pattern, proceeding with an initial burst 
and followed by a second release pulse between 14 and 35 or 35 and 63 days, depending 
on the polymer type used. The latter stage of release was ascribed to TT diffusion 
through the oily phase, once an appreciable loss of polymer mass had occurred. The 
authors claimed the mineral oil was the key to protecting the solid antigen during 
polymer erosion, where acidic degradants and moisture would otherwise have led 
to antigen inactivation. 
To improve solvent extraction, a novel method using a static micromixer was 
presented where a w 1 /o dispersion (aqueous BSA in organic PLGA solution) is fed 
into an array of microchannels and the extraction fl uid (w 2 ) into a second array of 
interdigitated channels [54] . The two fl uids, transported separately through the channels, 
are discharged through an outlet slit where alternating fl uid lamellae are 
formed with the w 1 /o fl uid lamella disintegrating into microdroplets, which harden 
quickly to form microspheres. This process offers easy scale - up, methodological 
robustness, continuous production, and a simple setup, making it ideally suited 
for aseptic production, a strongly needed feature for microsphere vaccine 
formulations. 
ProLease Technology (Cryogenic Spray Drying) A variation of the conventional 
spray drying method is a cryogenic method which will described below. A novel 
low - temperature spraying technique (called ProLease technology) for preparing 
PLA and PLGA microspheres has been reported by Khan et al. [55] and the group 
at Alkermes [56, 57] . The method relies on the use of stabilizing and release controlling 
agents, low processing temperature, and nonaqueous microencapsulation. Typically, 
a protein powder is micronized, possibly with a stabilizer, by spray freeze 
drying and then suspended in an organic polymer solution. The suspension is atomized 
into a vessel containing liquid N 2 underlaid by frozen ethanol (extraction 
solvent). The atomized droplets freeze in the liquid N 2 and deposit on the surface 
of the frozen ethanol. As liquid N 2 evaporates, the frozen ethanol liquefi es ( Tm 
approximately . 110 ° C) so that the frozen polymeric droplets will transfer into the 
ethanol where the polymer solvent is extracted, yielding solid microspheres [58, 59] . 
To date, the ProLease system has been effectively applied to the encapsulation of 
zinc - complexed human growth hormone in PLGA microspheres, resulting in a one - 
month effect after one single injection [37, 57, 60] . As a reference, the recombinant 
human growth hormone (rhGH) was unstable in contact with ethyl acetate or 
dichloromethane [61] . The only protein - containing PLGA microspheres, Nutropin 
Depot, is produced by this novel technique. However, this product containing rhGH 
PEPTIDE/PROTEIN-LOADED MICROSPHERE PRODUCTION METHODS 401

402 BIODEGRADABLE POLYMER-BASED MICROSPHERES 
marketed initially in the United States in 1999 was pulled from the market voluntarily 
by the manufacturer in June 2004 because of high costs of production and 
commercialization ( http://www.gene.com/gene/news/press-releases/ , accessed May 
25, 2006). 
ProLease technology was also used for encapsulating recombinant human vascular 
endothelial growth factor (rhVEGF) and rhIGF - 1 [62, 63] . Both proteins were 
stabilized in aqueous solution, prior to spray freeze drying, and encapsulated (9 – 
20% w/w) into PLGA microspheres. The microspheres also contained ZnCO 3 (3 – 
6% w/w) as release modifi er. The resistance of rhIGF - 1 to aggregation and oxidation, 
determined from in vitro release studies, hardly changed. Protein, released in 
an almost pulsatile fashion over 21 days, was composed of predominantly monomeric 
rhIGF - 1 with only minor amounts ( . 6%) of degradants forming toward day 21. 
Similarly, the integrity of rhVEGF dimer released over 21 days was good and its 
bioactivity remained largely unaffected, regardless of the extent of aggregation and 
degradation. In view of these studies, ProLease technology appears to have potential 
for sustaining antigen stability and release from microspheres. 
Techniques Using Supercritical Fluids Generally, the application of supercritical 
(SC) fl uids for the encapsulation of peptides and proteins has been fueled by the 
recognition that the established methods implicate some drawbacks. The application 
of supercritical fl uids, especially of supercritical carbon dioxide (CO 2 ), can minimize 
or even eliminate the use of organic solvents and renders work at moderate temperatures 
possible [64] . The term supercritical defi nes the area above the critical 
point, which specifi es the fi nal point of the liquid – gas phase transition curve. Beyond 
that critical point, isobar/isotherm alterations of pressure or temperature alter the 
density of the critical phase but do not lead to a separation into two phases. A 
density change is directly associated with a change of the solvent power, and thus 
the method features a high variability. Usually CO 2 is used as supercritical fl uid due 
to its critical point ( Tc = 31.1 ° C, Pc = 73.8 bars), which can be easily reached. That 
allows a moderate working temperature and leaves no toxic residues since it returns 
to the gas phase at ambient conditions. Two SC CO 2 - based processes have been 
reported for the preparation of drug - loaded polymeric microspheres: fi rst, the rapid 
expansion from supercritical solutions (RESS) process, whereby a SC CO 2 solution 
of an active agent and a polymeric carrier is rapidly expanded. This quickly transforms 
the SC CO 2 into a liquid that is a much poorer solvent, thereby precipitating 
the active agent – carrier mixture as small particles [65] . Second is the aerosol solvent 
extraction system (ASES), also referred as the gas antisolvent spray precipitation 
(GAS) process [66] . Here, a solution of the active agent and the polymeric carrier 
is sprayed into a chamber loaded with SC CO 2 . The SC CO 2 extracts the solvent 
from the spray droplets and induces coprecipitation of the active agent and the 
polymeric carrier in the form of small, solvent - free particles [67, 68] . However, the 
use of organic solvents cannot be avoided, which is to be deemed a major disadvantage 
of both techniques. 
In peptide/protein pharmaceuticals, the GAS process is predominantly applied 
for the preparation of microparticulate protein powders as an alternative to common 
drying processes. However, Winters et al. [69] reported an increase of . - sheet aggregates 
during the precipitation of lysozyme, trypsin, and insulin as a consequence of 
stress parameters such as organic solvent, pressure, and shear forces. One reason 

why these methods were not credited as encapsulation techniques for protein within 
PLGA may be the tendency of several polymers to rapidly precipitate and agglomerate 
during the process [70] . 
ASES has been compared with conventional spray drying in terms of effects on 
the stability of the peptide tetracosactide [71] . Almost no intact peptide was recovered 
from spray - dried PLA particles, whereas the tetracosactide was well protected 
against oxidation during ASES ( . 94% unmodifi ed peptide). In general, the particle 
formation step seems to be less detrimental to proteins than the loading step. For 
example, emulsifi cation in an aqueous phase or spray drying of rhEPO/PLGA 
emulsions was mild compared to the fi rst emulsifi cation step [72] . Also, variation of 
the particle formation step (spray drying or coacervation) had a minor impact on 
diphtheria toxoid (DTd) antigenicity when compared to other process variables 
[73] . 
A serious limitation of GAS, ASES, and RESS for producing microspheres is the 
need of polymer types that form discrete crystalline domains upon solidifi cation, 
such as l - PLA [74, 75] . The advantages of these methods offer (e.g., over spray 
drying) are the low critical temperatures for processing (34 ° C) and the avoidance 
of oxygen exposure during atomization, with both parameters being potentially 
important to peptide/protein stability. 
Ultrasonic Atomization Ultrasonic atomization of w/o dispersions is presently 
under investigation for preparing especially protein antigen containing microspheres. 
In one setup, the atomized antigen/polymer dispersion was sprayed into a 
nonsolvent where the polymer solvent was extracted, resulting in microspheres [76] . 
A comparable technique was proposed where the antigen or polymer dispersion 
was atomized into a reduced pressure atmosphere and the preformed microspheres 
hardened in a collection liquid [77] . Similarly, PLGA solutions were also atomized 
by acoustical excitation and the atomized droplets transported by an annular stream 
of a nonsolvent phase [aqueous polyvinyl alcohol (PVA)] into a vessel containing 
aqueous PVA [78] . Solvent evaporation and microsphere hardening occurred in the 
vessel over several hours. The main advantages of these atomization techniques 
encompass the possibility of easy particle size control and scale - up, processing at 
ambient or reduced temperature, and the suitability for aseptic manufacturing in 
a small containment chamber such as an isolator. 
In Situ Formed Injectable Microspheres All the encapsulation techniques discussed 
so far rely on the preparation of solid microspheres. However, a method for 
preparing a stable dispersion of protein containing semisolid PLGA microglobules 
has been reported [79] . Here, a protein dissolved in PEG 400 was added to a solution 
of PLGA in triacetin or triethyl citrate. This mixture, stabilized by Tween 80, 
was added dropwise and under stirring to a solution of Miglyol 812 or soybean oil, 
containing Span 80, resulting in a stable dispersion of protein inside semisolid 
PLGA microglobules. The microglobules remained in an embryonic state until 
mixed with an aqueous medium, so that the water - miscible components were 
extracted and protein containing matrix - type microspheres formed. Myoglobin was 
encapsulated and found to remain physically unchanged (circular dichroism 
analysis) after the process and during storage of the microglobular dispersion (15 
days, 4 ° C). 
PEPTIDE/PROTEIN-LOADED MICROSPHERE PRODUCTION METHODS 403

404 BIODEGRADABLE POLYMER-BASED MICROSPHERES 
Preformed Porous Microspheres A new approach for attaining sustained release 
of protein is introduced involving a pore - closing process of preformed porous 
PLGA microspheres [80] . Highly porous biodegradable PLGA microspheres were 
fabricated by a single w/o emulsion solvent evaporation technique using Pluronic 
F127 as an extractable porogen. The rhGH was incorporated into porous microspheres 
by a simple solution dipping method. For its controlled release, porous 
microspheres containing rhGH were treated with water - miscible solvents in the 
aqueous phase for production of pore - closed microspheres. These microspheres 
showed sustained - release patterns over an extended period; however, the drug 
loading effi ciency was extremely low. To overcome the drug loading problem, the 
pore - closing process was performed in an ethanol vapor phase using a fl uidized - bed 
reactor. The resultant pore - closed microspheres exhibited high protein loading 
amount as well as sustained rhGH release profi les. Also, the released rhGH exhibited 
structural integrity after the treatment. 
Charged (Anionic and Cationic) PLGA Microspheres PLGA or any other type 
of microspheres can be readily decorated with positive or negative surface charges 
by simply preparing the particles by a w/o/w solvent evaporation/extraction process 
where the second water phase contains a cationic emulsion stabilizer [hexadecyltrimethylammonium 
bromide; poly(ethyleneimine); stearylamine] or an anionic 
emulsifi er [sodium dioctyl - sulfosuccintate; sodium dodecyl sulfate (SDS)]. Such 
compounds attach tightly to PLGA surfaces during preparation and provide the 
necessary surface charge for ionic adsorption of counterions. It is known that a 
protein ’ s surface charge depends on its isoelectric point (pI) and the pH of the 
medium in which it is dispersed. The use of particles with ionic surface charge offers 
several advantages over classical microencapsulation, among which the mild conditions 
for loading are probably the most attractive. PLGA microspheres with surface - 
adsorbed protein antigens and DNA have been highly effi cient in inducing strong 
immune responses, as reviewed by Singh et al. [81] and Jilek et al. [82] . Nonetheless, 
it remains to be shown whether such particles are also suitable for eliciting long - 
term immunity after one or two injections. 
Jabbal - Gill et al. [83, 84] noted the tendency for microencapsulated protein antigens 
to distribute heavily at the surface of PLGA microspheres and developed 
polymeric lamellar substrate particles (PLSP) by precipitating a highly crystalline 
poly(l - lactic acid)/organic solvent solution with water, followed by removal of 
remaining organic solvent with nitrogen purge. The particles, which can be sterilized 
by gamma irradiation and stored as a suspension for several months without changes 
to antigen absorption [84] , possessed a large lamellar surface area and highly negative 
zeta potential ( .. 35 to . 42 mV) and could adsorb signifi cant amounts of antigen 
(up to 50 . g/mg microspheres) depending on pH, ionic strength, antigen – polymer 
ratio, and other factors. Release of protein antigen (TT) could be extended to over 
1 month with minimal antigenic losses in released antigen, although most of the 
antigen was lost to the initial burst or to apparent irreversible adsorption (as indicated 
by the absence of reaching 100% release). Elevated antibody responses in 
mice were elicited using PLSP similar to one dose of aluminum adjuvant following 
subcutaneous administration of OVA at elevated doses (100 or 300 . g). Both immunoglobulin 
IgG1 and IgG2a antibody subtypes were of similar magnitude over 28 
days in the PLSP/OVA groups, and cellular immunity was also observed following 

immunization with a 38 - kDa protein antigen against tuberculosis [85] . Similarly, 
Kazzaz et al. [86] created anionic PLGA microparticles by substituting the standard 
nonionic emulsifi er PVA with anionic SDS during microsphere preparation. In addition 
to eliciting elevated antibody responses in mice relative to the soluble antigen, 
the adsorbed antigen elicited a potent cytotoxic T - cell (CTL) response, similar to 
that observed after infection from virus expressing the p55 gag and polymerase 
proteins. Moreover, the CTLs were formed from the more challenging intramuscular 
route but not signifi cantly by the soluble antigen, even at elevated doses. The 
SDS - PLGA particles could also be gamma irradiated before adsorption and were 
shown to effectively boost antigen in nonhuman primates [87] . 
5.2.3 ANALYTICAL CHARACTERIZATION OF PEPTIDE/ 
PROTEIN - LOADED MICROSPHERES 
An area requiring additional efforts is analytical characterization of peptides and 
proteins encapsulated in PLGA microspheres. The high complexity of the therapeutic 
peptides and proteins requires not only physicochemical methodologies but 
also immunochemical and biological techniques for the characterization and quality 
control of these substances. In general, the analytical methods can be broadly 
viewed from the following study perspectives: methods meant for microsphere 
product quality checking, methods used for peptide/protein stability identifi cation 
inside the microspheres, and methods called for peptide integrity detection following 
liberation from the microspheres immediately upon placement in release 
medium either in vitro or in vivo. Therefore, in most cases, a combination of several 
analytical methods is necessary for a comprehensive characterization of the 
peptide/protein under investigation and for appropriate quality control of the 
product concerning identity, purity, and potency. However, some of the analytical 
methods have potentially appealing applications to interplay among the mentioned 
perspectives. In Table 4 , a selection of widely used analytical methods is given, 
showing which technology is applicable for the testing of identity, purity, and 
potency of peptides and proteins. In addition, peptide/protein integrity evaluation 
is indeed likely to be affected by artefacts during the sample preparation before 
analysis and during the analysis itself. Therefore, artefacts might prevent the scientist 
from critically ascribing detected protein denaturation to manufacturing 
conditions [88] . 
In order to measure the extent of peptide/protein degradation within the carriers 
and during release, the encapsulated molecule has to be removed from the polymeric 
matrix. Moreover, for avoiding artefacts such as underestimation of drug 
content, recovery methods need to be tried by an empirical trial - and - error approach 
as each peptide/protein is different one from the other. Recovery methods so far 
reported include extraction - based method with the help of potentially deleterious 
organic solvents, hydrolysis of the polymer matrix with alkaline medium, dissolution 
of polymer matrix in an organic solvent, recovery of suspended insoluble protein 
by fi ltration [89] , total protein quantifi cation after complete digestion of carriers 
followed by amino acid analysis [90, 91] , electrophoretic extraction of the protein 
using sodium dodecyl sulfate – polyacrylamide gel electrophoresis (SDS – PAGE) [92, 
93] , and direct dissolution of both the polymer and the protein drug in a single liquid 
ANALYTICAL CHARACTERIZATION 405

406 BIODEGRADABLE POLYMER-BASED MICROSPHERES 
TABLE 4 Analytical Methods for Characterization and Quality Control of 
Pharmaceutical Peptides and Proteins 
Methods 
Indicated Usage/Checking 
Identity Purity Potency 
Physicochemical 
Chromatography 
Reversed - phase high - performance liquid chromatography 
(HPLC, RP - 1) 
+ + . 
Ion exchange . + . 
Affi nity . + . 
Size exclusion chromatography (SEC) . + . 
Spectroscopy 
Infrared spectroscopy + + . 
Raman spectroscopy + + . 
Fluorescence spectroscopy . + . 
Ultraviolet/visible (UV/VIS) spectroscopy + + . 
NMR spectroscopy + + . 
Mass spectrometry + + . 
Circular dichroism (CD) + + . 
Matrix - assisted light desorption ionization – time - of - fl ight 
(MALDI - TOF) mass spectrometry 
+ + . 
Electrophoresis 
Capillary electrophoresis . + . 
SDS – polyacrylamide gel electrophoresis (PAGE) + + . 
Isoelectric focusing . + . 
Immunochemical 
Radioimmunoassay (RIA) . . + 
ELISA . . + 
Western blot + . . 
Biological 
In vivo assays . . + 
In vitro (cell culture) assays . . + 
Abbreviation: SDS: sodium do decyl sulfate. 
phase containing water - miscible organic solvents such as acetonitrile or dimethylsulfoxide 
(DMSO) [94, 95] . 
Following successful recovery of peptide/protein molecule from the microspheres, 
a simple spectrophotometric method does not always allow discrimination between 
the monomeric protein form and its aggregates. However, HPLC might separate 
these species and thus provides more accurate qualitative data [96] . But HPLC 
cannot quantify exclusively the amount of active protein antigen, as is the case with 
ELISA techniques [97] . Nowadays, Fourier transform infrared (FTIR) spectroscopy 
has become a popular, noninvasive method, as it is able to characterize the secondary 
structure of entrapped proteins [26, 95, 98 – 101] . Only recently, the integrity 
of their primary structure was evaluated, thanks to a new matrix - assisted laser 

desorption ionization – time - of - fl ight (MALDI - TOF) mass spectrometry method [94, 
102] . The method was shown to require little sample material and only a simple 
dissolution of the carrier was needed prior to the analysis. The MALDI - TOF allowed 
elucidation of a new degradation pathway, that is, peptide acylation within PLGA 
carriers resulting from a chemical interaction between peptide and degraded 
polymer [102] . Moreover, the method was also useful for quantifi cation, and it 
should be underlined that no interference from PLGA was detected during the 
measurements. For all the reasons cited above, mass spectrometry should be considered 
one of the most promising methods for protein analysis inside polymeric 
carriers including microspheres. Using erythropoietin as an example, an exploratory 
and elaborative discussion was made on the analytical techniques used for the 
characterization and quality control of pharmaceutical peptides and proteins [103] . 
A similar discussion was also done on the analytical techniques critical to (as a part 
of) the quality assurance after process changes of the production of a therapeutic 
antibody [104] . 
With an increasing level of sophistication in the design of new protein antigens 
and adjuvants (including polymer controlled - release systems), efforts both in the 
United States and the EU are underway to respond with more appropriate regulations 
[105 – 107] . For example, the Committee for Proprietary Medicinal Products 
(CPMP), the primary scientifi c body in EU regulatory matters, is currently updating 
its “ notes for guidance, ” which guide/direct industry and regulatory authorities on 
content and evaluation of marketing authorization applications for vaccines [105] . 
Early drafts of these updates include more rigorous guidelines for new non - 
aluminum - based adjuvants, including antigen stability requirements (see Sesardic 
and Dobbelaer [105] for a discussion). Similar discussions ongoing in the United 
States have attempted to standardize requirements of controlled - release parenterals 
[106, 107] , including specifi cs regarding in vitro release assays and the need to 
account for 80% or more of the encapsulating agent during the release period. 
5.2.4 IMMUNE SYSTEM INTERACTION WITH 
INJECTABLE MICROSPHERES 
Since microspheres are capable of forming a drug depot, the encapsulated peptide 
or protein is being slowly released over days or months at the injection site. Interestingly, 
the size of microspheres plays an important role in immune response. 
Microspheres with sizes smaller than 10 . m can be directly taken up macrophages 
(and dendritic cells) through a phagocytosis mechanism while sizes greater than 
10 . m need to undergo biodegradation before phagocytosis can occur [108] . It was 
shown that within a few days of intramuscular injection PLGA microspheres less 
than 10 . m are completely engulfed in a thin layer of connective tissue and thus 
evidenced infi ltration by macrophages as a consequence of wound - healing response 
to injected particles [109] . It is feasible that the infl ux of these macrophages may 
cause degradation of the encapsulated protein and available protein released in the 
vicinity of the microspheres. Furthermore, it has been suggested that these macrophages 
are capable of producing proteolytic enzymes [110] , which may result in the 
release and circulation of altered, inactive, or immunogenic forms of the encapsulated 
peptide or protein. 
IMMUNE SYSTEM INTERACTION WITH INJECTABLE MICROSPHERES 407

408 BIODEGRADABLE POLYMER-BASED MICROSPHERES 
On the other hand, degradation, protein antigen release, location, and antigen 
presentation of microspheres larger than 10 . m are expected to be different from 
smaller ones. Larger microspheres can provide an extracellular depot for secondary 
immune responses by way of B - cell stimulation [111 – 113] . In both cases, upon 
administration of microspheres, a foreign - body response occurs resulting in an acute 
initial infl ammation despite the excellent tissue compatibility and biodegradability 
properties of polymers such as PLGA. This initial infl ammation is followed by the 
infi ltration of small foreign - body giant cells and neutrophils [114] . These immune 
cells could consume the released peptide or protein and produce an immune 
response. However, if released protein is recognized as a self - protein (e.g., homologus), 
the probability of an immune response by these cells is reduced. It is therefore 
always essential to release the protein in its native conformation. The release of 
aggregated or denatured protein from the microspheres may, in fact, result in an 
unwanted immune response [115] . It should be added that systematic studies to 
explore the effects of tissue response on the bioavailability of incorporated peptide 
or protein drug have not appeared extensively in the literature, with a few exceptions 
as described below. Using a light microscopic technique, bumps containing 
residual amounts of microspheres were observed at the injection site two weeks 
after administration of TT - encapsulated PLGA microspheres to mice and guinea 
pigs [116] . These bump formations may be due to chronic reactions, long - term 
immunogenicity, and immunological priming of mice and guinea pigs against the 
injected polymeric microspheres. The immunogenicity of microsphere - encapsulated 
vaccines can be varied to some extent by changing the physicochemical properties 
of the microspheres, for example, size, surface properties, and release kinetics of 
the antigen from the microspheres [111] . An interesting review by Jiang et al. [117] 
details the various reports on the relationship between in vitro protein antigen stability 
and immunogenicity, modulation of cell - mediated immune responses, and 
different formulation approaches to achieve the appropriate immune response with 
microencapsulated vaccine antigens. There has been some debate, arising from some 
animal experiments, that the antigenicity does not directly correlate with immunogenicity. 
However, the stability of protein antigens is considered to play a signifi cant 
role in the quality and magnitude of immune response for the controlled - release 
single - dose vaccines as degraded or nonantigenic proteins may not be able to 
provide a continuous boost for generation of protective levels of high - affi nity 
antibodies. 
5.2.5 EXCIPIENT INCLUSION: INJECTABLE PEPTIDE/ 
PROTEIN - LOADED MICROSPHERES 
Peptide and protein molecules are highly prone to degradation mechanisms that 
can be divided into two classes: physical and chemical [118] . Whereas chemical 
degradation leads to the loss of the protein ’ s primary structure through oxidation, 
deamidation, peptide bond hydrolysis, isomerization, disulfi de exchange, and covalent 
aggregation, physical degradation refers to the changes in the higher order 
structure (secondary and above) mainly by noncovalent aggregation and precipitation. 
In particular, aggregates formation during the encapsulation process must be 
avoided because these aggregates always represent loss of therapeutic effi cacy and 

increased immunogenicity which can endanger the patient ’ s health [119, 120] . The 
following few examples indicate the fragility of peptide and protein molecules due 
to physical or chemical degradation: Aggregation of insulin has been well characterized 
and depends on unfolding of the insulin molecules [121] ; aggregation of lyophilized 
formulations of BSA, . - galactoglobulin, and glucose oxidase are attributed to 
disulfi de interchange [118] ; deamidation contributes to reduction in catalytic activity 
of lysozyme [122] and ribonuclease at high temperatures [123] ; and peptide bond 
hydrolysis results in loss of activity of lysozyme when heated to 90 – 100 ° C [122] . A 
recent introduction to this list is formaldehyde - mediated aggregation pathway 
(FMAP) unique to formaldehyde - treated protein antigens such as TT [117, 124, 
125] . 
The formulator of injectable microspheres for peptide and protein faces multiple 
challenges: (i) to maximize physical and chemical stability, (ii) to prolong biological 
half - life, (iii) to increase absorption, (iv) to decrease antigenicity, and (v) to minimize 
metabolism. Thus, it is quite obvious that the fabrication of peptide - and 
protein - loaded microspheres requires several kinds of excipients for effective stabilization 
or immobilization of encapsulated therapeutic molecules. Excipients of 
choice are included specifi cally for controlling protein degradation in microspheres 
due to (a) external and internal environmental changes, (b) manipulating the initial 
burst release, (c) preventing protein adsorption onto delivery devices, and (d) neutralizing 
the causative acidic microclimate formation due to the acids liberated by 
the biodegradable lactic/glycolic - based polymers. Therefore, it is generally best to 
fi nd conditions to stabilize the protein before other aspects of the formulation, such 
as controlled - release characteristics, are optimized. Typically, the appropriate excipients 
for the protein under investigation are experimentally selected among various 
substances by screening. This tedious experimental screening is partly necessary due 
to the present inability to predict protein stability after addition of such excipients. 
Moreover, since individual entrapped peptides and proteins differ in terms of physicochemical 
properties and chemical/therapeutic function, each species is expected 
to demonstrate a different degree of sensitivity to stress and react differently to the 
same stabilization strategy. For example, a sugar, amino acid, or antacid excipient 
may be required to stabilize protein, each of which can increase water uptake in the 
polymer matrix leading to an increase in release rate. In the scenario in which controlled 
- release conditions are optimized before such a stabilizer has been identifi ed, 
it is likely that upon addition of the new stabilizer the release kinetics may change 
enough to require reformulation. Certainly, there is a sharp contrast between encapsulating 
a highly water soluble protein [126] or a poorly soluble zinc – protein complex 
[37, 57] . Switching between these two cases would be expected to alter the requirements 
in the formulation necessary to attain the controlled - release function (e.g., 
low versus high polymer matrix permeability for the protein, respectively) because 
protein solubility in water may be important for any diffusion component of release. 
The principal stresses causing instability of encapsulated peptide/proteins in PLGA 
microspheres are elaborated in a book chapter [127] and in a jounal publication 
[128] . This subject was again reexamined in a review based on new fi ndings since 
the previous book chapter by the same author [129] . An interesting review from the 
same research group was published on the biodegradable PLGA microparticles for 
injectable delivery of vaccine antigens [117] , where they focused on mechanistic 
approaches to improve the stability of PLGA - encapsulated protein antigens. 
INJECTABLE PEPTIDE/PROTEIN-LOADED MICROSPHERES 409

410 BIODEGRADABLE POLYMER-BASED MICROSPHERES 
Another review by Bilati et al. [130] also envisioned the strategic approaches for 
overcoming peptide and protein instability within biodegradable nano - and microparticles. 
The reader is also referred to related publications edited by Sanders and 
Hendren [131] and Senior and Radomsky [132] for information on excipients used 
in injectable peptide/protein - loaded formulations including microspheres. This 
section will not cover all excipients used in parenteral protein formulations because 
the aforementioned publications already do so. Rather this section highlights examples 
of synergestic and antagonistic interactions that have been reported mainly 
between the excipients and the peptide/protein drugs, especially before microsphere 
preparation, followed by a brief discussion of a major instability problem of proteins/
peptides inside the microspheres. The published research paper is being organized 
according to major functions of parenteral excipients, namely, solubilization, 
stabilization, and preservation (see Figure 2 ) [133] . 
5.2.5.1 Solubility - and Stability - Increasing Excipients 
The traditional approach is to solubilize directly the peptide/protein in organic solvents. 
This can be achieved by different means. Cleland and Jones [61] assumed that 
native protein conformation could be maintained by precipitating the protein at its 
pI. The molecule is then free of charge and can be readily solubilized in organic 
solvents. Conversely, an alternative concept is based on the freeze drying of the 
protein at a pH away from its pI value before formulating it. It was thought that 
this strategy could increase protein solubility and stability in various polar and 
water - miscible organic solvents such as DMSO [134, 135] . It should be noted that 
a preformulation procedure consisting of using spray freeze drying with a suitable 
excipient was able to stabilize BSA before encapsulation by a nonaqueous method 
[35] . Using the dissolution approach, lysozyme was successfully formulated but 
incomplete lysozyme release from microspheres was observed and ascribed to 
aggregation [136] . Protein solubility can also be increased via an ion - pairing mechanism. 
The protein is modifi ed by adding an oppositely charged surfactant that binds 
to the protein, so as to obtain a neutral hydrophobic entity and thus reduce direct 
contact between the protein and the organic solvent. Positively charged proteins 
and negatively charged surfactants should be employed, since cationic surfactants 
might have toxic side effects. This technique was shown to improve lysozyme conformational 
stability after a hydrophobic complex between lysozyme and oleic acid 
[137, 138] . A new interesting concept is to encapsulate an aggregated protein in a 
reversibly dissociable form in order to avoid the formation of irreversible aggregates 
during processing and to promote the sustained release of the native monomeric 
form. Growth hormone was successfully formulated with this approach [139] . 
Solubilization Preservation Stabilization 
Excipients used 
Moisture-induced instability prevention Microclimate pH-induced instability prevention 
FIGURE 2 Flow chart of excipients used to prevent/minimize protein instability 
problems. 

Cyclodextrins (CD) have emerged as very effective additive compounds for 
solubilizing hydrophobic drugs. In the parenteral dosage form area, modifi ed cyclodextrins 
such as hydroxylpropyl - . - cyclodextrin and sulfobutylether - . - cyclodextrin 
have been reported to solubilize and stabilize many injectable drugs, including 
dexamethasone, estradiol, interleukin - 2, and other proteins and peptides [140] 
without apparent compatibility problems [141] . In addition, CD - containing formulations 
(either 0.1 M sulfobutylether - . - cyclodextrin or 0.1 M hydroxylpropyl - . - 
cyclodextrin) were shown to cause less damage to venous epithelial cells at the site 
of injection compared with formulations containing organic cosolvents [142] . When 
CD were coentrapped in the internal aqueous phase, erythropoietin (EPO) covalent 
aggregate formation was signifi cantly reduced during microsphere preparation by 
the double - emulsion method [72] and lysozyme stability was improved [88] . Although 
the precise mechanism is unclear, interactions between amino acids and the hydrophobic 
inner cavity of CD may play a role [143] . However, CD showed no protecting 
effect on insulinlike growth factor - 1 (IGF - 1) [144] and hepatitis B core antigen 
(HBcAg) [145] and even promoted the loss of superoxide dismutase activity at high 
CD concentrations [146] . By contrast, carboxymethylcellulose (CMC) did not effi - 
ciently stabilize HBcAg and GH against dichloromethane - induced denaturation 
[61, 145] . Various types ( . , . , and . ) of CD were examined for encapsulating TT in 
PLGA microspheres [147] , with . - hydroxypropyl - cyclodextrin effectively increasing 
TT encapsulation. However, CD also showed low effi ciency in retaining spray - dried 
TT antigenicity, probably due to antigenic epitopes being buried inside the molecular 
CD core [147] . 
Surfactants have the ability to lower surface tension of protein solutions and 
prevent protein adsorption and/or aggregation at hydrophobic surfaces such as 
PLGA. Among them, nonionic surfactants are generally preferred as ionic surfactants 
might bind to groups in proteins and cause denaturation. Tween 20 was 
shown to greatly reduce the rate of formation of insoluble aggregates of recombinant 
human factor XIII caused by both freeze thawing and agitation stresses 
[148] . Maximum protection occurs at concentrations close to the critical micelle 
concentration of Tween 20, independent of initial protein concentration. In 
another report, Tween 20 at a 1% (w/v) concentration caused precipitation of a 
relatively hydrophobic protein ( Humicola lanuginosa lipase) by inducing nonnative 
aggregates [149] . Similarly, nonionic surfactants such as Tween 20 or 80 were 
not good stabilizers for lysozyme and rhGH against the unfolding effect of the 
water – dichloromethane interface. It has been assumed that both the hydrophilic 
(PEG chains) and hydrophobic (fatty acid chain) parts of the polysorbate molecules 
were preferentially partitioned in the dichloromethane phase, leading 
to low protection effi cacy [61, 98] . Exchange of Tween 20 for a less hydrophobic 
surfactant, PEG 3350, provided almost complete rhGH recovery irrespective of 
protein concentration. However, an opposing trend was seen with EPO encapsulation 
in PLGA microspheres [72] . Encapsulated protein aggregates increased 
(. 15%) with different PEG types codissolved in the w 1 phase. Conversely, when 
three nonionic surfactants of different hydrophilic – lipophilic balances (HLBs) 
were coencapsulated with insulin by the w/o/w double - emulsion method, only 
Tween 20 was able to improve insulin stability within particles and to limit formation 
of high - molecular - weight products during the sustained - release period 
[150] . 
INJECTABLE PEPTIDE/PROTEIN-LOADED MICROSPHERES 411

412 BIODEGRADABLE POLYMER-BASED MICROSPHERES 
Tween 80 is well known to protect proteins against surface - induced denaturation 
[151] . Tween 80 was demonstrated to reduce hemoglobin aggregation in solution by 
preventing the protein from reaching the air – liquid interface or the liquid – surface 
interfaces [152] . Polyoxyethylene surfactants such as Tween 80 can form peroxide 
impurities after long - term storage. Knepp et al. [153] concluded that Tween 80 and 
other nonionic polyether surfactants undergo oxidation during bulk material storage 
and subsequent use and the resultant alkyl hydroperoxides formed can contribute 
to the degradation of proteins. In such formulations, they further reported that thiols 
such as cysteine, glutathione, and thioglycerol were most effective in stabilizing 
protein formulations containing peroxide - forming nonionic surfactants. 
The Pluronics, also known as poloxamers (e.g., poloxamer 188, British Pharmacopoeia 
standard) are a well - studied series of commercially available, nonionic, triblock 
copolymers with a central block composed of the relatively hydrophobic 
poly(propylene oxide) fl anked on both sides by blocks of the relatively hydrophilic 
poly(ethylene oxide) [154, 155] . The Pluronics possess an impressive safety profi le 
and are approved selectively by the Food and Drug Administration (FDA) for 
pharmaceutical and medical applications, including parenteral administration [156] . 
The strong safety profi le, commercial availability, ease of preparation, and well - 
studied physical properties make the Pluronics particularly appealing for drug delivery 
purposes. They have been used in several patented protein formulations as 
stabilizers and sustained - release injectables in development as solubilizing and stabilizing 
agents [157] . However, poloxamers, like Tweens, can form peroxide impurities 
over time. Poloxamer 188 was successfully used when mixed with PLGA for 
prolonged release of active interferon - . (INF - . ) [158] , but such a formulation had 
no effect on BSA secondary structure compared to PLGA alone [35] . Poloxamer 
188 was not effective in preventing nerve growth factor (NGF) aggregation during 
in vitro release from microspheres generated by spray drying [159] . Complex interactions 
between poloxamer, BSA, and PLGA were believed to have infl uenced BSA 
microencapsulation [160] . The gelling property of the amphiphilic poloxamer 407 
was successfully employed for urease encapsulation. The protein was likely protected 
during the microsphere preparation by a hydrated gelled structure due to the 
hydrophilic polyoxyethylene chains [161] . EPO aggregates in PLGA microspheres 
decreased when poloxamer 407 was incorporated at a level of 10% (w/w) [72] . 
Interleukin - 1 . (IL - 1 . ) was protected by phosphatidylcholine from damage 
during the double - emulsion process but underwent inactivation during microsphere 
incubation [162] . Sodium dodecyl sulfate signifi cantly reduced insulin aggregation 
at the dichloromethane – water interface, whereas dodecyl maltoside did not, this 
surfactant being more effi cient at air – water or solid – water interfaces [163] . It should 
be mentioned that surfactants are used along with sugars, proteins, and polymers 
effectively for solubilization and stabilization purposes of peptide/protein in microspheres. 
Bilati et al. [130] give an overview on various proteins and polymers that 
act as stabilizing excipients during the development of peptide/protein - loaded 
microspheres. 
5.2.5.2 Preservation - Imparting Excipients 
Prevention/Minimization of Moisture -Induced Instability Moisture - and microclimate 
acid pH – induced instability (typically the aggregation) of the peptide/protein 

encapsulated in PLGA microspheres has been monitored. Even several formulation 
strategies to inhibit these instability problems are being actively investigated. If the 
protein is expected to exist in the solid state within the PLGA polymer, the protein 
is remarkably prone to aggregation when formulated under conditions that allow 
moisture - and microclimate acid pH – induced instability. The two covalent aggregation 
mechanisms commonly described during exposure of the solid protein to moisture 
are the disulfi de interchange/exchange [164] and the FMAP, which is operative 
for protein antigens that have been detoxifi ed with formaldehyde exposure [124] . 
In the former pathway, the reaction is typically initiated by a thiolate ion on the 
protein or free thiolate ions that accompany . elimination of an intact disulfi de 
[165] . Decreasing the concentration of the reactive species (e.g., lowering pH to 
favor the nonionized thiol, covalently blocking the thiol group, or oxidizing free 
thiols as they appear with divalent copper ion) has been shown to block this mechanism 
[165] . To inhibit the FMAP, strongly formaldehyde - interacting amino acids 
such as histidine and lysine [166] have been colyophilized with the formalinized 
protein antigen. On exposure to moisture, the amino acids appear to bind with the 
reactive Schiff base or equivalent electrophile [167] in the protein before a neighboring 
protein nucleophile can react to form an intermolecular cross - link [124] . 
Sorbitol has also been identifi ed to inhibit the FMAP of TT at the maximal aggregating 
water content of the antigen, about 30 g H 2 O/g protein [168] , although whether 
this is a humectant effect [169] or a possible covalent reaction with the highly reactive 
electrophile in the antigen has not been determined. Several techniques have 
been developed to successfully bypass the destabilizing stress either by altering the 
role of water in the solid or immobilizing the protein or, alternatively, by directly 
inhibiting the aggregation. Clearly, one of the most signifi cant fi ndings in the fi eld 
of peptide/protein stability in polymers is the success of the immobilization strategy 
of Zn 2+ precipitation, as performed with human growth hormone [37, 57, 170, 171] . 
The 2 : 1 mole ratio Zn – protein complex, which immobilizes the rhGH as a solid 
precipitate in a near - native state [99] , has been shown to confer superior stability 
on the protein encapsulated in PLGA for a one - month release incubation. Since 
then, other proteins such as INF - . [172] and NGF [173] were also stabilized in 
PLGA microspheres by this approach. Another interesting approach originating in 
the patent literature is the precipitation of erythropoietin with salting - in salt, ammonium 
sulfate [174] , which is a technique commonly used in protein processing. Other 
methods to alter the role of water in the reaction involve the addition of agents that 
alter the amount of water sorbed in the polymer and/or the activity of the water 
present. For example, both water - soluble salts (NaCl) and antacid excipients 
(Mg(OH)2 ) are known to dramatically increase the amount of water sorbed in 
PLGAs, with the former due to osmosis and the latter to a complex effect of neutralizing 
acidic degradation products and end groups of the polymer (which also 
involves an osmotic component) [126] . In contrast, for a given moisture content, 
humectants such as sorbitol, which dissolve in water bound to the protein, reduce 
the available free water necessary to mobilize the protein or perform other roles in 
deleterious reactions [169] . 
The alternative to bypassing the deleterious role of moisture is to inhibit the 
aggregation mechanism directly. Several ways to accomplish this have been reported, 
particularly in the solid state and in the absence of the polymer. Well - referenced 
and useful book chapters by Johnson [175] and Carpenter and Chang [176] are 
INJECTABLE PEPTIDE/PROTEIN-LOADED MICROSPHERES 413

414 BIODEGRADABLE POLYMER-BASED MICROSPHERES 
available to thoroughly focus on the importance of making a lyophilized powder 
before loading the peptide or protein into an injectable microspheres. It has been 
stated that, in comparison to protein solution, the protein in the solid state would 
be less susceptible to shear forces that occur during an emulsifi cation procedure or 
denaturation at oil – water interfaces. However, special precautions should be taken 
during freeze drying because the drying process itself will expose the protein to 
destabilizing stresses. To circumvent this problem, cryo - and lyoprotectants and 
bulking agents are usually included along with a peptide or protein solution while 
it undergoes the drying stages of the lyophilization process. 
Cryo - and Lyoprotectants and Bulking Agents Various mechanisms are proposed 
to explain why excipients serve as cryo - or lyoprotectants. The most widely accepted 
mechanism to explain the action of cryoprotection is the preferential exclusion 
mechanism [177] . Excipients that will stabilize proteins against the effects of freezing 
do so by not associating with the surface of the protein. Such excipients actually 
increase the surface tension of water and induce preferential hydration of the 
protein. Examples of solutes that serve as cryoprotectants by this mechanism include 
amino acids, polyols, sugars, and polyethylene glycol. 
For lyoprotection, that is, stabilization of proteins during the drying stages of 
freeze drying and during storage in the dry state, two mechanisms are generally 
accepted. One is the water substitute hypothesis [178] and the other is the vitrifi cation 
hypothesis [179] . Both are legitimate theories, but both also have exceptions; 
that is, neither fully explain the stabilization of proteins by excipients during dehydration 
and dry storage [180] . The water substitute hypothesis states that a good 
stabilizer is one that hydrogen bonds to the protein just as water would do where 
it presents and, therefore, serves as a water substitute. Sugars are good water substitutes. 
(It may at fi rst appear contradictory that sugars can serve both as cryoprotectants 
because of being excluded from the surface of the protein and as 
lyoprotectants that hydrogen bond to the protein. However, keep in mind that the 
excluded solute concept involves a frozen aqueous system whereas the water substitute 
concept occurs in a dry system.) This is why many freeze - dried protein formulations 
contain sucrose or trehalose. Nevertheless, during a w/o/w procedure to 
prepare peptide/protein - loaded PLGA microspheres, sugars are often added to the 
inner aqueous phase. Trehalose was shown to partially improve the BSA secondary - 
structure protection within PLGA microspheres and to facilitate BSA monomer 
release [26] . Trehalose and mannitol had a signifi cant effect on the recovery of 
soluble nonaggregated interferon - . (INF - . ) and rhGH after emulsifi cation and 
ultrasonication [61] , whereas no or very little protecting effect on IGF - 1 against 
these stress factors was observed [144] . No effect of trehalose, mannitol, and sucrose 
was observed against o/w interface - induced degradation of lysozyme, whereas 
lactose and lactulose signifi cantly improved its structural stability and activity, mostly 
if these additives were also added to the second aqueous phase [88, 100] . Lysozyme 
and trypsin activity was not improved by addition of sucrose, which was unable to 
protect them from an emulsion - induced denaturation and from sonication [98, 181] . 
Mannitol and sucrose dissolved together in the inner aqueous phase had slight effect 
on NGF activity [182] and neither mannitol nor lactose improved HBcAg immunogenicity 
during dichloromethane/water emulsifi cation [145] . Surprisingly, sucrose 
and trehalose even decreased urease bioactivity, showing the opposite effect to that 

expected [161] . Coencapsulation of maltose reduced . - chymotrypsin aggregation 
[183] . With respect to microspheres generated by spray drying, trehalose was effective 
in retaining TT antigenicity [147] and in preventing BSA secondary - structure 
degradation [35] . Trehalose protected effi ciently NGF during the processing but did 
not prevent its aggregation during in vitro release [159] . 
The vitrifi cation hypothesis states that excipients that remain amorphous (glass 
formers) form a glassy matrix with the protein with the matrix serving as a stabilizer. 
Acceptance of this hypothesis requires formulators to determine glass transition 
temperatures of formulations to be freeze dried and to develop freeze - dry cycles 
that maintain drying temperatures below the glass transition temperature. Reports 
are available to indicate that excipient stabilizers, which are capable of undergoing 
crystallization during storage, caused degradation (typically aggregation and loss of 
potency) of the protein [176, 184, 185] . 
Freeze - dried formulations typically contain one or more of the following bulking 
agents: mannitol, lactose, sucrose, trehalose, dextran 40, and povidone. These excipients 
may also serve as cryo - and/or lyoprotectants in protein formulations. Fakes 
et al. [186] studied these bulking agents for moisture sorption behavior before and 
after freeze drying. Moisture uptake certainly can affect drug stability in the freeze - 
dried state, particularly with peptides and proteins. When selecting a bulking agent, 
these properties, particularly the tendency for moisture uptake, must be considered 
by the formulation scientist in developing an optimally stable freeze - dried formulation. 
Several excipients can serve as stabilizers for proteins that are unstable during 
the drying phases of freeze drying and/or during long - term storage in the dry state. 
Typically, additives that will crystallize during lyophilization (e.g., mannitol) or will 
remain amorphous but unable to hydrogen bond to the dried protein (e.g., dextran) 
are not effective lyoprotectants for proteins. Excipients that will crystallize during 
freeze drying will also be relatively ineffective, as was shown with sucrose in 
H. lanuginosa lipase formulations [149] . However, these authors also reported that 
sucrose crystallization could be inhibited by decreasing the mass ratio of sucrose to 
protein and by minimizing the moisture content that serves to decrease the glass 
transition temperature during storage. The reverse can also be true for certain small 
molecules. For example, excipients (mannitol or sodium bicarbonate) that promoted 
the crystallization of cyclophosphamide during freeze drying stabilized the fi nal 
product whereas excipients that did not allow crystallization (e.g., lactose) destabilized 
the fi nal product [187] . Costantino et al. [188] studied the effects of a variety 
of parenteral excipients on stabilizing human growth hormone in the lyophilized 
state. Mannitol, sorbitol, methyl a - d - mannopyranoside, lactose, trehalose, and cellobiose 
all provided signifi cant protection of the protein against aggregation, 
particularly at levels (131 : 1 excipient - to - protein molar ratio) to potentially satisfy 
water binding sites on the protein in the dried state. At higher excipient - to - protein 
ratios, mannitol and sorbitol crystallized and were not as effective in stabilizing the 
protein compared with low levels in which they remained in the amorphous, protein - 
containing phase. 
Reducing sugars may not be as effective as other bulking agents, cryoprotectants, 
or lyoprotectants because they may potentially react with proteins via the Maillard 
reaction. For example, glucose will form covalent adducts with side - chain amino 
acids lysine and arginine of human relaxin [189] . In addition, a signifi cant amount 
of serine cleavage from the C terminal of the B chain of relaxin was formed when 
INJECTABLE PEPTIDE/PROTEIN-LOADED MICROSPHERES 415

416 BIODEGRADABLE POLYMER-BASED MICROSPHERES 
glucose was used as the excipient. These reactions did not occur if mannitol and 
trehalose replaced glucose in the lyophilized formulation. Lactose will react with 
primary amines in the well - known Maillard - type condensation reaction to form 
brown - colored degradation products [190] . Thus, lactose is known to be incompatible 
with amine - containing compounds such as aminophylline, amphetamines, and 
amino acids/peptides. This reaction occurs more readily with amorphous lactose 
than crystalline lactose. 
Hydrophilic additives such as glucose are known to increase the porosity of 
microspheres, causing an increase in permeability to mass transport and a higher 
burst. However, a signifi cant reduction in initial burst release of a highly water - 
soluble model peptide, octreotide acetate, from poly( d , l - lactide - co - glycolide) 
microspheres by the coencapsulation of a small amount of glucose (e.g., 0.2% w/w) 
was reported [191] . Using the double emulsion – solvent evaporation method of 
encapsulation, the effect of glucose on initial burst in an acetate buffer pH 4 was 
found to depend on polymer concentration, discontinuous phase/continuous phase 
ratio, and glucose content. Extensive characterization studies were performed on 
two microsphere batches, ± 0.2% glucose, to elucidate the mechanism of this effect. 
However, no signifi cant difference was observed with respect to specifi c surface 
area, porosity, internal and external morphology, and drug distribution. Continuous 
monitoring of the fi rst 24 - h release of octreotide acetate from these two batches 
disclosed that, even though their starting release rates were close, the microspheres 
plus glucose exhibited a much lower release rate between 0.2 and 24 h compared to 
those without glucose. The microspheres plus glucose showed a denser periphery 
and a reduced water uptake at the end of the 24 - h release, indicating decreased 
permeability. However, this effect at times was offset as glucose content was further 
increased to 1%, causing an increase in surface area and porosity. In summary, these 
authors concluded that the effects of glucose on initial burst are determined by two 
factors: (1) increased initial burst due to increased osmotic pressure during encapsulation 
and drug release and (2) decreased initial burst due to decreased permeability 
of microspheres [191] . 
Mannitol is probably the most widely used bulking agent in lyophilized formulations 
because of its many positive properties with respect to crystallinity, high 
eutectic temperature, and matrix properties. However, some lots of mannitol can 
contain reducing sugar impurities that were implicated in the oxidative degradation 
of a peptide in a lyophilized formulation [192] . Mannitol at or above certain concentrations 
and volumes in glass vials is well known to cause vial breakage because 
of the unique crystallization properties of mannitol - ice during the primary drying 
states of freeze drying [193, 194] . 
Other Freeze - Dry Excipients High - molecular - weight carbohydrates such as dextran 
have higher glass transition temperatures than peptides/proteins. Therefore, when 
mixed with proteins, the overall glass transition temperature presumably can be 
increased with resultant increases in protein storage stability. Typically, carbohydrates 
(sucrose, trehalose, or dextran) alone do not result in appreciable increases 
in the storage stability of proteins. However, combinations of disaccharide and 
polymeric carbohydrates do tend to improve protein storage stability [195] . However, 
singular carbohydrates (sucrose or trehalose at 60 m M ) were also just as effective 
in stabilizing a model recombinant humanized monoclonal antibody as combinations 
of sucrose and mannitol or trehalose and mannitol. Interestingly, with this 

model monoclonal antibody, mannitol alone at 60 m M provided less protection 
during storage than sucrose or trehalose alone. A specifi c sugar/protein molar ratio 
was suffi cient to provide storage stability for this particular monoclonal antibody 
[196, 197] . 
Low - molecular - weight additives such as osmolytes ( N,N - dimethylglycine, trehalose, 
and sucrose) or salts (sodium chloride, sodium phosphate, ammonium sulfate, 
and sodium citrate) were found to be highly effective in stabilizing keratinocyte 
growth factor, both against thermal denaturation and enhancing long - term storage 
stability [198] . Nevertheless, the stabilizing properties of osmolytes appear to be 
balanced between their binding to (deteriorating effect) and exclusion from (stabilizing 
effect) the peptide/protein surface. As binding or exclusion predominantly 
results from hydrophobic interactions, hydrogen bonding, and electrostatic interactions, 
the sum of the various interaction parameters are dissimilar for different 
proteins. Therefore, it becomes crucial to examine the individual nature of the additive 
toward each individual protein and to assess whether it will offer a stabilizing 
or destabilizing effect [199, 200] . 
Polyvinyl pyrrolidone (PVP) and glycine were found to stabilize lyophilized 
sodium prasterone sulfate whereas dextran 40 or mannitol did not [201] . PVP and 
glycine stabilized the pH of the reconstituted solution by neutralizing the acidic 
degradation product, sodium bisulfate, formed by the hydrolysis of prasterone 
sulfate. Dextran 40 or mannitol was ineffective because of no buffer capacity. Buffering 
agents, such as phosphate – citrate buffer and some neutral and basic amino 
acids ( l - arginine, l - lysine, and l - histidine), also stabilized prasterone sulfate. l - Cysteine 
is an example of an amino acid that did not stabilize the drug, presumably 
because of its weak buffer capacity. Although the effi ciency of proteinic additives 
for protein stabilization has been clearly demonstrated in several occasions even 
during encapsulation processes [31, 72, 98, 144] , their use in pharmaceuticals is at 
present not desirable from a strictly regulatory point of view. Additionally, such 
agents might contribute to complicate all subsequent protein characterization within 
the formulation. Among these additives, albumins and gelatins are those mainly 
used for protection purposes. The protective effect of albumins against protein 
unfolding and aggregation has been extensively documented and is likely due to 
their surface - active properties (see Bilati et al. [130] for details). 
Prevention/Minimization of Microclimate pH -Induced Instability Evidence for 
acidifi cation within degrading microspheres is investigated and local pH values 
between 1.5 and 4.7 are being reported [202 – 204] . Methods to measure microclimate 
pH in PLGA microspheres include (i) ensemble average measurements using electron 
paramagnetic resonance (EPR) [203, 204] , nuclear magnetic resonance (NMR) 
[205] , and potentiometry and (ii) direct visualization techniques such as confocal 
imaging of pH - sensitive dyes [206, 207] . In the EPR method, the constant of hyper- 
fi ne splitting, 2 aN , was used to determine an average pH inside PLGA microspheres. 
Because the experiments relied on the mobility of spin - labeled protein, with an 
increase of the microviscosity in the later hours of the experiments, the spectra of 
EPR was changed and the signal - to - noise ratio decreased to prevent the measurement 
of pH throughout the release period [203] . The potentiometric measurements 
can give rapid values of pH for thin polymer fi lms, and the pH of the thin water fi lm 
between the electrode and polymer mimics the microclimate pH of aqueous pores 
inside the polymer - based drug delivery system. However, it is diffi cult to mimic 
INJECTABLE PEPTIDE/PROTEIN-LOADED MICROSPHERES 417

418 BIODEGRADABLE POLYMER-BASED MICROSPHERES 
microclimate pH of a small - scale system, such as microspheres or nanospheres, 
which may have unique microstructures, excipient/drug distributions, and transport 
characteristics. Overall, the ensemble average measurements described above could 
give a general picture of microclimate pH at the macroscopic level. However, the 
microscopic level of the detection can only be achieved through direct visualization 
techniques, such as microscopic imaging. Shenderova et al. [207] fi rst developed the 
confocal microscope imaging method to relate the microclimate pH with the fl uorescent 
intensity. Because of the diffi culty of controlling and predicting the fl uorescein 
concentration in the aqueous pore inside the microsphere, the method was only 
semiquantitative. Fu et al. [206] improved the confocal microscopic imaging method 
by coencapsulating two dextran fl uorescent dye (NERF and SNARF - 1) conjugates 
inside microspheres and related the ratio of the two dye images with microclimate 
pH in order to eliminate the poorly controlled effects of dye concentration and pore 
distribution. However, both of the dyes emit in the green range (535 nm for NERF 
and 580 nm of SNARF), giving rise to poor resolution without a narrow - bandwidth 
detector. Because of the high noise - to - signal ratio from the ratio images, the prediction 
of pH is also expected to be semiquantitative. In order to overcome the 
aforementioned drawbacks in microclimate pH measurement, a new quantitative 
ratiometric method based on laser scanning confocal microscopic imaging was 
developed to create a pixel - by - pixel neutral range microclimate pH map inside 
PLGA microspheres [208] . This method was then applied to both acid - neutralized 
and nonneutralized PLGA microspheres during extended incubation in physiological 
buffer. In another study, the PLGA water - soluble acid distribution has been 
measured with prederivatization HPLC [209] . 
Ongoing acidifi cation of the microsphere interior was shown to induce deamidation 
and covalent dimerization of nonreleased insulin [202] . Despite the evidence 
of acidifi cation mentioned above, there is controversy on this subject. It has been 
pointed out that the sampling scheme has a signifi cant impact on the degree of 
acidifi cation; frequent replenishment of the release medium or the use of a dialysis 
bag can effectively prevent the acidifi cation of the medium with subsequent 
reduced protein degradation [93, 210] . It is unsure, however, whether this also 
refl ects the situation in vivo, in which the PLGA microspheres are often surrounded 
by a fi brous capsule that may reduce effl ux of acidic degradation products 
from the PLGA matrix [93] . On the other hand, studies on rhGH - loaded PLGA 
microspheres showed a reasonable in vitro – in vivo correlation (IVIVC) only when 
a strong high - capacity buffer [200 m M N - (2 - hydroxyethyl)piperazine - N. - 2 - ethanesulfonic 
acid (HEPES), pH 7.4] was used, which effectively minimized the pH drop 
[211] . 
As indicated by the prevention of acid - induced physical aggregation of BSA in 
an abstract [212] , three principal ways have been identifi ed thus far to avoid the 
formation of highly acidic microclimate regions in the PLGAs during protein 
release: 
(i) Increasing the permeability of the polymer to facilitate escape of the water - 
soluble hydrolytic products of the PLGA polyester [125] 
(ii) Decreasing the degradation rate of the polyester [213] 
(iii) Coencapsulating additives to neutralize the weak acids formed by PLGA 
hydrolysis [126] 

In addition, two more ways that are likely to favor a lowering of microclimate pH 
are elevated initial acid content in the polymer [214] and low - frequency release 
media exchange [206] . 
The concept of controlling polymer permeability is diffi cult because attempts to 
increase permeability can spoil the controlled - release function of the polymer and 
cause the encapsulated protein to be released too rapidly. For example, Jiang and 
Schwendeman [213] increased the permeability of slow - degrading PLA (molecular 
weight (MW) 145 kDa) by blending in PEG (MW 10 or 30 kDa) at 0, 10, 20, and 
30%. Insoluble BSA aggregation in the PLA microspheres containing 4.5 – 5% w/w 
BSA was found in 0 and 10% PEG after a one - month incubation, but not in those 
preparations containing 20 or 30% PEG. The structural integrity of BSA was also 
intact in the stabilized formulations. However, between 10 and 30% PEG, the 
release rate of BSA increased rapidly and by 30% PEG, 60% of the protein encapsulated 
was released in only three days [213] . In contrast, an abstract [212] implied 
that 5% BSA encapsulated in a more permeable PLA (MW 77 kDa), the BSA 
formed < 2% insoluble aggregates over one month. This result suggested strongly 
that in some instances the slow degradation rate of the non - glycolic - acid - containing 
PLA is suffi cient to inhibit acid formation in the microclimate. 
In instances in which it is desirable to increase permeability and/or decrease the 
hydrolytic rate of PLGAs, that is, where a highly water - soluble protein requires 
release for one month or longer, it becomes necessary to coencapsulate a basic 
additive. Antacids such as MgCO 3 , Mg(OH) 2 or ZnCO 3 have been found to be particularly 
potent in preventing instabilty of acid - labile proteins [126, 215] . By means 
of thin fi lms coating pH glass electrodes to measure directly the microclimate acidic 
environment in PLGA microspheres, the stabilization against insoluble acid - induced 
noncovalent BSA aggregation afforded by a series of antacid excipients has been 
correlated with the ability of the antacid to neutralize acidic pores in fi lms of the 
same lot of PLGA coating pH glass electrodes [215] . Though much of the physical 
chemistry of microclimate pH adjustment with antacid additives is currently unclear, 
the strength of the base, the base solubility, and the association of the divalent 
cation with the carboxylate of the degradation products and/or polymer end groups 
appear to be important. For instance, Shenderova [216] has shown that from microclimate 
pH measurements in PLGA fi lms coating pH glass electrodes, MgCO 3 and 
Mg(OH)2 were found to be very similar under conditions which favor homogenous 
neutralization (i.e., high protein loading suffi cient to make pores for the base to 
diffuse all regions of the polymer matrix), but MgCO 3 was found to increase 
microclimate pH higher than Mg(OH) 2 . This result was consistent with the improved 
BSA stability in PLGA 50/50 microspheres when MgCO 3 was used in place of 
Mg(OH)2 [126] . 
5.2.6 PEPTIDE/PROTEIN ENCAPSULATED INTO BIODEGRADABLE 
MICROSPHERES: CASE STUDY 
Selected examples of therapeutic peptide and protein including vaccines which have 
been encapsulated into biodegradable polymer - based microspheres are discussed 
in this section. Besides what is mentioned below, many other proteins and 
vaccines have been encapsulated in biodegradable polymers, so a glimpse of ongoing 
PEPTIDE/PROTEIN ENCAPSULATED INTO BIODEGRADABLE MICROSPHERES 419

420 BIODEGRADABLE POLYMER-BASED MICROSPHERES 
research on microsphere delivery systems using biodegradable polymers is shown 
in Table 2 . 
5.2.6.1 Vaccines 
Group B Streptococcus Vaccine Group B streptococcus (GBS) is the leading 
bacterial cause of neonatal sepsis and meningitis. Although antibiotic prophylaxis 
has decreased the infection rate, the best long - term solution lies in the development 
of effective vaccines. The GBS capsular polysaccharide (CPS) is a major target of 
antibody - mediated immunity. The feasibility of producing a GBS having the ability 
to produce both a local IgA immune response at the mucosal surface and humoral 
IgG response having capability of transplacental passive immunization was 
investigated [217] . Inactivated GBS antigen was encapsulated in PLGA by a w/o/w 
multiple - emulsion technique along with immunostimulatory synthetic oligodeoxynucleotides 
containing cytosine phosphate guanosine (CpG) as potent adjuvant 
[217] . Immunization of female mice with normal immune systems was done with 
these PLGA microspheres containing GBS type III polysaccharide and CpG adjuvant 
(PLGA/GBS/CpG) and results indicated a signifi cantly higher GBS antibody 
response as compared to nonencapsulated GBS antigen or PLG - encapsulated GBS 
PS vaccine without the addition of the CpG. 
Diphtheria Toxoid ( DT) Diphtheria is a communicable disease caused by Corynebacterium 
diphtheriae which colonizes and forms a pseudomembrane at the 
infection site. This pathogen produces a potent protein toxin, diphtheria toxin, 
which is responsible for the typical systemic toxemia. DT is required for active 
immunization against diphtheria. DT was encapsulated in different types of PLA 
and PLGA microspheres by spray drying and coacervation. Immunization of 
guinea pigs with DT microspheres made with relatively hydrophilic PLGA 50 : 50 
resulted in specifi c and sustained antibody responses to alum adjuvanted toxoid in 
contrast to microspheres made with hydrophobic polymers where very low antibody 
responses were determined confi rming the feasibility of microsphere vaccines 
to induce strong, long - lasting protective antibody responses after single immunization 
[218] . 
In an endeavor toward development of multivalent vaccines based on biodegradable 
microspheres, Peyre et al. [219] tested the immunological performance of 
several divalent microsphere formulations against tetanus and diphtheria. Microspheres 
were made by separate microencapsulation of tetanus and diphtheria toxoid 
in PLGA by either spray drying or coacervation. Guinea pigs were subcutaneously 
immunized by a single injection of the divalent vaccines or, for control, an equivalent 
dose of a licensed vaccine containing both antigens adsorbed on aluminum hydroxide. 
All microsphere formulations were strongly immunogenic, irrespective of particle 
size and hydrophobicity. Endpoint titers of ELISA antibodies, mainly of the 
IgG1 subtype, were comparable to those obtained after immunization with the 
licensed vaccine. The microspheres provided increasing levels of antibodies, during 
the 16 weeks of testing, and the antibodies were weakly polarized toward tetanus. 
The induced antibodies were also toxin neutralizing, as determined for both diphtheria 
(1 – 4 IU/mL) and tetanus (5 – 9 IU/mL) eight weeks after immunization. These 
neutralization levels were several orders of magnitude above the level considered 

minimum for protection (0.01 IU/mL). When the animals were challenged with 
tetanus or diphtheria toxins six weeks after immunization, microsphere vaccines 
produced protective immunity that was comparable to or better than that induced 
by the licensed divalent vaccine. In conclusion, this study showed that a single 
administration of biodegradable microsphere vaccines provided protective immunity 
against diphtheria and tetanus and that this immunization approach might be 
feasible for multivalent vaccines. In a separate study, the same group have studied 
for the fi rst time the fate of immunogenic fl uorescent - labeled PLGA microspheres 
loaded with DT in vivo following a subcutaneous injection route [220] . 
A unique instability problem of DT is being foreseen when the DT would be 
encapsulated in PLGA microspheres along with a preservative such as thiomersal 
[221] . Thimerosal (TM) — also known as thiomersal, Merthiolate, or sodium 
ethylmercuri - thiosalicylate — is a water - soluble derivative complex of thiosalicylic 
acid (TSA) that has been used as bactericide in parenteral vaccines and ophthalmic 
products for decades. It has been reported that this preservative can be decomposed 
by oxidation to 2,2 - dithiosalicylic acid, ethyl mercuric ion, 2 - sulfenobenzoic acid, 2 - 
sulfobenzoic acid, and 2 - sulfi nobenzoic acid [222] . Namura et al. [221] demonstrated 
in vitro that the TSA, produced after the reduction of TM by lactic acid, reduces 
the S – S bridge of the previously incubated DT. This reduction is immediately followed 
by blocking the two SH groups formed by the same TSA molecules. In light 
of these conclusions, it is necessary now to reinterpret the in vitro protein degradation 
– stabilization data in the presence of PLGA microsphere, mainly for those 
proteins which contain S – S. The authors propose that all the PLGA microsphere 
microencapsulation studies and protein structural considerations should be done in 
the absence of TM as preservative. 
Tetanus Toxoid ( TT) Tetanus is considered a major health problem in developing 
and underdeveloped countries, with approximately one million new cases occurring 
each year. Tetanus is an intoxication manifested primarily by neuromuscular dysfunction. 
So vaccination is required for prevention of this disease. TT was encapsulated 
using PLGA with different molar compositions (50 : 50, 75 : 25) by the w/o/w 
multiple - emulsion technique and protein integrity was evaluated during antigen 
release in vitro in comparison to alum - adsorbed TT for in vivo induction of tetanus - 
specifi c antibodies [223] . TT microspheres elicited antibody titers as high as conventional 
alum - adsorbed TT, which lasted for 29 weeks, leading to the conclusion that 
TT microspheres can act as potential candidates for single - shot vaccine delivery 
systems. 
The study by Determan et al. [224] focuses on the effects of polymer degradation 
products on the primary, secondary, and tertiary structure of TT, OVA, and lysozyme 
after incubation for 0 or 20 days in the presence of ester (lactic acid and glycolic 
acid) and anhydride [sebacic acid and 1,6 - bis( p - carboxyphenoxy)hexane] monomers. 
The structure and antigenicity or enzymatic activity of each protein in the 
presence of each monomer was quantifi ed. SDS - PAGE, circular dichroism, and fl uorescence 
spectroscopy were used to assess/evaluate the primary, secondary, and 
tertiary structures of the proteins, respectively. ELISA was used to measure changes 
in the antigenicity of TT and OVA and a fl uorescence - based assay was used to 
determine the enzymatic activity of lysozyme. TT toxoid was found to be the most 
stable in the presence of anhydride monomers, while OVA was most stable in the 
PEPTIDE/PROTEIN ENCAPSULATED INTO BIODEGRADABLE MICROSPHERES 421

422 BIODEGRADABLE POLYMER-BASED MICROSPHERES 
presence of sebacic acid, and lysozyme was stable when incubated with all of the 
monomers studied. 
Jaganathan et al. [225] compared the effi ciency of microspheres produced from 
PLGA and chitosan polymers by using protein stabilizer (trehalose) and acid - 
neutralizing base [Mg(OH) 2 ]. The immunogenicity of PLGA - and chitosan microsphere 
– based single - dose vaccine was evaluated in guinea pigs and compared with 
multiple doses of alum - adsorbed TT. Results indicated that a single injection of 
PLGA and chitosan microspheres containing TT could maintain the antibody 
response at a level comparable to the booster injections of conventional alum - 
adsorbed vaccines. Both the PLGA - and chitosan - based stable vaccine formulations 
produced an equal immune response. Hence chitosan can be used to replace the 
expensive polymer PLGA. This approach should have potential application in the 
fi eld of vaccine delivery. 
The study by Kipper et al. [226] focuses on the development of single - dose vaccines 
based on biodegradable polyanhydride microspheres that have the unique 
capability to modulate the immune response mechanism. The polymer system 
employed consists of copolymers of 1,6 - bis( p - carboxyphenoxy)hexane and sebacic 
acid. Two copolymer formulations that have been shown to provide extended - 
release kinetics and protein stability were investigated. Using TT as a model antigen, 
in vivo studies in C3H/HeOuJ mice demonstrated that the encapsulation procedure 
preserves the immunogenicity of the TT. The polymer itself exhibited an adjuvant 
effect, enhancing the immune response to a small dose of TT. The microspheres 
provided a prolonged exposure to TT suffi cient to induce both a primary and a 
secondary immune response (i.e., high antibody titers) with high - avidity antibody 
production, without requiring an additional administration. Antigen - specifi c proliferation 
28 weeks after a single immunization indicated that immunization with the 
polyanhydride microspheres generated long - lived memory cells and plasma cells 
(antibody - secreting B cells) that generally do not occur without maturation signals 
from T helper cells. Furthermore, by altering the vaccine formulation, the overall 
strength of the T - helper type 2 immune response was selectively diminished, resulting 
in a balanced immune response, without reducing the overall titer. This result is 
striking, considering free TT induces a T - helper type 2 immune response and has 
important implications for developing vaccines to intracellular pathogens. The 
ability to selectively tune the immune response without the administration of additional 
cytokines or noxious adjuvants is a unique feature of this delivery vehicle that 
may make it an excellent candidate for vaccine development. 
Polylactide (PLA) polymer particles entrapping TT were evaluated in terms of 
particle size, antigen load, dose, and additional adjuvant for achieving high and 
sustained anti - TT antibody titers from single - point intramuscular immunization 
[227] . Admixture of polymer - entrapped TT and alum improved the immune response 
in comparison to particle - based immunization. High and long - lasting antibody titer 
was achieved upon immunization with 2 – 8 - . m size microparticles. Microspheres 
within the size range 50 – 150 . m elicited very low serum antibody response. Immunization 
with very small particles ( < 2 . m) and with intermediate - size - range particles 
(10 – 70 . m) elicited comparable antibody response from single - point immunization 
but lower in comparison to that achieved while immunizing with 2 – 8 - . m particles. 
Potentiation of antibody response on immunization of admixture of microspheres 
and alum was also dependent on particle size. These results indicate the need of 

optimal particle sizes in micrometer ranges for improved humoral response from 
single - point immunization. Increasing antigen load on polymer particles was found 
to have a positive infl uence on the generation of antibody titers from particle - based 
immunization. Maximum peak antibody titer of . 300 . g/mL was achieved on day 
50 upon immunization with particles having the highest load of antigen (94 . g/mg 
of polymer). Increase in dose of polymer - entrapped antigen resulted in concomitant 
increase in peak antibody titers, indicating the importance of antigen stability, particle 
size, and load on generating a reproducible immune response. Optimization of 
particle size, antigen load, dose, and use of additional adjuvant resulted in high and 
sustained anti - TT antibody titers over a period of more than 250 days from single - 
point immunization. Serum anti - TT antibody titers from single - point immunization 
of admixture of PLA particles and alum were comparable with immunization from 
two divided doses of alum - adsorbed TT. 
Vibrio Cholerae ( VC) Whole -Cell Vaccine Cholera, an acute intestinal infection 
caused by the bacterium Vibrio cholerae , produces an enterotoxin that causes a 
copious, painless, watery diarrhea that can quickly lead to severe dehydration and 
death if treatment is not promptly given. For prevention of cholera, cholera vaccine 
is usually given. VC was successfully entrapped in the PLGA microspheres by a 
double - emulsion method with trapping effi ciencies up to 98%. The immnunogenic 
potential of VC - loaded microspheres physically mixed with or without amphotericin 
B was evaluated in adult mice by oral immunization in comparison to VC solution. 
The immunogenicity of VC - loaded microparticles mixed with amphotericin B in 
evoking Vibrio - specifi c serum IgG and IgM responses was higher than that of VC - 
loaded microparticles only [228, 229] . However, VC was loaded in different polymer 
compositions (50 : 50 PLGA, 75 : 25 PLGA, and PLA/PEG blended), the higher 
antibody responses and serum IgG, IgA, and IgM responses were obtained when 
sera from both VC - loaded 75 : 25 PLGA and PLA/PEG - blended microparticles 
immunized mice were titrated against VC solution [230] . 
Japanese Encephalitis Virus ( JEV) Japanese encephalitis is a disease that is 
spread to humans by infected mosquitoes in Asia. It is one of a group of 
mosquito - borne viral diseases that can affect the central nervous system and 
cause severe complications and even death. Vaccination is one of the ways of 
treating it. JEV vaccine was encapsulated in PLGA microspheres by a double - 
emulsion technique and infl uences of various process variables such as stirring 
rate, types and concentration of emulsifi er, and polymer concentration were 
studied on size, size distribution, and biodegradation. The mean size of microspheres 
decreased with increasing speed, increasing concentration of emulsifi er, 
and decreasing polymer concentration. Rate of biodegradation of nonporous 
microspheres was slower than that of porous microspheres, leading to the conclusion 
that PLGA microspheres can be used to apply oral vaccination through 
Peyers patches across the gastrointestinal tract (GIT) [231] . 
Several approaches to develop an improved JEV vaccine are in progress in 
various laboratories. Of these, immunization of mice with plasmid DNA encoding 
JEV envelope (E) protein has shown great promise. The technology, developed by 
Kaur et al. [232] , involved the adsorption of DNA onto cetyltrimethyl - ammonium 
bromide (CTAB) containing cationic poly(lactide - co - glycolide) (PLG) microspheres. 
PEPTIDE/PROTEIN ENCAPSULATED INTO BIODEGRADABLE MICROSPHERES 423

424 BIODEGRADABLE POLYMER-BASED MICROSPHERES 
The microsphere - adsorbed DNA induced a mixed Th1 – Th2 immune response as 
opposed to Th1 immune responses elicited by the naked DNA. 
JEV - loaded poly(lactide) (PLA) lamellar and PLG microspheres were successfully 
prepared with low - molecular - weight PLA by the precipitate method and with 
6% w/v PLG in the organic phase, 10% w/v PVP, and 5% w/v NaCl in the continuous 
phase by using a w/o/w emulsion/solvent extraction technique, respectively [233] . 
The JEV incorporation, physicochemical characterization data, and animal results 
obtained in this study may be relevant in optimizing the vaccine incorporation and 
delivery properties of these potential vaccine targeting carriers. 
Hepatitis B Virus Hepatitis B is one of the most important infectious diseases in 
the world. Approximately 350 million people worldwide are chronic carriers of the 
hepatitis B virus (HBV), which accounts for approximately one million deaths annually. 
PLGA microspheres loaded with recombinant HBsAg were formulated using 
a double - emulsion technique. The pharmaceutical characteristics of size, surface 
morphology, protein loading effi ciency, antigen integrity, release of HBsAg - loaded 
PLGA microspheres, and degradation of the polymer in vitro were evaluated [234 – 
237] . Based on these fi ndings in vitro and in vivo, it was concluded that HBsAg was 
successfully loaded into the PLGA microspheres, which can autoboost an immune 
response, and the HBsAg - loaded PLGA microsphere is a promising candidate for 
the controlled delivery of a vaccine. 
5.2.6.2 Proteins 
Prolidase Defi ciency of this enzyme results in chronic intractable ulcerations of 
the skin, particularly of lower limbs, since it is involved in the fi nal stages of protein 
catabolism. To counteract the problem, the enzyme was encapsulated in PLGA 
microspheres by a double - or multiple - emulsion technique, in vitro and ex vivo 
evaluations were done, and the results indicated that microencapsulation stabilizes 
the enzymatic activity inside the PLGA microspheres resulting in both in vitro and 
ex vivo active enzyme release, hence opening the doors for the possibility of enzyme 
replacement therapy through microencapsulation [238] . Further evaluation from 
the same research group for prolidase - loaded PLGA microspheres is reported 
elsewhere [239, 240] . 
Insulin Insulin is the most important regulatory hormone in the control of glucose 
homeostasis. The World Health Organization (WHO) has indicated that more than 
50 million people around the world suffer from diabetes and require daily parenteral 
injections of insulin to stay healthy and live normally. For the treatment of type 
I diabetes insulin still is number one, with three subcutaneous injections to be taken 
per day. A controlled - release system for a long - term therapy of this disease is the 
need of the hour, as this can obviate the need for painful injection given a number 
of times to the diabetes patients. Insulin was encapsulated in blends of poly(ethylene 
glycol) with PLA homopolymer and PLGA copolymer by a w/o/w multiple - 
emulsion technique with entrapment effi ciencies up to 56 and 48% for PLGA/ PEG 
and PLA/ PEG, respectively [12] . Insulin - loaded microspheres were capable of 
controlling the release of insulin for 28 days with in vitro delivery rates of 0.94 and 
0.65 . g insulin/mg per particle per day in the fi rst 4 days and steady release with a 

rate of 0.4 and 0.43 . g insulin/mg per particle per day over the following 4 weeks, 
respectively, along with the extensive degradation of PLGA/ PEG microspheres as 
compared to PLA/ PEG blends which resulted in a stable particle morphology along 
with reduced fragmentation and aggregation of associated insulin. 
Two types of injectable cationized microspheres were prepared based on a native 
gelatin (NGMS) and aminated gelatin with ethylenediamine (CGMS) to prolong 
the action of insulin [241] . Release of rhodamin B isothiocyanate insulin from 
CGMS was compared with that from NGMS under in vitro and in vivo conditions. 
Lower release of insulin from CGMS compared with that from NGMS was caused 
by the suppression of initial release. The disappearance of 125 I - insulin from the injection 
site after intramuscular administration by NGMS and CGMS had a biphasic 
profi le in mice. Almost all the 125 I - insulin had disappeared from the injection site 
one day after administration by NGMS. The remaining insulin at the injection site 
after administration by CGMS was prolonged, with approximately 59% remaining 
after 1 day and 16% after 14 days. The disappearance of CGMS from the injection 
site was lower than that of NGMS. However, the difference in these disappearance 
rates was not great compared with those of 125 I - insulin from the injection site by 
NGMS and CGMS. The time course of disappearance of 125 I - CGMS from the injection 
site was similar to that of 125 I - insulin by CGMS. The initial hypoglycemic effect 
was observed 1 h after administration of insulin by NGMS, and thereafter its effect 
rapidly disappeared. The hypoglycemic effect was observed 2 – 4 h after administration 
by CGMS and continued to be exhibited for 7 days. The prolonged hypoglycemic 
action by CGMS depended on the time profi les of the disappearance of insulin 
from muscular tissues, which occurs due to the enzymatic degradation of CGMS. 
A novel controlled - release formulation was developed with PEGylated human 
insulin encapsulated in PLGA microspheres that produces multiday release in vivo 
[242] . The insulin is specifi cally PEGylated at the amino terminus of the B chain 
with a relatively low molecular weight PEG (5000 Da). Insulin with this modifi cation 
retains full biological activity but has a limited serum half - life, making microencapsulation 
necessary for sustained release beyond a few hours. PEGylated insulin can 
be codissolved with PLGA in methylene chloride and microspheres made by a 
single o/w emulsion process. Insulin conformation and biological activity are preserved 
after PEGylation and PLGA encapsulation. The monolithic microspheres 
have inherently low burst release, an important safety feature for an extended - 
release injectable insulin product. In PBS at 37 ° C, formulations with a drug content 
of approximately 14% show very low ( < 1%) initial release of insulin over one day 
and near - zero - order drug release after a lag of three to four days. In animal studies, 
PEG - insulin microspheres administered subcutaneously as a single injection produced 
< 1% release of insulin in the fi rst day but then lowered the serum glucose 
levels of diabetic rats to values < 200 mg/dL for approximately nine days. When the 
doses were given at seven - day intervals, steady - state drug levels were achieved after 
only two doses. PEG - insulin PLGA microparticles show promise as a once - weekly 
dosed, sustained - release insulin formulation. 
Shenoy et al. [243] developed an injectable, depot - forming drug delivery system 
for insulin based on microparticles technology to maintain constant plasma drug 
concentrations over a prolonged period of time for the effective control of blood 
sugar levels. Formulations were optimized with two well - characterized biodegradable 
polymers, namely PLGA and poly - . - caprolactone, and evaluated in vitro for 
PEPTIDE/PROTEIN ENCAPSULATED INTO BIODEGRADABLE MICROSPHERES 425

426 BIODEGRADABLE POLYMER-BASED MICROSPHERES 
physicochemical characteristics, drug release in phosphate - buffered saline (pH 7.4), 
and evaluated in vivo in streptozotocin - induced hypoglycemic rats. With a large 
volume of internal aqueous phase during a w/o/w double - emulsion solvent evaporation 
process and high molecular weight of the polymers used, they could not achieve 
high drug capture and precise control over subsequent release within the study 
period of 60 days. However, this investigation revealed that upon subcutaneous 
injection the biodegradable depot - forming polymeric microspheres controlled the 
drug release and plasma sugar levels more effi ciently than plain insulin injection. 
Preliminary pharmacokinetic evaluation exhibited steady plasma insulin concentration 
during the study period. These formulations, with their reduced frequency of 
administration and better control over drug disposition, may provide an economic 
benefi t to the user compared with products currently available for diabetes 
control. 
Interferon a2a ( IFN a2a) Interferon .2a is indicated for the treatment of adults 
with chronic hepatitis C virus infection who have compensated liver disease and 
have not been previously treated with interferon . . To improve the stability and 
loading effi ciency of protein drugs, a new microsphere delivery system comprises 
calcium alginate cores surrounded by PELA [poly - D,L - lactide - poly - (ethylene 
glycol)]. Recombinant IFN .2a as a model drug was entrapped within calcium alginate 
cores surrounded by PELA by a w/o/w multiple - emulsion technique [244] . 
Core - coated microspheres stabilized the IFN in the PELA matrix. The core - coated 
microspheres indicated high encapsulation effi ciency and biological retention as 
compared to conventional PLGA microspheres. The extent of burst release reduced 
to 14% in core - coated microspheres from 31% in conventional microspheres, indicating 
a new approach for water - soluble macromolecular drug delivery. 
5.2.7 CONCLUSION 
From this chapter, it has become apparent that a number microencapsulation 
methods are available today for the preparation of microspheres on an industrial 
scale. In fact, parenteral drug delivery systems based upon biodegradable microspheres 
are a true success story for the concept of drug delivery. However, the production 
of biodegradable microspheres containing a stable therapeutic peptide or 
protein still remains a major challenge in terms of technical obstacles. Ideally, peptides/
proteins of therapeutic interest should be studied case by case, so as to bring 
to the fore processing steps and stress factors which damage them. Continued efforts 
to establish methods for stable protein, especially antigen, delivery from microspheres 
may hopefully pave the way for future microsphere - based vaccines. Areas 
of further research should focus on the performance of peptide/protein - loaded 
microspheres under in vitro and in vivo conditions. Interestingly, the addition of 
medium - chain triglycerides (MCT) modifi es/shifts the triphasic release pattern of 
leuprolide acetate - loaded PLGA microspheres to a more continuous release in vitro 
[245] . Alternatively, BSA - loaded PLGA microspheres were coated with a thermosensitive 
gel, Pluronic F127 (PF127) [246] . The results demonstrated that PF127, 
which gelled at 37 ° C, inhibited the initial burst release of BSA from microspheres 
effectively. It is anticipated that more efforts will be invested in the future to develop 

REFERENCES 427 
novel ways to reduce the initial burst release of entrapped peptide/protein and to 
attain a more continuous release. In addition, an in vitro release model mimicking 
the fate of biodegradable microspheres applied through the parenteral route would 
be highly desirable. Also, new strategies to stabilize proteins in microspheres during 
manufacturing, shelf life, or in vivo could be of general interest. Moreover, the use 
of analytical techniques such as FTIR or MALDI - TOF mass spectrometry certainly 
constitutes a step forward for protein analysis in more appropriate conditions. 
REFERENCES 
1. Brooks , G. ( 1998 ), Biotechnology in Healthcare , Pharmaceutical Press , London . 
2. Walsh , G. ( 2002 ), Biopharmaceuticals and biotechnology medicines: An issue of nomenclature 
, Eur. J. Pharm. Sci ., 15 , 135 – 138 . 
3. Walsh , G. ( 2003 ), Pharmaceutical biotechnology products approved within the European 
Union , Eur. J. Pharm. Biopharm. , 55 , 3 – 10 . 
4. New biotechnology medicines in development report (2002), available: http://pharma. 
org , accessed May 25, 2006. 
5. Banga , A. K. , and Chien , Y. W. ( 1988 ), Systemic delivery of therapeutic peptides and 
proteins , Int. J. Pharm ., 48 , 15 – 50 . 
6. Lee , V. H. L. ( 1987 ), Ophthalmic delivery of peptides and proteins , Pharm. Technol ., 11 , 
26 – 38 . 
7. Athanasiou , K. A. , Niederauer , G. G. , and Agrawal , C. M. ( 1996 ), Sterilization, toxicity, 
biocompatibility and clinical applications of polylactic acid/polyglycolic acid copolymers . 
Biomaterials , 17 , 93 – 102 . 
8. Ueda , H. , and Tabata , Y. ( 2003 ), Polyhydroxyalkanonate derivatives in current clinical 
applications and trials , Adv. Drug Deliv. Rev ., 55 , 501 – 518 . 
9. Ramesh , V. D. , Medlicott , N. , Razzak , M. , and Tucker , I. G. ( 2002 ), Microencapsulation 
of FITC - BSA into poly(e - caprolactone) by a water - in - oil - in - oil solvent evaporation 
technique . Trends Biomater. Artif. Organs , 15 , 31 – 36 . 
10. Dhanaraju , M. D. , Vema , K. , Jayakumar , R. , and Vamsadhara , C. ( 2003 ), Preparation and 
haracterization of injectable microspheres of contraceptive hormones , Int. J. Pharm ., 268 , 
23 – 29 . 
11. Tracy , M. A. ( 1998 ), Development and scale - up of a microsphere protein delivery system , 
Biotechnol. Prog ., 14 , 108 – 115 . 
12. Yeh , M. K. ( 2000 ), The stability of insulin in biodegradable microparticles based on 
blends of lactide polymers and polyethylene glycol , J. Microencapsul ., 17 , 743 – 756 . 
13. Han , K. , Lee , K. D. , Gao , Z. G. , and Park , J. J. ( 2001 ), Preparation and evaluation of 
poly(l - lactic acid) microspheres containing rhEGF for chronic gastric ulcer healing , 
J. Controlled Release , 75 , 259 – 269 . 
14. Jackson , J. , Liang , L. , Hunter , W. , Reynolds , M. , Sandberg , J. , Springate , C. , and Burt , H. 
( 2002 ), The encapsulation of ribozymes in biodegradable polymeric matrices , Int. J. 
Pharm ., 243 , 43 – 47 . 
15. Blanco - Prieto , M. J. , Besseghir , K. , Zerbe , O. , andris , D. , Orsolini , P. , Heimgartner , F. , 
Merkle , H. P. , and Gander , B. ( 2000 ), In vitro and in vivo evaluation of a somatostatin 
analogue released from PLGA microspheres , J. Controlled Release , 67 , 19 – 28 . 
16. Yuksel , E. , Weinfeld , A. B. , Cleek , R. , Waugh , J. M. , Jensen , J. , Boutros , S. , Shenaq , S. M. , 
and Spira , M. ( 2000 ), De novo adipose tissue generation through long - term, local delivery 
of insulin and insulin like growth factor - 1 by PLGA/PEG microspheres in an in vivo 
rat midel: A novel concept and capability , Plastic Reconstruct. Surg ., 105 , 1721 – 1729 . 

428 BIODEGRADABLE POLYMER-BASED MICROSPHERES 
17. Kostanski , J. W. , Thanoo , B. C. , and Deluca , P. P. ( 2000 ), Preparation, characterization 
and in vitro evaluation of 1 - and 4 - month controlled release ornitide PLA and PLGA 
microspheres , Pharm. Dev. Technol ., 5 , 585 – 596 . 
18. King , T. W. , and Patrick , Jr ., C. W. ( 2000 ), Development and in vitro characterization of 
vascular endothelial growth factor (VEGF) loaded poly( dl - lactic - co - glycolicacid)/ 
poly(ethylene glycol) microspheres using a solid encapsulation/single emulsion/solvent 
extraction technique , J. Biomed. Mater. Res ., 51 , 383 – 390 . 
19. Zhu , K. J. , Jiang , H. L. , Du , X. Y. , Wang , J. , Xu , W. X. , and Liu , S. F. ( 2001 ), Preparation 
and characterization of hCG - loaded polylactide or poly(lactide -co - glycolide) microspheres 
using a modifi ed water - in - oil - in - water (w/o/w) emulsion solvent evaporation 
technique , J. Microencapsul ., 18 , 247 – 260 . 
20. Prabhu , S. , Sullivan , J. L. , and Betageri , G. V. ( 2002 ), Comparative assessment of in vitro 
release kinetics of calcitonin polypeptide from biodegradable microspheres , Drug Deliv ., 
9 , 195 – 198 . 
21. Oldham , J. B. , Lu , L. , Zhu , X. , Porter , B. D. , Hefferan , T. E. , Larson . D. R. , Currier , B. L. , 
Mikos , A. G. , and Yaszemski , M. J. ( 2000 ), Biological activity of rhBMP - 2 released from 
PLGA microspheres , J. Biomech. Eng ., 122 , 289 – 292 . 
22. Woo , B. H. , Fink , B. F. , Page , R. , Schrier , J. A. , Jo , Y. W. , Jiang , G. , DeLuca , M. Vasconez , 
H. C. , and DeLuca , P. P. ( 2001 ), Enhancement of bone growth by sustained delivery of 
recombinant human bone morphogenetic protein - 2 in a polymeric matrix , Pharm. Res ., 
18 , 1747 – 1753 . 
23. Weber , F. E. , Eyrich , G. , Gratz , K. W. , Maly , F. E. , and Sailer , H. F. ( 2002 ), Slow and continuous 
application of human recombinant bone morphogenetic protein via biodegradable 
poly(lactide -co - glycolide) foamspheres , Int. J. of Oral Maxillofacial Surg ., 31 , 
60 – 65 . 
24. Bordem , M. D. , Khan , Y. , Attawia , M. , and Laurencin , C. T. ( 2001 ), Tissue engineered 
microsphere - based matrices for bone repair: Design, evaluation and optimisation , Biomaterials 
, 23 , 551 – 559 . 
25. Lu , L. , Stamatas , G. N. , and Mikos , A. G. ( 2000 ), Controlled release of transforming 
growth factor beta 1 from biodegradable polymer microparticles , J. Biomed. Mater. Res ., 
50 , 440 – 451 . 
26. Fu , K. , Griebenow , K. , Hsieh , L. , Klibanov , A. M. , and Langer , R. ( 1999 ), FTIR characterization 
of the secondary structure of proteins encapsulated within PLGA microspheres 
, J. Controlled Release , 58 , 357 – 366 . 
27. Bezemer , J. M. , Radersma , R. , Grijpma , D. W. , Dijkstra , P. J. , van Blitterswijk , C. A. , and 
Feijen , J. ( 2000 ), Microspheres for protein delivery prepared from amphiphilic multiblock 
copolymers. I. Infl uence of preparation techniques on particle characteristics and 
protein delivery , J. Controlled Release , 67 , 233 – 248 . 
28. Rosas , J. E. , Pedraz , J. L. , Hernandez , R. M. , Gascon , A. R. , Igartua , M. , Guzman , F. , 
Rodrignuez , R. , Cortes , J. , and Patarroyo , M. E. ( 2002 ), Remarkably high antibody levels 
and protection against P. falciparum malaria in Aotus monkey after a single immunization 
of SPf66 encapsulated in PLGA microspheres , Vaccine , 20 , 1707 – 1710 . 
29. Boehm , G. , Peyre , M. , Sesardie , D. , Huskisson , R. J. , Mawas , F. , Douglas , A. , Xing , D. , 
Merkle , H. P. , Gander , B. , and Johansen , P. ( 2002 ), On technological and immunological 
benefi ts of multivalent single - injection microspheres vaccines . Pharm. Res ., 19 , 
1330 – 1336 . 
30. Sturesson , C. , Artursson , P. , Ghaderi , R. , Johansen , K. , Mirazimi , A. , Uhnoo , I. , Svensson , 
L. , Alberstsson , A. C. , and Carlfors , J. ( 1999 ), Encapsulation of rotavirus into poly(lactide - 
co - glycolide) microspheres , J. Controlled Release , 59 , 377 – 389 . 

REFERENCES 429 
31. Morlock , M. , Kissel , T. , Li , Y. X. , Koll , H. , and Winter , G. ( 1998 ), Erythropoietin loaded 
microspheres prepared from biodegradable LPLG - PEO - LPLG triblock copolymers: 
Protein stabilization and in - vitro release properties , J. Controlled Release , 56 , 105 – 115 . 
32. Zambaux , M. F. , Bonneaux , F. , Gref , R. , Dellacherie , E. , and Vigneron , C. ( 1999 ), Preparation 
and characterization of protein C loaded PLA nanoparticles , J. Controlled Release , 
60 , 179 – 188 . 
33. Cho , S. W. , Song , S. H. , and Choi , Y. W. ( 2000 ), Effects of solvent selection and fabrication 
on the characteristics of biodegradable poly(lactide -co - glycolide) microspheres containing 
ovalbumin , Arch. Pharm. Res ., 23 , 385 – 390 . 
34. Li , X. , Deng , X. , and Huang , Z. ( 2001 ), In vitro protein release and degradation of poly - 
dl - lactide - poly(ethylene glycol) microspheres with entrapped human serum albumin: 
Quantitative evaluation of the factors involved in protein release phase , Pharm. Res ., 
18 , 117 – 124 . 
35. Carrasquillo , K. G. , Stanley , A. M. , Aponte - Carro , J. C. , DeJesus , P. , Costantino , H. R. , 
Bosques , C. J. , and Griebenow , K. ( 2001 ), Non - aqueous encapsulation of excipient stabilized 
spray freeze dried BSA into poly(lactide -co - glycolide) microspheres results in 
release of native protein , J. Controlled Release , 76 , 199 – 208 . 
36. Lee , V. H. L. ( 1986 ), Peptide and protein drug delivery: Opportunities and challenges , 
Pharm. Int ., 7 , 208 – 212 . 
37. Johnson , O. L. , Cleland , J. L. , Lee , H. J. , Charnis , M. , Duenas , E. , Jaworowicz , W. , Shepard , 
D. , Shihzamani, A. , Jones , A. J. S. , and Putney , S. D. (1996), A month-long effect from a 
single injection of microencapsulated human growth hormone , Nat. Med ., 2 , 795 – 799 . 
38. Costantino , H. R. , Firouzabadian , L. , Hogeland , K. , Wu , C. , Beganski , C. , Carrasquillo , 
K. G. , Cordova , M. , Griebenow , K. , Zale , S. E. , and Tracy , M. A. ( 2000 ), Protein spray - 
freeze drying: Effect of atomization conditions on particle size and stability , Pharm. Res ., 
17 , 1374 – 1383 . 
39. Bustami , R. T. , Chan , H. K. , Dehghani , F. , and Foster , N. R. ( 2000 ), Generation of microparticles 
of proteins for aerosol delivery using high pressure modifi ed carbon dioxide , 
Pharm. Res ., 17 , 1360 – 1366 . 
40. Alder , M. , Unger , M. , and Lee , G. ( 2000 ), Surface composition of spray - dried particles 
of bovine serum albumin/trehalose/surfactant , Pharm. Res ., 17 , 863 – 870 . 
41. Ruiz , J. M. , Tissier , B. , and Benoit , J. P. ( 1989 ), Microencapsulation of peptides: A study 
of the phase separation of poly( d , l - lactic acid -co - glycolic acid) copolymers 50/50 by 
silicone oil , Int. J. Pharm ., 49 , 69 – 77 . 
42. Redding , T. W. , Schally , A. V. , Tice , T. R. , and Meyers , W. E. ( 1984 ), Long acting delivery 
systems for peptides: Inhibition of rat prostate tumors by controlled release of DTrp6 
leutinizing hormone releasing hormone injectable microcapsules , Proc. Nat. Acad. Sci. 
USA , 81 , 5845 . 
43. Heron , I. , Thomas , F. , Dero , M. , Poutrain , J. R. , Henane , S. , Catus , F. , and Kuhn , J. M. 
( 1993 ), Traitement de l ’ acromegalie par une forme a liberation prolongee du lanreotide, 
un nouvel analogue de la somatostatine , Presse Med ., 22 , 526 – 531 . 
44. O ’ Donnell , P. B. , and McGinity , J. W. ( 1997 ), Preparation of microspheres by the solvent 
evaporation technique , Adv. Drug Deliv. Rev ., 28 , 25 – 42 . 
45. Cleland , J. L. ( 1998 ), Solvent evaporation processes for the production of controlled 
release biodegradable microsphere formulations for therapeutics and vaccines , Biotechnol. 
Prog ., 14 , 102 – 107 . 
46. Ogawa , Y. , Yamamoto , M. , Takada , S. , and Shimamoto , T. ( 1988 ), Controlled release of 
leuprolide acetate from polylactic acid or copolymer ratio of polymer , Chem. Pharm. 
Bull ., 36 , 1502 – 1507 . 

430 BIODEGRADABLE POLYMER-BASED MICROSPHERES 
47. Ogawa , Y. , Yamamoto , M. , Okada , H. , Yashiki , T. , and Shimamoto , T. ( 1988 ), A new 
technique to effi ciently entrap leuprolide acetate into microcapsules of polylactic acid 
or copoly(lactic/glycolic) acid , Chem. Pharm. Bull ., 36 , 1095 – 1103 . 
48. Bittner , B. , Morlock , M. , Koll , H. , Winter , G. , and Kissel , T. ( 1998 ), Recombinant human 
erythropoietin (rHEPO) loaded poly(lactide -co - glycolide) microspheres: Infl uence of 
the encapsulation technique and polymer purity on microspheres characteristics , Eur. J. 
Pharm. Biopharm ., 45 , 295 – 305 . 
49. Kissel , T. , Brich , Z. , Bantle , S. , Lancranjam , I. , NimmerFall , F. , and Vit , P. ( 1991 ), Parentral 
depot systems as the basis of biodegradable polyesters , J. Controlled Release , 6 , 27 – 34 . 
50. Herrmann , J. , and Bodmeier , R. ( 1998 ), Biodegradable, somatostatin acetate containing 
microspheres prepared by various aqueous and non - aqueous solvent evaporation 
methods . Eur. J. Pharm. Biopharm ., 45 , 75 – 82 . 
51. Singh , M. , Shirley , B. , Bajwa , K. , Samara , E. , Hora , M. , and O ’ Hagan , D. ( 2001 ), Controlled 
release of recombinant insulin - like growth factor from a novel formulation of 
polylactide - coglycolide microparticles , J. Controlled Release , 70 , 21 – 28 . 
52. Viswanathan , N. B. , Thomas , P. A. , Pandit , J. K. , Kulkarni , M. G. , and Mashelkar , R. A. 
( 1999 ), Preparation of non - porous microspheres with high entrapment effi ciency of 
proteins by a (water - in - oil) - in - oil emulsion technique , J. Controlled Release , 58 , 9 – 20 . 
53. Sanchez , A. , Gupta , R. K. , Alonso , M. J. , Siber , G. R. , and Langer , R. ( 1996 ), Pulsed 
controlled - release system for potential use in vaccine delivery , J. Pharm. Sci ., 85 , 
547 – 552 . 
54. Freitas , S. , Walz , A. , Merkle , H. P. , and Gander , B. ( 2003 ), Solvent extraction employing 
a static micromixer: A simple, robust and versatile technology for the microencapsulation 
of proteins , J. Microencapsul ., 20 , 67 – 85 . 
55. Khan , M. A. , Healy , M. S. , and Bernstein , H. ( 1992 ), Low temperature fabrication of 
protein loaded microspheres , Proc. Int. Symp. Controlled Release Bioactive Mater ., 19 , 
518 – 519 . 
56. Herberger , J. D. , Wu , C. , Dong , N. , and Tracy , M. A. ( 1996 ), Characterization of Prolease ® 
human growth hormone PLGA microspheres produced using different solvents , Proc. 
Int. Symp. Controlled Release Bioactive Mater ., 23 , 835 – 836 . 
57. Johnson , O. L. , Jaworowicz , W. , Cleland , J. L. , Bailey , L. , Charnis , M. , Duenas , E. , Wu , C. , 
Shepard, D. , Magil, S. , Last, T. , Jones , A. J. S. , and Putney , S. D. (1997), The stabilization 
and encapsulation of human growth hormone into biodegradable microspheres , Pharm. 
Res ., 14 , 730 – 735 . 
58. Gombotz , W. R. , Healy , M. , Brown , L. R. , and Auer , H. E. ( 1990 ), Process for producing 
small particles of biologically active molecules, Pabst Patrea, Patent No. WO 90/13285 . 
59. Gombotz , W. R. , Healy , M. S. , and Brown , L. R. ( 1989 ), Very low temperature casting of 
controlled release microspheres, Enzytech Inc., Application No. 89 - 346143, U.S. Patent 
5,019,400 . 
60. Costantino , H. R. , Johnson , O. L. , and Zale , S. F. ( 2004 ), Relationship between encapsulated 
drug particle size and initial release of recombinant human growth hormone from 
biodegradable microspheres , J. Pharm. Sci ., 93 , 2624 – 2634 . 
61. Cleland , J. L. , and Jones , A. J. S. ( 1996 ), Stable formulations of recombinant human 
growth hormone and interferon - g for microencapsulation in biodegradable microspheres 
, Pharm. Res ., 13 , 1464 – 1475 . 
62. Cleland , J. L. , Duenas , E. T. , Park , A. , Daugherty , A. , Kahn , J. , Kowalski , J. , and 
Cuthbertson , A. ( 2000 ), Development of poly - (d , l - lactide - co - glycolide) microsphere 
formulations containing recombinant human vascular growth factor to promote local 
angiogenesis , J. Controlled Release , 72 , 13 – 24 . 

REFERENCES 431 
63. Lam , X. M. , Duenas , E. T. , Daugherty , A. L. , Levin , N. , and Cleland , J. L. ( 2000 ), Sustained 
release of recombinant human insulin - like growth factor - I for treatment of diabetes . 
J. Controlled Release , 67 , 281 – 292 . 
64. Ribeiro Dos Santos , I. , Richard , J. , Pech , B. , Thies , C. , and Benoit , J. P. ( 2002 ), Microencapsulation 
of protein particles within lipids using a novel supercritical fl uid process , 
Int. J. Pharm ., 242 , 69 – 78 . 
65. Debenedetti , P. G. , Tom , J. W. , Yeo , S. , Lim , G. B. ( 1993 ), Application of supercritical 
fl uids for the production of sustained delivery devices , J. Controlled Release , 24 , 
27 – 44 . 
66. Randolph , T. W. , RAndolph , A. D. , Mebes , M. , and Yeung , S. ( 1993 ), Submicrometer - 
sized biodegradable particles of poly( l - lactic acid) via the gas antisolvent spray precipitation 
process , Biotechnol. Prog ., 9 , 429 – 435 . 
67. Wa . nus , W. , Bleich , J. , and M u ller , B. W. ( 1991 ), Microparticle production by using 
supercritical gases , Clin. Pharmacol ., 13 , 367 . 
68. Bleich , J. , Mueller , B. W. , and Wassmus , W. ( 1993 ), Aerosol solvent extraction system. A 
new microparticle production technique , Int. J. Pharm ., 97 , 111 – 117 . 
69. Winters , M. A. , Debenedetti , P. G. , Carey , J. , Sparks , H. G. , Sane , S. U. , and Przybycien , 
T. M. ( 1997 ), Long - term and high - temperature storage of supercritically - processed 
microparticulate protein powders , Pharm. Res ., 14 , 1370 – 1378 . 
70. Bodmeier , R. , Wang , H. , Dixon , D. J. , Mawson , S. , and Johnston , K. P. ( 1995 ), Polymeric 
microspheres prepared by spraying into compressed carbon dioxide , Pharm. Res ., 12 , 
1211 – 1217 . 
71. Witschi , C. , and Doelker , E. ( 1998 ), Peptide degradation during preparation and in vitro 
release testing of poly( l - lactic acid) and poly( dl - lactic - co - glycolic acid) microparticles , 
Int. J. Pharm ., 171 , 1 – 18 . 
72. Morlock , M. , Koll , H. , Winter , G. , and Kissel , T. ( 1997 ), Microencapsulation of rh - 
erythropoietin using biodegradable poly( d , l - lactide - co - glycolide): Protein stability and 
the effects of stabilizing excipients , Eur. J. Pharm. Biopharm ., 43 , 29 – 36 . 
73. Bittner , B. , and Kissel , T. ( 1999 ), Ultrasonic atomization for spray drying: A versatile 
technique for the preparation of protein loaded bio - degradable microspheres , J. Microencapsul 
., 16 , 325 – 341 . 
74. Breitenbach , A. , Mohr , D. , Kissel , T. ( 2000 ), Biodegradable semicrystalline comb polymers 
infl uence the microsphere production by means of a supercritical fl uid extraction 
technique , J. Controlled Release , 63 , 53 – 68 . 
75. Engwicht , A. , Girreser , U. , and Muller , B. W. ( 1999 ), Critical properties of lactide -co - 
glycolide polymers for the use of microparticle preparation by the aerosol solvent 
extraction system , Int. J. Pharm ., 185 , 61 – 72 . 
76. Felder , C. B. , Blanco - Pr i eto , M. J. , Heizmann , J. , Merkle , H. B. , and Gander , B. ( 2003 ), 
Ultrasonic atomization and subsequent polymer desolvation for peptide and protein 
microencapsulation into biodegradable polyesters , J. Microencapsul ., 20 , 553 – 567 . 
77. Freitas , S. , Merkle , H. P. , and Gander , B. ( 2004 ), Ultrasonic atomisation into reduced 
pressure atmosphere — envisaging aseptic spray drying for microencapsulation , J. Controlled 
Release , 95 , 185 – 195 . 
78. Berkland , C. , Kim , K. , and Pack , D. W. ( 2001 ), Fabrication of PLG microspheres with 
precisely controlled and monodispersed size distributions , J. Controlled Release , 73 , 
59 – 74 . 
79. Jain , R. A. , Rhodes , C. T. , Railkar , A. M. , Malik , A. W. , and Shah , N. H. ( 2000 ), Controlled 
release of drugs from injectable in situ formed biodegradable PLGA microspheres: 
Effect of various formulation variables , Eur. J. Pharm. Biopharm ., 50 , 257 – 262 . 

432 BIODEGRADABLE POLYMER-BASED MICROSPHERES 
80. Kim , H. K. , Chung , H. J. , and Park , T. G. ( 2006 ), Biodegradable polymeric microspheres 
with “ open/closed ” pores for sustained release of human growth hormone , J. Controlled 
Release , 112 , 167 – 174 . 
81. Singh , M. , Kazzaz , J. , Ugozzoli , M. , Chesko , J. , and O ’ Hagan , D. ( 2004 ), Charged 
poly(lactide -co - glycolide) microparticles as novel antigen delivery systems , Expert Opin. 
Biolog. Ther ., 4 , 483 – 491 . 
82. Jilek , S. , Merkle , H. P. , and Walter , E. (2005), DNA-loaded biodegradable microparticles 
as vaccine delivery systems and their interaction with dendritic cells , Adv. Drug Deliv. 
Rev ., 57 , 377 – 390 . 
83. Jabbal - Gill , I. , Lin , W. , Jenkins , P. , Watts , P. , Jimenez , M. , Illum , L. , Davis , S. S. , Wood , J. 
M. , Major , D. , Minor , P. D. , Li , X. W. , Lavelle , E. C. , and Coombes , A. G. A. ( 1999 ), 
Potential of polymeric lamellar substrate particles (PLSP) as adjuvants for vaccines , 
Vaccine , 18 , 238 – 250 . 
84. Jabbal - Gill , I. , Lin , W. , Kistner , O. , Davis , S. S. , and Illum , L. ( 2001 ), Polymeric lamellar 
substrate particles for intranasal vaccination , Adv. Drug Deliv. Rev ., 51 , 97 – 111 . 
85. Venkataprasad , N. , Coombes , A. G. A. , Singh , M. , Rohde , M. , Wilkinson , K. , Hudecz , F. , 
Davis , S. S. , and Vordermeier , H. M. ( 1999 ), Induction of cellular immunity to a mycobacterial 
antigen adsorbed on lamellar particles of lactide polymers , Vaccine , 17 , 
1814 – 1819 . 
86. Kazzaz , J. , Neidleman , J. , Singh , M. , Ott , G. , and O ’ Hagan , D. T. ( 2000 ), Novel anionic 
microparticles are a potent adjuvant for the induction of cytotoxic T lymphocytes against 
recombinant p55 gag from HIV - 1 , J. Controlled Release , 67 , 347 – 356 . 
87. Otten , G. , Schaefer , M. , Greer , C. , Calderon - Cacia , M. , Coit , D. , Kazzaz , J. , Medina - Selby , 
A. , Selby , M. , Singh , M. , Ugozzoli , M. , zur Megede , J. , Barnett , S. W. , O ’ Hagan , D. , Donnelly 
, J. , and Ulmer , J. ( 2003 ), Induction of broad and potent antihuman immunodefi - 
ciency virus immune responses in rhesus macaques by priming with a DNA vaccine and 
boosting with protein - adsorbed polylactide coglycolide microparticles , J. Virol ., 77 , 
6087 – 6092 . 
88. Kang , F. , Jiang , G. , Hinderliter , A. , DeLuca , P. P. , and Singh , J. ( 2002 ), Lysozyme stability 
in primary emulsion for PLGA microsphere preparation: Effect of recovery methods 
and stabilizing excipients , Pharm. Res ., 19 , 629 – 633 . 
89. Sanchez , A. , Villamayor , B. , Guo , Y. , McIver , J. , and Alonso , M. J. ( 1999 ), Formulation 
strategies for the stabilization of tetanus toxoid in poly(lactide - co - glycolide) microspheres 
, Int. J. Pharm ., 185 , 255 – 266 . 
90. Sharif , S. , and O ’ Hagan , D. T. ( 1995 ), A comparison of alternative methods for determination 
of the levels of proteins entrapped in poly(lactide - co - glycolide) microparticles , 
Int. J. Pharm ., 115 , 259 – 263 . 
91. Gupta , R. K. , Chang , A. C. , Griffi n , P. , Rivera , R. , Guo , Y. Y. , and Siber , G. R. ( 1997 ), 
Determination of protein loading in biodegradable polymer microspheres containing 
tetanus toxoid , Vaccine , 15 , 672 – 678 . 
92. Lu , W. , and Park , T. G. ( 1995 ), Protein release from poly(lactic -co - glycolic acid) microspheres: 
Protein stability problems , PDA J. Pharm. Sci. Technol ., 49 , 13 – 19 . 
93. Park, T. G. , Lu, W. , and Crotts , G. (1995), Importance of in vitro experimental conditions 
on protein release kinetics, stability and polymer degradation in protein encapsulated 
poly(d , l - lactic acid -co - glycolic acid) microspheres , J. Controlled Release , 33 , 211 – 222 . 
94. Bilati , U. , Pasquarello , C. , Corthals , C. L. , Hochstrasser , D. F. , All e mann , E. , and Doelker , 
E. ( 2005 ), Matrix - assisted laser desorption/ionization time - of - fl ight mass spectrometry 
for quantitation and molecular stability assessment of insulin entrapped within PLGA 
nanoparticles , J. Pharm. Sci ., 94 , 1 – 7 . 

REFERENCES 433 
95. van de Weert , M. , Van ’ t Hof , R. , Van der Weerd , J. , Heeren , R. M. , Posthuma , G. , 
Hennink , W.E. , and Crommelin , D. J. ( 2000 ), Lysozyme distribution and conformation 
in a biodegradable polymer matrix as determined by FTIR techniques , J. Controlled 
Release , 68 , 31 – 40 . 
96. Blanco , M. D. , and Alonso , M. J. ( 1997 ), Development and characterization of protein - 
loaded poly(lactide -co - glycolide) nanospheres , Eur. J. Pharm. Biopharm ., 43 , 287 – 294 . 
97. Johansen , P. , Tamber , H. , Merkle , H. P. , and Gander , B. ( 1999 ), Diphtheria and tetanus 
toxoid microencapsulation into conventional and end - group alkylated PLA/PLGAs , 
Eur. J. Pharm. Biopharm ., 47 , 193 – 201 . 
98. van de Weert , M. , Hoechstetter , J. , Hennink , W. E. , and Crommelin , D. J. ( 2000 ), The 
effect of a water/organic solvent interface on the structural stability of lysozyme , 
J. Controlled Release , 68 , 351 – 359 . 
99. Yang , T. H. , Dong , A. , Meyer , J. , Johnson, O. L. , Cleland, J. L. , and Carpenter , J. F. (1999), 
Use of infrared spectroscopy to assess secondary structure of human growth hormone 
within biodegradable microspheres , J. Pharm. Sci ., 88 , 161 – 165 . 
100. Perez , C. , De Jesus , P. , and Griebenow , K. ( 2002 ), Preservation of lysozyme structure and 
function upon encapsulation and release from poly(lactic -co - glycolic) acid microspheres 
prepared by the water - in - oil - in - water method , Int. J. Pharm ., 248 , 193 – 206 . 
101. Jorgensen , L. , Vermehren , C. , Bjerregaard , S. , and Froekjaer , S. ( 2003 ), Secondary structure 
alterations in insulin and growth hormone water - in - oil emulsions , Int. J. Pharm ., 
254 , 7 – 10 . 
102. Na , D. H. , Youn , Y. S. , Lee , S. D. , Son , M. W. , Kim , W. B. , DeLuca , P. P. , and Lee , K. C. 
( 2003 ), Monitoring of peptide acylation inside degrading PLGA microspheres by capillary 
electrophoresis and MALDI - TOF mass spectrometry , J. Controlled Release , 92 , 
291 – 299 . 
103. Gilg , D. , Riedl , B. , Zier , A. , and Zimmermann , M. F. ( 1996 ), Analytical methods for the 
characterization and quality control of pharmaceutical peptides and proteins , Pharm. 
Acta Helv ., 71 , 383 – 394 . 
104. Brass , J. M. , Krummen , K. , and Moll - Kaufmann , C. ( 1996 ), Quality assurance after 
process changes of the production of a therapeutic antibody , Pharm. Acta Helv ., 71 , 
395 – 403 . 
105. Sesardic , D. , and Dobbelaer , R. ( 2004 ), European union regulatory developments for 
new vaccine adjuvants and delivery systems , Vaccine , 22 , 2452 – 2456 . 
106. Burgess , D. J. , Crommelin , D. J. A. , Hussain , A. S. , and Chen , M. L. ( 2004 ), Assuring 
quality and performance of sustained and controlled release parenterals: EUFEPS 
workshop report , AAPS PharmSciTech , 6 , 1 – 12 . 
107. Burgess , D. J. , Hussain , A. S. , Ingallinera , T. S. , and Chen , M. L. ( 2002 ), Assuring quality 
and performance of sustained and controlled release parenterals: Workshop report , 
AAPS PharmSciTech , 4 , (article 7). 
108. Gupta , R. K. , Chang , A. C. , and Siber , G. R. ( 1998 ), Biodegradable polymer microspheres 
as vaccine adjuvants and delivery systems , Dev. Biol. Stand ., 92 , 63 – 78 . 
109. Visscher , G. E. , Robison , R. L. , Maulding , H. V. , Fong , J. W. , Pearson , J. E. , and Argentieri , 
G. J. ( 1985 ), Biodegradation of and tissue reaction to 50 : 50 poly( dl - lactide - co - glycolide) 
microcapsules , J. Biomed. Mater. Res ., 19 , 349 – 365 . 
110. Schakenraad , J. M. , Hardonk , M. J. , Feijen , J. , Molenaar , I. , and Nieuwenhuis , P. ( 1990 ), 
Enzymatic activity toward poly( l - lactic) acid implants , J. Biomed. Mater. Res ., 24 , 529 – 
545 . 
111. Johansen , P. , Men , Y. , Merkle , H. P. , and Gander , B. ( 2000 ), Revisiting PLG/PLGA 
microspheres: An analysis of their potential in parenteral vaccination , Eur. J. Pharm. 
Biopharm ., 50 , 129 – 146 . 

434 BIODEGRADABLE POLYMER-BASED MICROSPHERES 
112. Men , Y. , Audran , R. , Thomasin , C. , Eberl , G. , Demotz , S. , Merkle , H. P. , Gander , B. , and 
Corradin , G. ( 1999 ), MHC class I - and class II - restricted processing and presentation of 
microencapsulated antigens , Vaccine , 17 , 1047 – 1056 . 
113. Raychaudhuri , S. , and Rock , K. L. ( 1998 ), Fully mobilizing host defense: Building better 
vaccines , Nat. Biotechnol ., 16 , 1025 – 1031 . 
114. Visscher , G. E. , Robinson , R. L. , and Argentieri , G. J. ( 1987 ), Tissue response t biodegradable 
injectable microcapsules , J. Biomater. Applic ., 2 , 118 – 131 . 
115. Cleland , J. L. , Powell , M. F. , and Shire , S. J. ( 1993 ), The development of stable protein 
formulations: A close look at protein aggregation, deamidation, and oxidation , Crit. Rev. 
Therap. Drug Carrier Syst ., 10 , 307 – 377 . 
116. Gupta , R. K. , Alroy , J. , Alonso , M. J. , Langer , R. , and Siber , G. R. ( 1997 ), Chronic tissue 
reactions, long term immunogenicity and immunological priming of mice and guinea 
pigs to tetanus toxoids encapsulated in biodegradable polymer microspheres composed 
of polylactide -co - glycolide polymers , Vaccine , 15 , 1716 – 1723 . 
117. Jiang , W. , Gupta , R. K. , Deshpande , M. C. , and Schwendeman , S. P. ( 2005 ), Biodegradable 
poly(lactic -co - glycolic acid) microparticles for injectable delivery of vaccine antigens , 
Adv. Drug Deliv. Rev ., 57 , 391 – 410 . 
118. Johnson , O. L. , and Tracy , M. A. ( 1999 ), Peptide and protein drug delivery , in Mathiowitz , 
E. , Ed., Encyclopedia of Controlled Drug Delivery , Vol. 2, Wiley , New York , pp. 
816 – 833 . 
119. Braun , A. , and Alsenz , J. ( 1997 ), Development and use of enzyme - linked immunosorbent 
assays (ELISA) for the detection of protein aggregates in interferon - alpha (IFN - 
alpha) formulations , Pharm. Res ., 14 , 1394 – 1400 . 
120. Hochuli , E. ( 1997 ), Interferon immunogenicity: Technical evaluation of interferon - alpha 
2a , J. Interferon Cytokine Res ., 17 ( Suppl 1 ), 15 – 21 . 
121. Volkin , D. B. , and Middaugh , C. R. ( 1992 ), in: Ahern , T. J. , Manning , M. C. , Eds., Stability 
of Protein Pharmaceuticals Part A: Chemical and Physical Pathways of Protein Degradation 
, Plenum , New York , pp. 109 – 134 . 
122. Manning , M. C. , Patel , K. , and Borchardt , R. T. ( 1989 ), Stability of protein pharmaceuticals 
, Pharm. Res ., 6 , 903 – 918 . 
123. Zale , S. E. , and Klibanov , A. M. ( 1986 ), Why does ribonuclease irreversibly inactivate at 
high temperatures? Biochemistry , 25 , 5432 – 5443 . 
124. Jiang , W. , and Schwendeman , S. P. ( 2000 ), Formaldehyde - mediated aggregation of protein 
antigens: Comparison of untreated and formalinized model antigens , Biotechnol. Bioeng ., 
70 , 507 – 517 . 
125. Jiang , W. , and Schwendeman , S. P. ( 2001 ), Stabilization of a model formalinized protein 
antigen encapsulated in poly(lactide -co - glycolide) - based microspheres , J. Pharm. Sci ., 
90 , 1558 – 1569 . 
126. Zhu , G. , Mallery , S. R. , and Schwendeman , S. P. ( 2000 ), Stabilization of proteins encapsulated 
in injectable poly(lactide -co - glycolide) , Nat. Biotechnol ., 18 , 52 – 57 . 
127. Schwendeman , S. P. , Cardamone , M. , Klibanov , A. , and Langer , R. ( 1996 ), Stability of 
proteins and their delivery from biodegradable polymer microspheres , in Cohen , S. , and 
Bernstein , H. , Eds., Microparticulates Systems for the Delivery of Proteins and Vaccines , 
Marcel Dekker , New York , pp. 1 – 49 . 
128. van de Weert , M. , Hennink , W. E. , and Jiskoot , W. ( 2000 ), Protein instability in poly(lactic - 
co - glycolic acid) microparticles , Pharm. Res ., 17 , 1159 – 1167 . 
129. Schwendeman , S. P. ( 2002 ), Recent advances in the stabilization of proteins encapsulated 
in injectable PLGA delivery systems , Crit. Rev. Therap. Drug Carrier Syst ., 19 , 73 – 
98 . 

REFERENCES 435 
130. Bilati , U. , All e mann , E. , and Doelker , E. ( 2005 ), Strategic approaches for overcoming 
peptide and protein instability within biodegradable nano - and microparticles , Eur. J. 
Pharm. Biopharm ., 59 , 375 – 388 . 
131. Sanders , L. , and Hendren . W. ( 1998 ), Protein Delivery: Physical Systems , Plenum , New 
York . 
132. Senior , J. , and Radomsky , M. ( 2000 ), Sustained - Release Injectable Products , Interpharm , 
Denver . 
133. Akers , M. J. ( 2002 ), Excipient - drug interactions in parenteral formulations , J. Pharm. 
Sci ., 91 , 2283 – 2300 . 
134. Stevenson , C. L. ( 2000 ), Characterization of protein and peptide stability and solubility 
in non - aqueous solvents , Curr. Pharm. Biotechnol ., 1 , 165 – 182 . 
135. Chin , J. T. , Wheeler , S. L. , and Klibanov , A. M. ( 1994 ), On protein solubility in organic 
solvents , Biotechnol. Bioeng ., 44 , 140 – 145 . 
136. Park , T. G. , Lee , H. Y. , and Nam , Y. S. ( 1998 ), A new preparation method for protein 
loaded poly( d , l - lactic - co - glycolic acid) microspheres and protein release mechanism 
study , J. Controlled Release , 55 , 181 – 191 . 
137. Yoo , H. S. , Choi , H. K. , and Park , T.G. ( 2001 ), Protein – fatty acid complex for enhanced 
loading and stability within biodegradable nanoparticles , J. Pharm. Sci ., 90 , 194 – 201 . 
138. Quintanar - Guerrero , D. , All e mann , E. , Fessi , H. , and Doelker , E. ( 1997 ), Applications 
of the ion - pair concept to hydrophilic substances with special emphasis on peptides , 
Pharm. Res ., 14 , 119 – 127 . 
139. Kim , H. K. , and Park , T. G. ( 2001 ), Microencapsulation of dissociable human growth 
hormone aggregates within poly( d , l - lactic - co - glycolic acid) microparticles for sustained 
release , Int. J. Pharm ., 229 , 107 – 116 . 
140. Johnson , M. D. , Hoesterey , B. L. , and Anderson , B. D. ( 1995 ), Solubilization of a tripeptide 
HIV protease inhibitor using a combination of ionization and complexation with 
chemically modifi ed cyclodextrins , J. Pharm. Sci ., 83 , 1142 – 1146 . 
141. Brewster , M. E. , Simpkins , J. W. , Hora , M. S. , Stern , W. C. , and Bodor , N. ( 1989 ), The 
potential use of cyclodextrins in parenteral formulations , J. Parenteral Sci. Technol ., 43 , 
231 – 240 . 
142. Medlicott , N. J. , Foster , K. A. , Audus , K. L. , Gupta , S. , and Stella , V. J. ( 1998 ), Comparison 
of the effects of potential parenteral vehicles for poorly water soluble anticancer drugs 
(organic cosolvents and cyclodextrin solutions) on cultured endothelial cells (HUV - EC) , 
J. Pharm. Sci ., 87 , 1138 – 1143 . 
143. Matsuyama , K. , El - Gizway , S. , and Perrin , J. H. ( 1987 ), Thermodynamics of binding 
of aromatic amino acids to . , . , and . cyclodextrins , Drug Dev. Ind. Pharm ., 13 , 
2687 – 2691 . 
144. Meinel , L. , Illi , O. E. , Zapf , J. , Malfanti , M. , Peter , M. H. , and Gander , B. ( 2001 ), Stabilizing 
insulin - like growth factor - I in poly( d , l - lactide - co - glycolide) microspheres , J. Controlled 
Release , 70 , 193 – 202 . 
145. Uchida , T. , Shiosaki , K. , Nakada , Y. , Fukada , K. , Eda , Y. , Tokiyoshi , S. , Nagareya , N. , and 
Matsuyama , K. ( 1998 ), Microencapsulation of hepatitis B core antigen for vaccine preparation 
, Pharm. Res ., 15 , 1708 – 1713 . 
146. Youan , B. B. C. , Gillard , J. , and Rollmann , B. ( 1999 ), Protein - loaded poly( . - caprolactone) 
microparticles. III. Entrapment of superoxide dismutase by the (water - in - oil) - in water 
solvent evaporation method , STP Pharma Sci ., 9 , 175 – 181 . 
147. Johansen , P. , Men , Y. , Audran , R. , Corradin , G. , Merkle , H. P. , and Gander , B. ( 1998 ), 
Improving stability and release kinetics of microencapsulated tetanus toxoid by co - 
encapsulation of additives , Pharm. Res ., 15 , 1103 – 1110 . 

436 BIODEGRADABLE POLYMER-BASED MICROSPHERES 
148. Krielgaard , L. , Jones , L. S. , Randolph , T. W. , Frokjaer , S. , Flink , J. M. , Manning , M. C. , 
Carpenter , J. F. ( 1998 ), Effect of Tween 20 on freeze - thawing - and agitation - induced 
aggregation of recombinant human factor XIII , J. Pharm Sci ., 87 , 1593 – 1603 . 
149. Krielgaard , L. , Frokjaer , S. , Flink , J .M. , Randolph , T. W. , and Carpenter , J. F. ( 1999 ), 
Effects of additives on the stability of Humicola lanuginosa lipase during freeze - drying 
and storage in the dried solid , J. Pharm. Sci ., 88 , 281 – 290 . 
150. De Rosa , G. , Iommelli , R. , La Rotonda , M. I. , Miro , A. , and Quaglia , F. ( 2000 ), Infl uence 
of the co - encapsulation of different non - ionic surfactants on the properties of PLGA 
insulin - loaded microspheres , J. Controlled Release , 69 , 283 – 295 . 
151. Chang , B. S. , Kendrick , B. S. , and Carpenter , J. F. ( 1996 ), Surface - induced denaturation 
of proteins during freezing and its inhibition by surfactants , J. Pharm. Sci ., 85 , 1325 – 
1330 . 
152. Kerwin, B. A. , Heller , M. C. , Levin, S. H. , and Randolph, T. W. (1998), Effects of Tween 
80 and sucrose on the acute short term stability and long term stability storage at . 20 
degrees of a recombinant hemoglobin , J. Pharm. Sci ., 87 , 1062 – 1068 . 
153. Knepp , V. M. , Whatley , J. L. , Muchnik , A. , and Calderwood , T. S. ( 1996 ), Identifi cation of 
antioxidants for prevention of peroxide - mediated oxidation of recombinant human 
ciliary neurotrophic factor and recombinant human nerve growth factor , J. Parenteral 
Sci. Technol ., 50 , 163 – 171 . 
154. Nace , V. ( 1996 ), Nonionic Surfactants: Polyoxyalkylene Block Copolymers , Marcel 
Dekker , New York . 
155. Kabanov , A. , Batrakova , E. , and Alakhov , V. ( 2002 ), Pluronic block copolymers as novel 
polymer therapeutics for drug and gene delivery , J. Controlled Release , 82 , 189 – 212 . 
156. U.S. Food and Drug Administration ( 2003, Nov. 7 ), Inactive ingredient search for 
approved drug products, “ poloxamer, ” available http:/www.accessdata.fda.gov/scripts/ 
cder/iig/index.cfm , accessed May 21, 2006. 
157. Johnston , T. P. , and Miller , S. C. ( 1985 ), Toxicological evaluation of poloxamer vehicles 
for intramuscular use , J. Parenteral Sci. Technol ., 39 , 83 – 88 . 
158. Sanchez , A. , Tobio , M. , Gonzalez , L. , Fabra , A. , and Alonso , M. J. ( 2003 ), Biodegradable 
micro - and nanoparticles as long - term delivery vehicles for interferon -. , Eur. J. Pharm. 
Sci ., 18 , 221 – 229 . 
159. Lam , X. M. , Duenas , E. T. , and Cleland , J. L. ( 2001 ), Encapsulation and stabilization of 
nerve growth factor into poly(lactic - co - glycolic) acid microspheres , J. Pharm. Sci ., 90 , 
1356 – 1365 . 
160. Nihant , N. , Schugens , C. , Grandfi ls , C. , Jer o me , R. , and Teyssie , P. ( 1994 ), Polylactide 
microparticles prepared by double emulsion/evaporation technique: I. Effect of primary 
emulsion stability , Pharm. Res ., 11 , 1479 – 1484 . 
161. Sturesson , C. , and Carlfors , J. ( 2000 ), Incorporation of protein in PLG - microspheres with 
retention of bioactivity , J. Controlled Release , 67 , 171 – 178 . 
162. Chen , L. , Apte , R. N. , and Cohen , S. ( 1997 ), Characterization of PLGA microspheres for 
the controlled delivery of IL - 1a for tumor immunotherapy , J. Controlled Release , 43 , 
261 – 272 . 
163. Kwon , Y. M. , Baudys , M. , Knutson , K. , and Kim , S. W. ( 2001 ), In situ study of insulin 
aggregation induced by water – organic solvent interface , Pharm. Res ., 18 , 1754 – 1759 . 
164. Costantino , H. R. , Langer , R. , and Klibanov , A. ( 1995 ), Aggregation of a lyophilized 
pharmaceutical protein, recombinant human albumin: effect of moisture and stabilization 
excipients , BioTechnology , 13 , 493 – 496 . 
165. Costantino , H. R. , Langer , R. , and Klibanov , A. M. ( 1994 ), Solid - phase aggregation of 
proteins under pharmaceutically relevant conditions , J. Pharm. Sci ., 83 , 1662 – 1669 . 

REFERENCES 437 
166. Means , G. E. , and Feeney , R. E. ( 1971 ), Chemical Modifi cation of Proteins , Holden - Day , 
San Francisco . 
167. Schwendeman , S. P. , Costantino , H. R. , Gupta , R. K. , Siber , G. R. , Klibanov , A. M. , and 
Langer , R. ( 1995 ), Stabilization of tetanus and diphtheria toxoids against moisture - 
induced aggregation , Proc. Nat. Acad. Sci. USA , 92 , 11234 – 11238 . 
168. Costantino , H. R. , Schwendeman , S. P. , Griebenow , K. , Klibanov , A. M. , and Langer , R. 
( 1996 ), The secondary structure and aggregation of lyophilized tetanus toxoid , J Pharm. 
Sci ., 85 , 1290 – 1293 . 
169. Hageman , M. J. ( 1988 ), The role of moisture in protein stability , Drug Dev. Ind. Pharm ., 
14 , 2047 – 2070 . 
170. Lee , H. J. , Riley , G. , Johnson , O. , Cleland , J. L. , Kim , N. , and Charnis , M. ( 1997 ), In vivo 
characterization of sustained release formulations of human growth hormone , J. Pharmacol. 
Exp. Ther ., 281 , 1431 – 1439 . 
171. Herbert , P. , Murphy , K. , Johnson , O. , Dong , N. , Jaworowicz , W. , and Tracy , M. A. ( 1998 ), A 
large - scale process to produce microencapsulated proteins , Pharm. Res ., 15 , 357 – 361 . 
172. Tracy , M. A. , Bernstein , H. , and Kahn , M. A. ( 2000 ), Controlled release of metal cation - 
stabilized interferon, U.S. Patent 6,165,508 . 
173. Lam , X. M. , Duenas , E. T. , and Cleland , J. L. ( 1998 ), Stabilization of nerve growth factor 
during microencapsulation and release from microspheres [abstract] , Proc. Int. Symp. 
Controlled Release Bioactive Mater ., 25 , 491 . 
174. Zale , S. E. , Burke , P. A. , Bernstein , H. , and Brickner , A. ( 1997 ), Composition for sustained 
release of non - aggregated erythropoietin, U.S. Patent 5,674,534 . 
175. Johnson , O. L. ( 2000 ), Stabilization of proteins in solutions by lyophilization , in Wise , D. 
E. , Handbook of Pharmaceutical Controlled Release Technology , Marcel Dekker , New 
York , pp. 693 – 749 . 
176. Carpenter , J. F. , and Chang , B. S. ( 1996 ), Lyophilization of protein pharmaceuticals , in 
Avis , K. E. , and Wu , V. L. , Eds., Biotechnology and Biopharmaceutical Manufacturing, 
Processing, and Preservation , Interpharm , Buffalo Grove, IL , pp. 199 – 264 . 
177. Carpenter , J. F. , and Crowe , J. H. ( 1988 ), The mechanism of cryoprotection of proteins 
by solutes , Cryobiology , 25 , 244 – 255 . 
178. Arakawa , T. , Prestrelski , S. , Kinney , W. , and Carpenter , J. F. ( 1993 ), Factors affecting 
short - term and long - term stabilities of proteins , Adv. Drug Deliv. Rew ., 10 , 1 – 28 . 
179. Franks , F. ( 1990 ), Freeze drying: From empiricism to predictability , Cryoletters 11 , 
93 – 110 . 
180. Prestrelski , S. J. , Tedeschi , N. , Arakawa , T. , and Carpenter , J. F. ( 1993 ), Dehydration - 
induced conformational changes in proteins and their inhibition by stabilizers , Biophys. 
J ., 65 , 661 – 671 . 
181. Tabata, Y ., Gutta , S. , and Langer , R. ( 1993 ), Controlled delivery systems for proteins 
using polyanhydride microspheres , Pharm. Res ., 10 , 487 – 496 . 
182. P e an , J. M. , Boury , F. , Venier - Julienne , M. C. , Menei , P. , Proust , J. E. , and Benoit , J. P. 
( 1999 ), Why does PEG 400 co - encapsulation improve NGF stability and release from 
PLGA biodegradable microspheres? Pharm. Res ., 16 , 1294 – 1299 . 
183. Perez - Rodriguez , C. , Montano , N. , Gonzalez , K. , and Griebenow , K. ( 2003 ), Stabilization 
of a - chymotrypsin at the CH 2 Cl 2 /water interface and upon water - in - oil - in - water encapsulation 
in PLGA microspheres , J. Controlled Release , 89 , 71 – 85 . 
184. Izutsu , K. , Yoshioka , S. , and Teroa , T. ( 1993 ), Decreased protein - stabilizing effects of 
cryoprotectants due to crystallization , Pharm. Res ., 10 , 1232 – 1237 . 
185. Izutsu , K. , Yoshioka , S. , and Teroa , T. ( 1994 ), Effect of mannitol crystallinity on the stabilization 
of enzymes during freeze drying , Chem. Pharm. Bull ., 42 , 5 – 8 . 

438 BIODEGRADABLE POLYMER-BASED MICROSPHERES 
186. Fakes , M. G. , Dali , M. V. , Haby , T. A. , Morris , K. R. , Varia , S. A. , and Serajuddin , A. T. M. 
( 2000 ), Moisture sorption behavior of selected bulking agents used in lyophilized products 
, PDA J. Pharm. Sci. Technol ., 54 , 144 – 149 . 
187. Kovalcik , T. R. , and Guillory , J. K. ( 1988 ), The stability of cyclophosphamide in lyophilized 
cakes. Part I. Mannitol, lactose, and sodium bicarbonate as excipients , J. Parenterol 
Sci. Technol ., 43 , 80 – 83 . 
188. Costantino , H. R. , Carrasquillo , K. G. , Cordero , R. A. , Mumenthaler , M. , Hsu , C. C. , and 
Griebenow , K. ( 1998 ), Effect of excipients on the stability and structure of lyophilized 
recombinant human growth hormone , J. Pharm. Sci ., 87 , 1412 – 1420 . 
189. Li , S. , Patapoff , T. W. , Overcashier , D. , Hsu , C. , Nguyen , T. H. , and Borchardt , R. T. ( 1996 ), 
Effects of reducing sugars on the chemical stability of human relaxin in the lyophilized 
state , J. Pharm. Sci ., 85 , 873 – 877 . 
190. Hartauer , K. J. , and Guillory , J. K. ( 1991 ), A comparison of diffuse refl ectance FT - IR 
spectroscopy and DSC in the characterization of a drug - excipient interaction , Drug Dev. 
Ind. Pharm ., 17 , 617 – 630 . 
191. Wang , J. , Wang , B. M. , and Schwendeman , S. P. ( 2004 ), Mechanistic evaluation of the 
glucose - induced reduction in initial burst release of octreotide acetate from poly( d , l - 
lactide - co - glycolide) microspheres , Biomaterials , 25 , 1919 – 1927 . 
192. Dubost , D. C. , Kaufman , M. J. , Zimmerman , J. A. , Bogusky , M. J. , Coddington , A. B. , and 
Pitzenberger , S. M. ( 1996 ), Characterization of a solid state reaction product from a 
lyophilized formulation of a cyclic heptapeptide: A novel example of an excipient - 
induced oxidation , Pharm. Res ., 13 , 1811 – 1814 . 
193. Williams , N. A. , Lee , Y. , Polli , G. P. , and Jennings , T. A. ( 1986 ), The effects of cooling rate 
on solid phase transitions and associated vial breakage occurring in frozen mannitol 
solutions , J. Parenteral Sci. Technol ., 40 , 135 – 141 . 
194. Williams , N. A. , and Dean , T. ( 1991 ), Vial breakage by frozen mannitol solutions: Correlation 
with thermal characteristics and effect of stereoisomerism, additives, and vial 
confi guration , J. Parenteral Sci. Technol ., 45 , 94 – 100 . 
195. Allison , S. D. , Manning , M. C. , Randolph , T. W. , Middleton , K. , Davis , A. , and Carpenter , 
J. F. ( 2000 ), Optimization of storage stability of lyophilized actin using combinations of 
disaccharides and dextran , J. Pharm. Sci ., 89 , 199 – 214 . 
196. Cleland , J. L. , Lam , X. , Kendrick , B. , Yang , J. , Yang , T. , Overcashier , D. , Brooks , D. , Hsu , 
C. , and Carpenter , J. F. ( 2001 ), A specifi c molar ratio of stabilizer to protein is required 
for storage stability of a lyophilized monoclonal antibody , J. Pharm. Sci ., 90 , 310 – 321 . 
197. Lam , X. M. , Patapoff , T. W. , and Nguyen , T. H. ( 1997 ), Effect of benzyl alcohol on stability 
of recombinant human interferon gamma , Pharm. Res ., 14 , 725 – 729 . 
198. Chen , B. L. , and Arakawa , T. ( 1996 ), Stabilization of recombinant human keratinocyte 
growth factor by osmolytes and salts , J. Pharm. Sci ., 85 , 419 – 422 . 
199. Arakawa , T. , Kita , Y. , and Carpenter , J. F. ( 1991 ), Protein – solvent interactions in pharmaceutical 
formulations , Pharm. Res ., 8 , 285 – 291 . 
200. Zhou , Y. , and Hall , C. K. ( 1996 ), Solute excluded - volume effects on the stability of 
globular proteins: A statistical thermodynamic theory , Biopolymers , 38 , 273 – 284 . 
201. Sugimoto , L. , Ishihara , T. , Habata , H. , and Nakagawa , H. ( 1981 ), Stability of lyophilized 
sodium prasterone sulfate , J. Parenteral Sci. Techn ., 35 , 88 – 92 . 
202. Shao , P. G. , and Bailey , L. C. ( 2000 ), Porcine insulin biodegradable polyester microspheres: 
Stability and in vitro release characteristics , Pharm. Dev. Technol ., 5 , 1 – 9 . 
203. M a der , K. , Bittner , B. , Li , Y. , Wohlauf , W. , and Kissel , T. ( 1998 ), Monitoring microviscosity 
and microacidity of the albumin microenvironment inside degrading microparticles 
from poly(lactide - coglycolide) (PLG) or ABA - triblock polymers containing hydropho

REFERENCES 439 
bic poly(lactide -co - glycolide) A blocks and hydrophilic poly(ethyleneoxide) B blocks , 
Pharm. Res ., 15 , 787 – 793 . 
204. Brunner , A. , Mader , K. , and Gopferich , A. ( 1999 ), pH and osmotic pressure inside biodegradable 
microspheres during erosion , Pharm. Res ., 16 , 847 – 853 . 
205. Burke , P. A. ( 1996 ), Determination of internal pH in PLGA microspheres using 31 P 
NMR spectroscopy , Proc. Inter. Symp. Controlled Release Bioactive Mater ., 23 , 133 – 
134 . 
206. Fu , K. , Pack , D. W. , Klibanov , A. M. , and Langer , R. ( 2000 ), Visual evidence of acidic 
environment within degrading poly(lactic -co - glycolic acid) (PLGA) microspheres , 
Pharm. Res ., 17 , 100 – 106 . 
207. Shenderova , A. , Burke , T. G. , and Schwendeman , S. P. ( 1999 ), The acidic microclimate 
in poly(lactide -co - glycolide) microspheres stabilizes camptothecins , Pharm. Res ., 16 , 
241 – 248 . 
208. Li , L. , and Schwendeman , S. P. ( 2005 ), Mapping neutral microclimate pH in PLGA 
microspheres , J. Controlled Release , 101 , 163 – 173 . 
209. Ding , A. G. , and Schwendeman , S. P. ( 2004 ), Determination of water - soluble acid distribution 
in poly(lactide -co - glycolide) , J. Pharm. Sci ., 93 , 322 – 331 . 
210. Cleland , J. L. , Mac , A. , Boyd , B. , Yang , J. , Hsu , C. , Chu , H. , Mukku , V. , and Jones , A. J. S. 
( 1997 ), The stability of recombinant human growth hormone in poly(lactic - co - glycolic 
acid) (PLGA) microspheres , Pharm. Res ., 14 , 420 – 425 . 
211. Cleland , J. L. , Duenas , E. , Daugherty , A. , Marian , M. , Yang , J. , Wilson , M. , Celniker , A. 
C. , Shahzamani , A. , Quarmby , V. , Chu , H. , Mukku , V. , Mac , A. , Roussakis , M. , Gillette , 
N. , Boyd , B. , Yeung , D. , Brooks , D. , Maa , Y. F. , Hsu , C. , and Jones , A. J. S. ( 1997 ), Recombinant 
human growth hormone poly(lactic -co - glycolic acid) (PLGA) microspheres 
provide a long lasting effect , J. Controlled Release , 49 , 193 – 205 . 
212. Schwendeman , S. P. ( 2001 ), Stabilization of vaccine antigens encapsulated in PLGA 
microspheres [abstract 320] , Proc. Int. Symp. Controlled Release Bioactive Mater ., 28 . 
213. Jiang , W. , and Schwendeman , S. P. ( 2001 ), Stabilization and controlled release of bovine 
serum albumin encapsulated in poly( d , l - lactide) and poly(ethylene glycol) microsphere 
blends , Pharm. Res ., 18 , 878 – 885 . 
214. Bittner , B. , Witt , C. , M a der , K. , and Kissel , T. ( 1999 ), Degradation and protein release 
properties of microspheres prepared from biodegradable poly(lactide -co - glycolide) and 
ABA triblock copolymers: Infl uence of buffer media on polymer erosion and bovine 
serum albumin release , J. Controlled Release , 60 , 297 – 309 . 
215. Shenderova , A. , Zhu , G. , and Schwendeman , S. P. ( 2000 ), Correlation of measured microclimate 
pH with the stability of BSA encapsulated in PLGA microspheres abstract 
0413] , Proc. Int. Symp. Controlled Release Bioactive Mater ., 27 . 
216. Shenderova , A. ( 2000 ), The microclimate in poly(lactide -co - glycolide) microspheres and 
its effect on the stability of encapsulated substances, Ph.D thesis, The Ohio State 
University. 
217. Hunter , S. K. , andracki , M. E. , and Kreig , A. M. ( 2001 ), Biodegradable microspheres 
containing group B Streptococcus vaccine: Immune response in mice , Am. J. Obstet. 
Gynecol ., 185 , 1174 – 1179 . 
218. Johansen , P. , Moon , L. , Tamber , H. , Merkle , H. P. , Gander , B. , and Sesardic , D. ( 1999 ), 
Immunogenicity of single - dose diphtheria vaccines based on PLA/PLGA microspheres 
in guinea pigs , Vaccine , 18 , 209 – 215 . 
219. Peyre , M. , Sesardic , D. , Merkle , H. P. , Gander , B. , and Johansen , P. ( 2003 ), An experimental 
divalent vaccine based on biodegradable microspheres induces protective immunity 
against tetanus and diphtheria , J. Pharm. Sci ., 92 , 957 – 966 . 

440 BIODEGRADABLE POLYMER-BASED MICROSPHERES 
220. Peyre , M. , Fleck , R. , Hockley , D. , Gander , B. , and Sesardic , D. ( 2004 ), In vivo uptake of 
an experimental microencapsulated diphtheria vaccine following sub - cutaneous immunisation 
, Vaccine , 22 , 2430 – 2437 . 
221. Namura , J. A. M. , Takata , C. S. , Moroc , A. M. , Politi , M. J. , de Araujo , S. , Cuccovia , I. M. , 
and Bueno da Costa , M. H. ( 2004 ), Lactic acid triggers, in vitro, thiomersal to degrade 
protein in the presence of PLGA microspheres , Int. J. Pharm ., 273 , 1 – 8 . 
222. Tan , M. , and Parkin , J. E. ( 2000 ), Route of decomposition of thiomersal (Thimerosal ® ) , 
Int. J. Pharm ., 208 , 23 – 34 . 
223. Jung , T. , Koneberg , R. , Hungerer , K. D. , and Kissel , T. ( 2002 ), Tetanus toxoid microspheres 
consisting of biodegradable poly(lactide -co - glycolide) - and ABA - triblock - 
copolymers: Immune response in mice , Int. J. Pharm ., 234 , 75 – 90 . 
224. Determan , A. S. , Wilson , J. H. , Kipper , M. J. , Wannemuehler , M. J. , and Narasimhan , B. 
( 2006 ), Protein stability in the presence of polymer degradation products: Consequences 
for controlled release formulations , Biomaterials , 27 , 3312 – 3320 . 
225. Jaganathan , K. S. , Rao , Y. U. B. , Singh , P. , Prabakaran , D. , Gupta , S. , Jain , A. , and Vyas , 
S. P. ( 2005 ), Development of a single dose tetanus toxoid formulation based on polymeric 
microspheres: a comparative study of poly( d , l - lactic - co - glycolic acid) versus chitosan 
microspheres , Int. J. Pharm ., 294 , 23 – 32 . 
226. Kipper , M. J. , Wilson , J. H. , Wannemuehler , M. J. , and Narasimhan , B. ( 2006 ), Single dose 
vaccine based on biodegradable polyanhydride microspheres can modulate immune 
response mechanism , J. Biomed. Mater. Res. Part A , 76 , 798 – 810 . 
227. Katare , Y. K. , Muthukumaran , T. , and Panda , A. K. ( 2005 ), Infl uence of particle size, 
antigen load, dose and additional adjuvant on the immune response from antigen loaded 
PLA microparticles , Int. J. Pharm ., 301 , 149 – 160 . 
228. Yeh , M. K. , Liu , Y. T. , Chen , J. L. , and Chiang , C. H. ( 2002 ), Oral immunogenicity of the 
inactivated Vibrio cholerae whole cell vaccine encapsulated in biodegradable microparticles 
, J. Controlled Release , 82 , 237 – 247 . 
229. Yeh , M. K. , Chen , J. L. , and Chiang , C. H. ( 2002 ), Vibrio cholerae - loaded poly( dl - lactide - 
co - glycolide) microparticles , J. Microencapsul ., 19 , 203 – 212 . 
230. Yeh, M. K. , and Chiang , C. H. (2004), Inactive Vibrio cholerae whole-cell vaccine-loaded 
biodegradable microparticles: In vitro release and oral vaccination , J. Microencapsul ., 
21 , 91 – 106 . 
231. Khang , G. , Cho , J. C. , Lee , J. W. , Rhee , J. M. , and Lee , H. B. ( 1999 ), Preparation and 
characterization of Japanese encephalitis virus vaccine loaded poly( l - lactide - co - 
glycolide) microspheres for oral immunization , Biomed. Mater. Eng ., 9 , 49 – 59 . 
232. Kaur , R. , Rauthan , M. , and Vrati , S. ( 2004 ), Immunogenicity in mice of a cationic 
microparticle - adsorbed plasmid DNA encoding Japanese encephalitis virus envelope 
protein , Vaccine , 22 , 2776 – 2782 . 
233. Yeh , M. K. , Coombes , A. G. A. , Chen , J. L. , and Chiang , C. H. ( 2002 ), Japanese encephalitis 
virus vaccine formulations using PLA lamellar and PLG microparticles , J. Microencapsul 
., 19 , 671 – 682 . 
234. Feng , L. , Qi , X. R. , Zhou , X. J. , Maitani , Y. , Wang , S. C. , Jiang , Y. , and Nagai , T. ( 2006 ), 
Pharmaceutical and immunological evaluation of a single - dose hepatitis B vaccine using 
PLGA microspheres , J. Controlled Release , 112 , 35 – 42 . 
235. Jaganathan , K. S. , Singh , P. , and Prabakaran , D. ( 2004 ), Development of a single - dose 
stabilized poly( d , l - lactide - co - glycolide) microspheres - based vaccine against hepatitis B , 
J. Pharm. Pharmacol ., 56 , 1243 – 1250 . 
236. Shi , L. , Caulfi eld , M. J. , Chern , R. T. , Wilson , R. A. , Sanyal , G. , and Volkin , D. B. ( 2002 ), 
Pharmaceutical and immunological evaluation of a single - shot hepatitis B vaccine formulated 
with PLGA microspheres , J. Pharm. Sci ., 91 , 1020 – 1035 . 

REFERENCES 441 
237. Singh , M. , Li , X. M. , McGee , J. P. , Zamb , T. , Koff , W. , Wang , C. Y. , and O ’ Hagan , D. T. 
( 1997 ), Controlled release microparticles as a single dose hepatitis B vaccine: Evaluation 
of immunogenicity in mice , Vaccine , 15 , 475 – 481 . 
238. Genta , I. , Perugini , P. , Pavanetto , F. , Maculotti , K. , Modena , T. , Casado , B. , Lupi , A. , 
Iadarola , P. , and Conti , B. ( 2001 ), Enzyme loaded biodegradable microspheres in vitro 
ex vivo evaluation , J. Controlled Release , 77 , 287 – 295 . 
239. Perugini , P. , Genta , I. , Pavanetto , F. , Modena , T. , Maculotti , K. , and Conti , B. ( 2002 ), 
Evaluation of enzyme stability during preparation of polylactide -co - glycolide microspheres 
, J. Microencapsul ., 19 , 591 – 602 . 
240. Lupi , A. , Perugini , P. , Genta , I. , Modena , T. , Conti , B. , Casado , B. , Cetta , G. , Pavanetto , 
F. , and Iadarola , P. ( 2004 ), Biodegradable microspheres for prolidase delivery to human 
cultured fi broblasts , J. Pharm. Pharmacol ., 56 , 597 – 603 . 
241. Morimoto , K. , Chono , S. , Kosai, T. , Seki, T. , and Tabata, Y. (2005), Design of novel injectable 
cationic microspheres based on aminated gelatin for prolonged insulin action , 
J. Pharm. Pharmacol ., 57 , 839 – 844 . 
242. Hinds , K. D. , Campbell , K. M. , Holland , K. M. , Lewis , D. H. , Pich e , C. A. , and Schmidt , 
P. G. ( 2005 ), PEGylated insulin in PLGA microparticles. In vivo and in vitro analysis , 
J. Controlled Release , 104 , 447 – 460 . 
243. Shenoy , D. B. , D ’ Souza , R. J. , Tiwari , S. B. , and Udupa , N. ( 2003 ), Potential applications 
of polymeric microsphere suspension as subcutaneous depot for insulin , Drug Dev. Ind. 
Pharm ., 29 , 555 – 563 . 
244. Zhou , S. , Deng , X. , He , S. , Li , X. , Jin , W. , Wei , D. , Zhang , Z. , and Ma , J. ( 2002 ), Study on 
biodegradable microspheres containing recombinant interferon - alpha - 2a , J. Pharm. 
Pharmacol ., 54 , 1287 – 1292 . 
245. Luan , X. , and Bodmeier , R. ( 2006 ), Modifi cation of the tri - phasic drug release pattern 
of leuprolide acetate - loaded poly(lactide - co - glycolide) microparticles , Eur. J. Pharm. 
Biopharm ., 63 , 205 – 214 . 
246. Wang , Y. , Gao , J. Q. , Chen , H. L. , Zheng , C. H. , and Liang , W. Q. ( 2006 ), Pluronic F127 
gel effectively controls the burst release of drug from PLGA microspheres , Die Pharma ., 
61 , 367 – 368 . 


443 
5.3 
LIPOSOMES AND DRUG DELIVERY 
Sophia G. Antimisiaris, 1 Paraskevi Kallinteri, 2 and 
Dimitrios G. Fatouros 3 
1 School of Pharmacy, University of Patras, Rio, Greece 
2 Medway School of Pharmacy, Universities of Greenwich and Kent, England 
3 School of Pharmacy and Biomedical Sciences, Portsmouth, England 
Contents 
5.3.1 Introduction 
5.3.2 Liposome Structure and Characteristics 
5.3.2.1 Phospholipids: Structure Stability and Characterization of Lipid 
Membranes 
5.3.2.2 Physicochemical Properties of Liposomes 
5.3.2.3 Preparation of Liposomes 
5.3.2.4 Functionalization of Liposomes 
5.3.3 In Vivo Distribution 
5.3.3.1 Conventional Liposomes 
5.3.3.2 Long - Circulating or PEGylated Liposomes 
5.3.3.3 Other Routes of Administration 
5.3.4 Applications of Liposomes in Therapeutics 
5.3.4.1 Anticancer Drug Delivery 
References 
5.3.1 INTRODUCTION 
Liposomes are vesicles in which an aqueous volume is entirely surrounded by a 
phospholipid membrane and their size can range between 30 and 50 nm up to 
several micrometers. They can consist of one (unilamellar) or more (multilamellar) 
homocentric bilayers of amphipathic lipids (mainly phospholipids). Based on their 
lamellarity (number of lamellae) — and size — they are characterized as SUVs/LUVs 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

444 LIPOSOMES AND DRUG DELIVERY 
(small or large unilamellar vesicles) or MLVs (multilamellar vesicles). MLV liposomes 
are always large (at least cannot be considered small) and aqueous spaces 
exist in their center and also between their bilayers. 
Liposomes were initially invented by Alec Bangham [1] to serve as a model for 
cell membranes in biophysical studies. In the 1970s they started to be investigated 
as promising drug carriers [2, 3] . The main advantages of liposomes as a drug delivery 
system are the following: (i) They have a very versatile structure which can be 
easily tairored in order to bear the properties needed for each specifi c application. 
(ii) They can accommodate any type of drug molecules either in their aqueous 
compartments (hydrophilic drugs) or in their bilayers (lipophilic drugs) or both 
(amphiphilic drugs). (iii) Last, but not least, they are nontoxic, nonimmunogenic, 
and fully biodegradable. Early attempts to use liposomes as a drug delivery system 
revealed the main limitation of the system and resulted in disappointment due to 
the fast nonspecifc clearance of liposomes from circulation by reticuloendothelial 
system (RES) cells [4] . However, it was realized that recognition by the RES macrophage 
system could be useful in antigen presentation, macrophage activation or 
killing, and elimination of parasitic infections (what is called passive targeting). 
From then on many different types of liposomes or methods to obtain longer circulation 
half - lives have been successfully invented by controlling the physicochemical 
characteristics of lipid bilayers and their interaction with the biological 
environment. Some of the basic methods to control the properties of liposomes are 
presented in Table 1 . Today (Figure 1 ), several different liposome types are available, 
mainly conventional, long circulating (or “ stealth, ” sterically stabilized, or PEGylated), 
targeted [or ligand bearing or immunoliposomes (when antibodies are used as 
targeting ligands)], cationic (for genetic material delivery), and deformable or 
elastic (or “ transferosomes ” ) [will be discussed in the paragraph about of skin 
delivery in Section 5.3.3.3 ]. As a consequence of these advances in liposome technology, 
a very broad range of liposome applications for drug delivery are being explored, 
some of which have resulted in life - saving products on the market or under late - 
stage clinical testing. 
In this chapter we will provide information about the basic characteristics of 
liposomes staring from their building blocks, that is, phospholipids. After this, liposome 
structure, physicochemical properties, and stability, which are most important 
for their in vivo performance, will be discussed as well as methods used for liposome 
preparation, characterization, and stabilization. Following this fi rst part which is 
more technological, we will move into the biological part and talk about the fate of 
conventional liposomes and sterically stabilized liposomes, as well as liposomal 
drugs, after in vivo administration by different routes [mainly intravenous (i.v.), 
intraperitoneal (i.p.), or subcutaneous (s.c.)] and also give some information about 
other possible routes for in vivo administration of liposomes. Finally, specifi c applications 
of liposomes in therapeutics will be presented, some in more detail, mainly 
for the therapy of different types of cancer. 
5.3.2 LIPOSOME STRUCTURE AND CHARACTERISTICS 
The main building blocks of most liposomal drug formulations are phospholipids. 
This chapter will start with an introduction of phospholipid structure and briefl y 

TABLE 1 Methods and Results of Modifying Physicochemical Properties of Liposomes 
Properties Method Result 
Physicochemical properties 
Size distribution Sonication, extrusion, 
microfl uidization 
Control of circulation time, 
Increased extravasation 
Membrane 
permeability 
Lipid composition modifi cation 
(cholesterol, thermotropic 
transitions) 
Increased liposome stability, 
proton or temperature - induced 
sensitivity 
Tendency for 
aggregation 
or fusion 
Lipid composition modifi cation, 
addition of cations 
Formation of structures (as 
cochleates) that can carry 
antigens, deoxyribonucleic acid 
(DNA), vaccine formulations 
Surface 
hydrophilicity 
Steric stabilization by grafting 
hydrophilic molecules on 
liposome surface (linear 
dextrans [5] , sialic acid – 
containing gangliosides [6] , 
lipid derivatives of hydrophilic 
polymers [as PEG [7, 8] , poly - 
N - vinylpyrrolidones [9] , and 
polyvinyl alcohol [10] ) 
Increase of circulation time, 
modifi cation of 
pharmacokinetics and tissue 
dispotition of liposomes and 
encapsulated drugs 
Drug 
encapsulation 
effi ciency 
Active or remote loading [11] Stable liposome encapsulation of 
drugs at high drug - to - lipid 
ratios, Many applications in 
drug delivery 
Elasticity, 
rigidity 
Introduction of detergents or 
edge activators and “ skin 
lipids ” (as ceramides) in 
liposome preparations [12] 
Increased skin penetration and 
retention in skin which results 
in increased transdermal 
absortption of liposomal drugs 
Surface 
properties, 
liposome 
stability, and 
other properties 
Addition of spefi fi c ligand on 
liposome surface, together 
with steric stabilization 
Increased potential for effi cient 
targeting 
describe the most important (for liposomes) physichochemical characteristics of 
lipids and lipid membranes. Afterward properties of liposomes and fi nally methods 
used for the preparation of liposomal drug formulations will be discussed. 
5.3.2.1 Phospholipids: Structure Stability and Characterization 
of Lipid Membranes 
Phospholipids are naturally occurring biomacromolecules that play an important 
role in the physiology of humans as they serve as structural components of biological 
membranes and support organisms with energy [13] . They are amphiphilic molecules 
with poor aqueous solubility and typically consist of two parts: a water - soluble 
group, the so called polar head, and an insoluble one, the backbone (Figure 2 ). The 
polar head group contains hydroxyl groups which are responsible for the surface 
LIPOSOME STRUCTURE AND CHARACTERISTICS 445

446 LIPOSOMES AND DRUG DELIVERY 
charge of the lipids that can be positively or negatively charged, zwitterionic, or 
noncharged. During liposome formation, these molecules arrange themselves by 
exposing their polar parts toward the water phase, while the hydrocarbon moieties 
(hydrophobic) adhere together in the bilayer. Two classes of lipids are mainly used 
for liposome preparation: double - chain polar lipids and sterols (mainly cholesterol). 
Such lipids form bilayers, in contrast to single - chain lipids (e.g., short - chain 
phosphatidylcholine) that form micelles, upon their dispersion in water. [14] . 
Double - chain lipids are either naturally occurring or synthesized in the laboratory. 
They consist of glycerol or sphingosine and a polar head containing a phosphor - 
or glyco - group. Glycerophopsholipids or phospholipids are the most popular, among 
the other lipids, for the preparation of liposomal dispersions [15] . Phosphatidylcho- 
Conventional PEGylated or 
Longcirculating 
Targeted or 
immunoliposomes 
Cationic 
Phospholipid 
PEGlipid 
Cationic lipid 
Helper lipid 
Antibody or 
targeting ligand 
FIGURE 1 Liposome types. Conventional liposomes are composed of phospholipids that 
form bilayers enclosing an aqueous compartment. Cholesterol may be included in the bilayer 
to increase membrane rigidity. Hydrophilic drugs can be encapsulated in the aqueous interior 
of the vesicles and lipophilic drugs can be included in their membranes. Pegylated or long - 
circulating liposomes have a surface coating of polyethylene glycol (PEG) molecules that 
permits liposomes to escape opsonization (coating with plasma proteins — opsonins — that 
make liposomes visible by RES macrophages). PEG - conjugated lipids are used for the preparation 
of this type of vesicle. Targeted liposomes or immunoliposomes are liposomes that in 
addition to a PEG coating (in most cases) have targeting moiety on their surface that directs 
them to the preferred target. This targeting moiety may be a sugar (i.e., galactose, to target 
cells with galactose receptors on their membranes) or other type of molecule or an antibody 
(usually monoclonal antibody), in which case the liposomes are characterized as immunoliposomes. 
Cationic liposomes are vesicles that consist of positively charged lipids (cationic 
lipids) which may form complexes with negatively charged DNA molecules and thus are used 
for gene delivery or targeting applications. For their preparation an additional lipid (helper 
lipid) is usually required. Cationic liposomes can also have PEG molecules on their surface 
(for longer circulation in the bloodstream) and/or targeting moieties. The last type of liposome 
is the so called transformable liposome or elastic liposome . ( structure is presented and 
explained in Figure 10 ). 

line (PC) or lecithin, one of the main components of the liposomal bilayer, belongs 
to this group [1] . Phosphatidylcholine is zwitterionic in the range of physiological 
pH [16] and can be found in egg yolk. Other naturally occurring glycerophospholipids 
are the following: phosphatidylethanolamine (PE), which is isolated from 
brain lipids and is zwitterionic; phosphatidylserine (PS), which is found in bovine 
brains and posseses a negative charge; cardiolipin (CL), which is isolated from heart 
tissue or mitochondrial membranes and is negatively charged; phosphatidylglycerol 
(PG), which can be found in mitochondria or chloroplasts of mammalian cells and 
is negatively charged; and fi nally phosphatidylinositol (PI), a negatively charged 
lipid found in mammalian tissues [13] . 
Another group of naturally occurring lipids with applications in liposome technology 
is comprised of the sphingophospholipids (mainly sphingomyelin) which are 
derivatives of ceramides [17] . Sphingomyelin (SM) is found in the outer leafl et of 
plasma membranes [17] and has many similarities with PC since they both have the 
same zwitterionic polar group and two hydrophobic acyl chains. 
Custom - made lipids can be produced by de - or reacylation of natural lipids. Commonly 
used phospholipids with polar heads containing myristoyl (14 : 0), palmitoyl 
(16 : 0), stearoyl (18 : 0) fatty acids are all classifi ed by four - letter abbreviations, for 
example, DMPC, where DM stands for the number and type of fatty acids (di - myristoyl) 
and PC for the type of polar head (phosphatidylglycero - choline), and similarly 
DPPC and DSPC (Figure 2 ). 
Positively charged lipids are capable of making complexes with deoxyribonucleic 
acid (DNA) (since it is negatively charged) and are currently very popular. 
Examples of such lipids are: N - [1 - (2,3 - dioleyloxy)propyl] - N , N , N - trimethylammonium 
chloride (DOTMA), 1,2 - dioleoyl - 3 - trimethylammoniopropane (DOTAP), 
FIGURE 2 Typical phospholipid structure. 
Polar head 
(hydrophilic) 
O– O– 
O 
O O 
O O O 
O O 
O 
–O O –O 
O 
O P P 
Backbone 
(lipophilic) 
H2C 
H2C H2C 
H2C CH2 CH2 
HC 
HC 
C C C C 
H2C H2C 
LIPOSOME STRUCTURE AND CHARACTERISTICS 447

448 LIPOSOMES AND DRUG DELIVERY 
and dicetyl phosphate (DCP), and dioctadecyldimethylammonium bromide 
(DODAB). 
The last group of amphiphiles contains sterols that are present in the membranes 
of cells. The most popular among them is cholesterol (Chol), which can be easily 
incorporated in lipid bilayers, increasing their rigidity and making them less permeable, 
due to the interactions taking place with phospholipids in lipid membranes 
which result in modifi cation of the lipid acyl - chain conformation. 
Recently polyethyleneglycol (PEG, of varying molecular weight) – lipid conjugates 
have become commercially available and are frequently used in liposome 
applications. Aditionally, functionalized phospholipids exist for the covalent or noncovalent 
attachment of proteins, peptides, or drugs to the liposome surface. Most of 
these lipids fall into three major classes of functionality: Conjugation through amide 
bond formation, disulfi de or thioether formation, or biotin/streptavidin binding. 
Active lipids — mostly with anticancer activity — have also been added in liposome 
membrane for production of active liposomes. Examples of such lipids are 
ether lipids [18] and arsonolipids [19] . 
Several techniques are employed for the physicochemical characterization of 
lipid membranes, as summarized in Table 2 . Thermal analysis, mainly differential 
scanning calorimetry (DSC), has been used extensively, offering information on the 
thermodynamics of various types of liposomes. The phase behavior of lipid components 
of membranes determines membrane fl uidity. Each lipid has a characteristic 
lipid chain transition temperature, T m . Changes in the structure of lipids occur below 
and above this temperature [20] . The temperature at which these changes occur 
depends on the head group, the chain length, and the degree and type of unsaturation 
of each lipid [21] . Using DSC studies in has been demonstrated that heat capacity 
curves are affected by the size of vesicles [20, 22] , and can be modifi ed by 
introduction of drugs [23, 24] or peptides [25, 26] in the lipid membranes (due to 
interactions between incorporated molecules and lipids). 
TABLE 2 Methods for Physicochemical Characterization of Lipid Membranes 
Method Information References 
Thermal analysis, mainly Membrane fl uidity 20 – 26 
Differential scanning calorimetry 
(DSC) 
Lipid chain transition temperature, T m 
Fluorescence spectroscopy Phase transitions 27, 28 
Membrane dynamics 29, 30 
Nuclear magnetic resonance (NMR) Polymorphism 31, 32 
Lamellarity 33 
Membrane dynamics 34, 35 
Electron paramagnetic resonance 
(EPR) 
Fluidity of membranes 36 
Liposomal internal volume 37 
Membrane dynamics 38 
Membrane – drug interactions 39 
Fluorescence quenching Fusion processes 40, 41 
X - ray diffraction Structural information; thickness of 
the membrane and water layers 
42 – 45 

5.3.2.2 Physicochemical Properties of Liposomes 
The in vitro and in vivo performance of liposomes is highly dependent on their structural 
and surface properties. Liposome size and size distribution, surface charge (zeta 
potential), and trapping effi ciency of the drug incorporated in the liposomes are 
important parameters that should be measured when developing a liposomal drug 
formulation. To obtain optimum performance of a liposomal preparation, parameters 
infl uencing both the liposome and the drug need to be carefully considered during 
early stages of development. In this chapter the most important physicochemical 
properties of liposomes will be discussed in terms of the way they may affect liposomal 
drug performance as well as the techniques used for their measurement. 
Liposome Size Distribution Liposomes have to be smaller than the vascular pore 
cutoff (380 – 780 nm) to extravasate and reach solid tumors [46] . Liposome size also 
plays an important role in complement activation and RES clearance of liposomes 
[47] and [48] . In general, vesicles that are larger than 100 nm require additional 
strategies for preventing surface opsonization and prolonging their circulation half - 
life (Table 1 ). Light scattering, fi eld fl ow fractionation, microscopy, size exclusion 
chromatography, and turbidity are commonly used techniques for the physicochemical 
characterization of liposomal dispersions. 
Quasi - elastic light - scattering or photon correlation spectroscopy is the most 
popular light - scattering technique. The Brownian motion of the particles causes 
fl uctuations in the light intensity versus time. The hydrodynamic radius and the 
polydispersity index of liposomes can be easily determined from these studies [49 – 
51] . Field fl ow fractionation is another approach to measure the particle size and 
the surface charge of liposomes [52 – 54] . The liposomes are exposed to a perpendicular 
fi eld under laminal fl ow and their size and mass distribution can be determined. 
Size exclusion chromatography is a simple method to determine the size 
distribution of liposome dispersions [13, 55] , especially on a routine basis, if other 
sophisticated equipment is not available. Samples with high heterogeneity are suitable 
to be analyzed with this method [55] . The selection of the proper gel, pretreatment 
of the column with sonicated vesicles to avoid any loss of material by adsorption, 
and use of isotonic buffer to avoid osmotic shock and frequent column calibration 
can secure the reproducibility of the method [13, 55] . 
Turbidity is a spectroscopic technique determining the optical density of colloidal 
particles. A wavelength between 350 and 500 nm is the fi rst choice for such studies 
[56] . Turbidity measurements can offer important information on the kinetics of 
membrane – surfactant interactions since membrane solubilization changes refl ect 
changes to the optical density of the dispersion [57 – 60] . It is also widely used as a 
technique to investigate liposome aggregation and fusion [61] . However the exact 
particle size of liposomes cannot be determined using turbidity techniques. 
Optical microscopy is applicable for LUVs, MLVs, and especially giant liposomes 
[62, 63] . For instance, mechanical properties of the liposomal membrane can be 
studied by combining optical microscopy and a micropipette technique [64] . Electron 
microscopy can give information about both the morphology and homogeneity 
of liposomes. Quantifi cation in terms of number can be carried out measuring at 
least 300 particles from different images [65] . Transmission electron microscopy 
(TEM) is a powerful technique since magnifi cations up to 200,000 and a resolution 
of approximately 1 nm can be achieved. Negative staining is a quite common 
LIPOSOME STRUCTURE AND CHARACTERISTICS 449

450 LIPOSOMES AND DRUG DELIVERY 
technique for visualization studies. A small amount of the sample is dried on a grid 
coated with carbon fi lm. Then the fi lm is coated with an electron - dense solution 
(e.g., tungsten molybdate). However the technique suffers from some drawbacks, 
such as artifacts due the fi xation process or the extraction of lipid material by 
embedments [66, 67] . With freeze - fracture electron microscopy (FFEM) samples are 
quickly frozen and fractured. Compared with negative staining technique, FFEM 
has the advantage of preserving water - dependent lipid phases because no dehydration 
steps are involved. Therefore phase transitions, lipid polymorphism, or fusion 
processes can be visualized with this approach [68 – 71] . Recently cryogenic transmission 
electron microscopy (cryo - TEM) has been employed for visualization studies. 
With this technique very precise morphological assessment of liposome interior 
FIGURE 3 Morphology of liposome of liposome by cryo - TEM (transmission electron 
microscopy). ( a ) Empty liposomes (liposomes with entrapped unbuffered CuSO 4 in the 
absence of drug). ( b ) Topotecan - encapsulating liposomes (drug was added to the empty 
liposomes of ( a ) to achieve a fi nal drug - to - lipid ratio of 0.2 mol/mol, and the system was 
incubated at 20 ° C). ( Reproduced from ref. 72 with permission of Elsevier .) 
(a) 
(b)

(Figure 3 ) as well as liposome surface (Figure 4 ) can be carried out as recently 
demonstrated for Copper - Topexan encapsulating [72] and transferring - coated liposomes 
[73] , respectively. Compared with the previous techniques the main advantage 
of cryo - TEM is avoidance of any fi xation of the grid, which can create artifacts 
induced by staining and thus keeps the sample close to the original state [74, 75] . 
The samples for the cryo - TEM studies are prepared in a controlled environment 
vitrifi cation system (CEVS). A small amount of the sample is placed on a grid - supported 
fi lm. The grid is quenched in liquid ethane and it is vitrifi ed. Then the samples 
are characterized with a TEM microscope. In a manner similar to the previous discussion, 
lipid polymorphism or fusion can be investigated [61, 76 – 79] . Finally scanning 
probe microscopy (SPM) including atomic force microscopy (AFM) has been 
recently applied in the liposome fi eld [73, 80] . Some of the advantages of the technique 
are the high resolution in atomic dimensions (Figure 5 [73] ), the production 
of three - dimensional images with high resolution, and the versatility of the operation 
conditions (vacuum air liquid). Aditionally, sample preparation does not involve 
any staining, freezing, embedding, or fracturing procedures. 
Surface Charge of Liposomes The electrical properties of liposomal surfaces can 
infl uence the physical stability of liposomal dispersions during storage as well as the 
behavior of liposomes in the biological milieu and their interaction with cells 
[47, 81] . 
Microelectrophoresis is used to measure the electrophoretic mobility or, in other 
words, the movement of liposomes under the infl uence of an electric fi eld. From the 
electrophoretic mobility the electrical potential at the plane of shear or . (zeta) 
potential can be determined (by the Helmoholtz – Smoluchowski equation). From 
the zeta potential values the surface charge density ( . ) can be calculated. 
Aggregation of liposomes both in vitro and in vivo is one of their main stability 
problems. According to the Derjaguin-Landau-Verwey-Overbeek (DLVO) theory, 
or theory of colloidal stability, a colloidal system is stable if the electrostatic repulsion 
forces between two particles are larger than the attraction van der Waals forces. 
Therefore charged liposomal formulations are highly desirable. Manipulation of 
FIGURE 4 Morphological assessment of liposome surface coating using cryo - TEM. Liposomes 
that have been conjugated with transferrin display small particles on their surface ( a ), 
while control preparations appear to be smooth and undecorated ( b ). ( Reproduced from ref. 
73 with permission of Elsevier .) 
(a) (b) 
LIPOSOME STRUCTURE AND CHARACTERISTICS 451

452 LIPOSOMES AND DRUG DELIVERY 
liposome surface renders them stable against self - aggregation or nonspecifi c interactions, 
and this can be achieved by introducing other molecules on the surface, 
which may be natural molecules such as glycolipids [82, 83] or antibodies [84] or 
lectins [85] (that are usually grafted on the liposome surface by chemical linking). 
As also mentioned in the introduction, the RES has been the “ Achilles heel ” to 
the delivery of liposomes by injection to the blood stream because of their rapid 
FIGURE 5 Morphological assessment of liposome surface coating using atomic force 
microscopy (AFM). AFM images of liposomes prepared with DSPE – PEG 200 – COOH. 
( a ) Plain liposomal formulations. The liposomes show a smooth surface morphology. ( b ) 
Liposomes covalently modifi ed with transferring. Small globular structures are visible at the 
surface. ( Reproduced from ref. 73 with permission of Elsevier .) 
(a) 
(b) 
250 nm 
250 nm

uptake by the macrophages of the RES. Grafting of liposome surface with hydrophilic 
polymers has been used successfully as a method to protect the liposomes 
with a steric barrier which inhibits the adsorption of blood components [7, 86, 87] . 
It has been shown that sterically stabilized liposomes have signifi cantly lower zeta 
potential values compared with conventional liposomes [87] , indicating that the 
presence of PEG might shift the plane of shear away from the phosphate moieties. 
Thereby, zeta - potential modifi cation can serve as proof of successful coating of 
liposomes by polymers. 
Drug Loading Effi ciency and Techniques The amount of drug incorporated (for 
amphiphilic or lipophilic drugs) or encapsulated (for hydrophilic drugs) is a very 
important parameter that determines largely the selection of liposome type and 
components. Terms used to quantitate drug loading are drug – lipid (mol/mol) ratio 
(or D/L) and trapping or encapsulation effi ciency (which is the percent D/L in the 
fi nal liposomal formulation compared to the initial D/L used for liposome preparation). 
In many cases, the percent encapsulation is mentioned (percentage of drug 
encapsulated in relation with the amount of drug offered for encapsulation during 
liposome preparation); however, this number is practically useless and misdirecting, 
since it highly depends on the initial amount of drug offered to the specifi c lipid 
quantity, while the amount of lipid is not quantifi ed. 
Several parameters infl uence the encapsulation of drugs into liposomes. The 
types of vesicle and drug used play signifi cant roles in the percentage of encapsulation. 
Hydrophobic drugs have higher loading effi ciency in MLVs since they consist 
of high numbers of bilayers and low aqueous volumes [88 – 91] . In contrast, hydrophilic 
drugs have higher encapsulation in LUVs [91, 92] . Generally, for hydrophilic 
drugs, the percentage of encapsulation increases in the order of SUV . MLV < 
LUV [92] . 
The method used for liposome preparation has a signifi cant impact on drug 
loading as well. Larger surface areas for the formation of thin fi lms are preferable 
since they facilitate the hydration process of the bilayer, as demonstrated for doxorubicin 
(DOX) using fi ve different hydration protocols [93] . The highest encapsulation 
was achieved when a thin fi lm with large surface area was formed. Glass beads 
can be used for this purpose. From 8 to 10 times higher values for hydrophobic drug 
encapsulation were obtained after the addition of such beads [94] . The type of 
hydration (conditions and media used) also infl uences drug loading. 
Drug encapsulation and stability of liposomes are also affected by the length of 
the acyl chain and the degree of saturation of the lipids used for their formation. 
As the acyl chain length of the lipid increases, so does the partitioning of hydrophobic 
drugs in the lipid membrane [95] , as demonstrated for atenolol and propranolol 
in MLVs and SUVs [96] . The impact of the head group of different lipids used for 
the preparation of liposomes on encapsulation of citicoline was investigated by 
Puglisi et al. [97] . The fl uidity of the membrane has also been demonstrated to infl uence 
encapsulation of different compounds [98 – 100] . 
When galactocerebroside was incorporated into liposomes, the percentage of 
encapsulation of mintoxantrone increased proportionally to the amount of glycolipid 
present in the membrane [101] . Numerous studies have demonstrated the 
impact of vesicle surface charge on drug loading effi ciency, which is important for 
charge - bearing molecules. Encapsulation of hydroxycobalamin [102] and doxorubicin 
[103] was higher in negatively charged liposomes compared to neutral ones. In 
LIPOSOME STRUCTURE AND CHARACTERISTICS 453

454 LIPOSOMES AND DRUG DELIVERY 
a similar manner, higher amounts of calcitonin were encapsulated in positively 
charged compared to neutral liposomes [104] while the highest loading of indomethacin 
in liposomes was obtained in the following order: positively charged > 
negatively charged > neutral liposomes [105] . 
Cholesterol and . - tocopherol are used quite often to increase the rigidity and 
stability of liposomal membranes [88, 106 – 108] . In most cases cholesterol appears 
to improve the encapsulation of both hydrophilic and hydrophobic compounds. 
However, if the drug is lipophilic and partitions in the liposome membrane, there 
is a good chance that it might be displaced by adding increasing amounts of cholesterol 
in the bilayer (as observed in the case of dexamethasone encapsulating liposomes 
in our laboratory). 
A novel system for enhancing the drug loading of lipophilic drugs combining 
liposomes and cyclodextrin – drug complexes by forming drug - in - cyclodextrins - 
in - liposome preparations has been proposed [109 – 111] . Cyclodextrins (CDs) are 
hydrophobic, cavity - forming, water - soluble oligosaccharides that can accommodate 
water - insoluble drugs in their cavities, increasing their water solubility. The basic 
intention is to encapsulate a stable water - soluble drug – cyclodextrin complex in the 
aqueous compartments of liposomes (Figure 6 ). As will be discussed below, this 
system can also serve as a method to increase the retention of lipophilic drugs in 
diluted liposome dispersions. 
Finally, remote loading and active loading [11] are other methods used to achieve 
high trapping effi ciency in liposome formulations, but unfortunately they can only 
be applied to a small number of drugs with specifi c physicochemical properties. This 
technique will be discussed below in Section 5.3.2.3 . 
= = 
Lipid bilayer 
Aqueous interior 
Drug–CD 
complex 
(High aqueous solubility) 
CD Drug 
(Low aqueous solubility) 
FIGURE 6 Representation of drug in – cyclodextrin in liposome technique for encapsulation 
of lipophilic drugs in aqueous interior of vesicles. Drug molecules have low aqueous 
solubility and thus cannot be encapsulated in the aqueous compartment of the vesicle. 
However, the drug – cyclodextrin complex has high aqueous solubility and can thus be encapsulated 
in high concentrations in the vesicles. 

Stability of Liposomes A shelf life of at least two years is requested for pharmaceutical 
products. Therefore, chemical stability and physical stability are important 
parameters for the overall performance of liposomal formulations. Additionaly, 
another very important factor is the retention of encapsulated drug. 
Several studies have suffi ciently addressed the chemical degradation of liposomes 
during storage [112 – 115] . This is due to the hydrolysis of the phospholipids 
to fatty acids and 1 - and 2 - acyl - lysophospholipids. Further hydrolysis leads to the 
production of glycerol phospho compounds. Antioxidants ( . -tocopherol), complexing 
agents [e.g., ethylenediaminetetraacetic acid (EDTA)], and inert atmosphere 
(e.g., nitrogen) are most commonly used to overcome this problem. Moreover, the 
presence of . - tocopherol can reduce the auto - oxidation of lipids, which is usually 
induced by light, metal ions, and temperature. Prevention of the chemical decomposition 
of the lipids by adding . - tocopherol in liposomal dispersions can increase 
the shelf life of liposomes [116] . Furthermore, the coexistence of cholesterol and . - 
tocopherol in the lipid bilayer can improve the antioxidant activity of tocopherol 
[117] . 
An alternative to circumvent problems related to the chemical decomposition 
of liposomes is their storage in the dry state (freeze dried). However, the protection 
against damage by freezing (cryoprotection) [118 – 120] and the protection 
against damage by dehydration (lyoprotection) [121, 122] require special attention 
for the proper storage of liposomes. Depending on the drug encapsulated in the 
liposomes and possible interactions between drug molecules and components of 
the lipid membrane, initial studies should be carried out in order to fi nd the proper 
cryo - and lyoprotectant which preserves the integrity of the specifi c liposomal 
formulation. 
The physical stability of liposome dispersions is mainly related with possible 
aggregation and leakage of the liposomal membrane. The size and surface charge 
play a signifi cant role for the stability of liposomal dispersions, as has been discussed 
in the previous paragraph. 
In addition to physical stability, the retention of drug in liposomal formulations 
is particularly important, not only during storage, but also during in vivo administration. 
Especially when targeted or long - circulating liposomes are used, association 
of the drug in the liposome carrier until the carrier reaches its biological target is 
a prerequisite for achieving any therepeutic benefi t. However, in many cases, 
although the amount of drug loaded in liposomes is initially high, for different 
reasons most of the liposome - associated drug is rapidly released from the vesicles. 
The main causes for such behavior are different according to the physicochemical 
properties of the drug. For hydrophilic drugs: (i) The low integrity of liposome 
membranes after in vivo administration and contact with blood compomnents, 
which results in removal of some lipid molecules and concurrent opening of pores 
in the liposomal bialyer through which the loaded drug molecules may leak out, 
and (ii) the physical instability of liposomes that results in aggregation and fusion. 
and fi nally release of drug from liposomes. For amphiphilic and lipophilic drugs , 
the main problem is caused by the dilution of liposome dispersions, which usually 
occurs immediately after in vivo administration [by most routes, especially i.v. 
(diluted in 4 L of blood)]. This results in release of drug (since the drug can permeate 
membranes) until the drug saturates the full aqueous volume in which the 
liposome vesicles are diluted. 
LIPOSOME STRUCTURE AND CHARACTERISTICS 455

456 LIPOSOMES AND DRUG DELIVERY 
For hydrophilic drugs, the problem can be solved by increasing the rigidity of the 
liposome membrane while concurrently decreasing their tendency for aggregation, 
after proberly selecting the liposome lipid components. However, for membrane - 
permeable drugs, drug leaking upon dilution cannot be easily, if at all, confronted. 
Theoretically, one method may be to increase the affi nity of the drug molecule with 
the lipid membrane (and at the same time decrease its aqueus solubility) by chemical 
modifi cation (conjugation to a lipid or fatty acid). However, this is neither easy 
nor generally applicable. The association of liposomes with cyclodextrins has been 
recently proposed as a method to ensure high and stable entrapment of lipophilic 
drugs in aqueous compartments of liposomes [109 – 111, 123] (Figure 6 ), providing 
that the drug has high affi nity for the cyclodextrin molecule and is not displaced 
from the cyclodextrin cavity by components of the lipid membrane (mostly cholesterol). 
In such cases, this approach may not increase drug retention, but encapsulation 
of drug may be substantially improved [111] . 
5.3.2.3 Preparation of Liposomes 
Liposome Preparation Techniques In most cases, liposomes are named by the 
preparation method used for their formation, Such as sonicated, dehydrated – rehydrated 
vesicle (DRV), reverse - phase evaporation (REV), one step, and extruded. 
Several reviews have summarized available liposome preparation methods [91, 124, 
125] . Liposome formation happens spontaneously when phospholipids are dispersed 
in water. However, the preparation of drug - encapsulating liposomes with high drug 
encapsulation and specifi c size and lamellarity is not always an easy task. The most 
important methods are highlighted below. 
Thin Film Method It was in 1964 that Alec Bangham introduced the “ thin - fi lm 
method ” for liposome preparation [1, 126] . Lipids are dissolved in organic solvents 
(chloroform or mixtures with methanol) and the solvent is removed under a high - 
vacuum rotor evaporator forming a thin fi lm on the walls of round - bottomed fl ask. 
Depending on the phase transitions of the lipids used for the preparation of liposomes, 
the aqueous phase for the rehydration should be prewarmed at temperatures 
above the phase transition of the lipid. After addition of the aqueous phase the thin 
fi lm is detached from the fl ask walls by agitation and a highly heterogeneous population 
of MLVs is produced. Depending on the physicochemical properties of drugs, 
they can be introduced either in the thin fi lm together with the lipids (lipophilic 
compound) or in the rehydration solution (hydrophilic compound). 
Sonication This approach uses energy (ultrasound) and can be applied to a dispersion 
of MLVs [127] or to solid lipids mixed with aqueous solution. The fl ask with the 
liposome dispersion is placed in a bath sonicator or a probe sonicator (tip) is immersed 
in the tube containing the liposome dispersion. With the fi rst setup it is diffi cult to 
reduce the size to the nanometer level since the energy produced by the bath sonicators 
is rather low. However, it has the advantage that there is no contact with the liposome 
dispersion. The position of the fl ask in the sonicator is equally important. It is 
easy to understand if it is in the right place from the noise produced by ultrasound 
waves. For instance, if foams are produced or there is no noise at all, that implies the 
sample is misplaced and fi nally the size of vesicles will not be reduced. 

In the second approach (probe sonicator), the size of the liposomes can be 
reduced to nanometers and SUVs can be produced. As previously, the position of 
the probe plays an important role on the ability to minimize vesicle size. Because 
the energy produced from the transducer is high, overheating of the system is quite 
common; therefore, a water bath fi lled with ice is recommended. After sonication, 
fragments of Ti originated from the probe are scattered in the dispersion. Centrifugation 
for 4 – 5 min at 10.000 rpm will cause sedimentation of these fragments, giving 
clear liposome dispersions. 
Injection Methods 
Ethanol Injection Small unilamellar vesicles (with diameter of 30 nm) can be 
prepared with the ethanol injection technique [128] . Lipids are dissolved in 
ethanol and injected rapidly in the aqueous solution under stirring (fi nal concentrations 
up to 7.5% (v/v) ethanol can be applied). The method is very easy, 
having the advantage of avoiding chemical or physical treatment of lipids. 
However, there is an extra step to remove ethanol and the concentration of 
vesicles produced is rather low. Also encapsulation of hydrophilic drugs is also 
low, due to the high volumes used. 
Ether Injection The general principle of this method is the same as ethanol 
injection. The only difference is that the lipid is injected slowly in the aqueous 
solution that is warm [129] . Furthermore, the concentrations used in this case 
are somewhat higher (up to 10 m M ) compared to the ethanol injection 
approach. 
Extrusion (Extruded Vesicles) The extrusion method, which today is very popular 
for the production of homogenous vesicle samples of a predetermined size, was 
introduced by the group of D. Papahadjopoulos [65] . Multilamellar vesicles are 
extruded through fi lters with well - defi ned pores under pressure. Polycarbonate is 
the most commonly used material for these membranes, which have pore sizes from 
30 nm up to several micrometers. For lipids with high melting point, the extrusion 
should be carried out above their phase transition temperature. The operating 
volumes are from 1 to 50 mL with a liposome dispersion concentration up to 150 m M . 
Generally, repeated extrusions reduce the number of lamellae and the produced 
liposomes are mainly unilamellar. High pressure can cause disruption of membranes. 
The reproducibility of the method is good; however, it is quite time consuming 
and membrane rupture problems occur quite often. 
French Press With this approach MLV liposomes are introduced in a cell and a 
piston presses the dispersion [130] . Pressures up to 25,000 psi can be achieved and 
SUVs are produced. The main disadvantage of this technique is that it is not applicable 
for lipids with phase transition temperatures lower than 20 ° C and the concentration 
of liposomes that can be used is relatively low (maximum 20 m M ). 
Microfl uidization During microfl uidization, MLVs are circulated with a pneumatic 
pump operating under high pressure through a prefi lter and then to the interaction 
chamber [131] . From there, they are separated into two streams and they pass 
through defi ned microchannels under high velocity to a heat exchanger which is 
LIPOSOME STRUCTURE AND CHARACTERISTICS 457

458 LIPOSOMES AND DRUG DELIVERY 
connected with a water bath. This is repeated many times until the size of the liposomes 
is signifi cantly reduced. They can operate with volumes from 0.1 up to 10 L 
of liposomes and with concentrations up to 300 m M , which is by far the highest 
capacity from all other methods. Small unilamellar vesicles less than 100 nm can be 
produced, but the population is not completely homogeneous. 
Reverse - Phase Evaporation The REV method was developed by Szoka and Papahadjopoulos 
[132] . Lipids are dissolved in organic solvent and the solvent is removed 
with evaporation. The thin fi lm is resuspended in diethyl ether (1 mL solvent/mL 
liposomes) followed by the addition of one - third of water and sonication in a bath 
sonicator for 1 min. This water - in - oil (w/o) - emulsion is evaporated until a dry gel is 
formed, and fi naly the gel is broken by agitation and water addition. Sometimes this 
step is quite diffi cult. The remnants of the organic solvent are removed by evacuation 
and the resulting dispersion is REV liposomes. 
Dehydrated – Rehydrated Vesicles DRV liposomes were developed by Kirby and 
Gregoriadis in 1984 [133] and are capable of encapsulating high amounts of aqueous 
soluble molecules under mild conditions (conditions that do not cause decomposition 
or loss of activity). The high entrapment ability of this type of liposomes is due 
to the fact that preformed, “ empty ” SUVs are disrupted during a freeze - drying step 
in the presence of the solute destined for entrapment. Subsequently, during controlled 
rehydration that is carried out in the presence of concentrated solution of 
the solute (to be encapsulated), the vesicles fuse into large oligolamellar vesicles 
entrapping high amounts of solute. The produced liposomes are multilamellar and 
their size is between 200 and 400 nm up to a few micrometers. Recently, with the 
addition of certain amounts of sucrose, DRV liposomes with diameter between 90 
and 200 nm were obtained entrapping considerable proportions (up to 87) of the 
solute [134] . 
Giant Vesicles Large or giant liposomes have been developed by Reeves and 
Dowben [135] . Briefl y, lipids are dissolved in chloroform/methanol 2 : 1 and dispersed 
on a piece of glass. Water is added for their rehydration; however, their 
population is quite heterogeneous. Other types of particle - encapsulating giant liposomes 
[136] can be prepared by applying a double - emulsion technique followed by 
a freeze – drying step. 
Detergent Depletion With this method phospholipid – detergent mixed micelles are 
initially produced, and during controlled - rate detergent removal, liposomes are 
formed. The rate and method of detergent removal determine the size and size 
homogeneity of the liposomes produced. Gel fi ltration and dialysis are the most 
popular approaches [137, 138] . Although liposomes are produced under mild conditions 
(low temperature and low shear mechanical forces applied), this method 
suffers from low encapsulation effi ciency of hydrophilic drugs. 
Large Unilamellar Vesicles from Cochleates Large unilamellar vesicles can be produced 
with the “ cochleate ” approach [139] . Small unilamellar vesicles consisted 
from phosphatidylserine adopt a cochleate shape after addition of calcium. Addition 
of EDTA creates complexes with calcium, turning the cochleates to LUVs. 

One - Step Method The “ one - step method ” has been introduced by Talsma et al. 
[140] . Lipid dispersions are hydrated at high temperatures in the presence of a steam 
of N 2 . Liposomes between 200 and 500 nm can be prepared with this approach. 
Large - Scale Manufacturing Despite the fact that there are many methods to 
prepare liposomal dispersions not all of them are applicable for scaling up. In fact, 
scaling up to larger batches could be a monumental task. Among all the methods 
reviewed so far, microfl uidization and homogenization are the most powerful 
methods to produce large quantities of liposomes. New homogenizers have a capacity 
of 1000 L/h and require a minimal sample volume of 2 L [141] . The fact that lipid 
concentrations up to 300 m M can be used secures high encapsulation capacity. 
Remode and Active Drug Loading Techniques The main advantages of these 
approaches are the high encapsulation effi ciency and low leakage of the encapsulated 
material. 
An in situ method for “ remote ” drug loading based on the development of a pH 
gradient across the internal and external water phases of the membrane has been 
established. A transmembrane pH gradient induces the uptake of charged drugs 
into liposomes. Drug encapsulation is based on its partitioning between the lipid 
and aqueous parts. This process is governed mainly by pH and to some extent by 
the ionic strength of the medium. Drugs that are weak bases can diffuse through 
the lipid membrane as unprotonated species. The presence of a proton gradient 
makes them more hydrophilic, allowing them to accumulate in the intraliposomal 
aqueous phase. Encapsulations up to 90% have been reported for doxorubicin 
[142 – 144] and vincristine [145] . 
An alternative but similar technique is based on an ammonium sulfate gradient 
used to obtain “ active ” loading of amphipathic weak bases into the aqueous compartment 
of liposomes. This has been used for active loading of anthracyclines, 
acridine orange, epirubicin, and doxorubicin [11, 146, 147] at very high effi ciency 
( > 90%). In the case of doxobubicide most of the intraliposomal drug is present in 
the aggregated state. Additionally, antracycline accumulation in liposomes is stabilized 
for prolonged periods of storage due to aggregation and gelation of antracycline 
sulfate salt. 
Active entrapment and loading stability are dependent on liposome lipid composition, 
lipid quality, medium composition, and temperature as well as on the p K a and 
hydrophobicity of the base. The ammonium sulfate gradient approach differs from 
most other chemical approaches used for remote loading of liposomes, since it does 
not require liposomes with acidic pH interior or an alkaline extraliposomal phase. 
In addition to the remote or active loading techniques mentioned above, metal 
complexation reactions have been demonstrated to achieve accumulation of doxorubicin 
in liposomes [148, 149] . Furthermore, copper – topotecan complexation has 
been recently seen to mediate drug accumulation into liposomes and is proposed 
as a methodology to prepare liposomal camptothecin formulations [72] . 
5.3.2.4 Functionalization of Liposomes 
“ Active targeting ” is used to describe the specifi c liposomal drug localization achieved 
by grafting various moieties (antibodies, lectins, polymers, etc.) on the carrier surface. 
LIPOSOME STRUCTURE AND CHARACTERISTICS 459

460 LIPOSOMES AND DRUG DELIVERY 
There are a number of techniques available to attach the suitable ligand on the liposome 
surface, either by covalent or noncovalent coupling [150, 151] . 
These techniques should be fast, effi cient, and reproducible, yielding stable nontoxic 
bonds, while the conjugated ligands should maintain the ability to recognize 
the target site with high binding affi nity. Also, the coupling method should not affect 
the blood clearance of the formulation, colloidal stability, drug incorporation, and 
release in a negative way. For example, when antibodies were attached on the liposome 
surface where PEG molecules were grafted as well to ensure prolonged carrier 
retention in the blood, it was shown that the ligand binding effi ciency on the bilayer 
was low as well as the binding ability to the target [152, 153] . The latter problem 
was opposed by attaching the ligand at the distal end of the PEG molecules already 
grafted on the liposomal bilayer [154] . 
Covalent Binding of Ligands The majority of ligand coupling is achieved by 
covalent reactions with hydrophobic anchors. The procedure could be carried out 
in two patterns: Either the hydrophobic anchor is included already in the liposomal 
bilayer and the ligand interacts with the anchor on preformed liposomes [155] or 
the ligand – hydrophobic anchor conjugate in the form of micelles is mixed with the 
liposomes [156] . In the fi rst instance, where a hydrophobic anchor is mediated 
between the liposomal surface and ligand, covalent attachment can occur via a 
thioether bond [157 – 161] , via a disulfi de bridge, between carboxylic acid and the 
primary amine group [162] , via hydrazone, or via cross - linking between two primary 
amine groups (Figure 7 ). 
The formation of the stable thioether bond is a reaction between thiol moieties 
of proteins mainly with maleimide groups. Usually, PE or PEG – PE or PEG – DSPE 
(distearoyl - phosphatidylethanolamine) has been functionalized with maleimido 
(Mal - ), maleimido - phenylpropionate (MP), or pyridil - dithio - propionylamino 
(PDP) groups, which eventually will react with the thiol groups. Also, sometimes 
the ligand does not carry enough thiol groups or those are completely absent, so 
they have to be introduced using a heterobifunctional cross - linker, such as SPDP 
[ N - hydroxysuccinimidyl 3 - (2 - pyridyldithio) propionate] or SATA (succinimidyl - 
S - acetylthioacetate), which introduce one amine group. Still, in the case of SATA, 
deacetylation using hydroxylamine is necessary to uncover the thiol group, while if 
SPDP is used, the produced disulfi de bond has to be reduced to thiol groups with 
dithiothreitol (DTT). The former cross - linker is more preferable as only mild conditions 
are used to make the thiol group available. 
It has been shown that attaching the ligand at the distal end of PEG molecules 
combines the advantages of a specifi c drug delivery system with steric stabilization 
for higher stability in the blood [163, 164] . However, Longmuir et al. showed that 
introducing a peptide from Plasmodium at the free end of PEG 3400 – AP (aminopropane) 
was not capable of retaining the liposome stability, so PEG 5000 – PE molecules 
were added to the liposomal composition resulting in PC/PE – PEG 5000 /AP – PEG 3400 - 
peptide liposomes with molar ratio of 86 : 10 : 4 [165] . The coupling effi ciency of 
ligand attached on PEG was higher (60 – 70% or even 100% in some cases) compared 
to that achieved using, for example, N - (4 . - 4 . - maleimidophenyl)butyrol) - 
dioleoylphosphatidylethanolamine (MPB – DOPE) (only 10%) [164, 166] . 
Antibodies, whole or fragments, have been attached successfully on liposomal 
surfaces and more commonly at the free end of PEG molecules, as has been reported 

FIGURE 7 Schematic of different coupling methods used: ( a ) reaction between meleimide 
and thiol functions; ( b ) formation of disulfi de bond; ( c ) reaction between carboxylic acid and 
primary amine group; ( d ) reaction between hydrazide and aldehyde functions; ( e ) cross - 
linking between two primary amine functions. ( Reproduced from ref. 150 with permission of 
Elsevier .) 
(a) 
R1 R2 R2 
R1S 
SH 
O
O 
N N 
O
O 
+ 
(b) 
R1 R1 R2 R2 SH HS S S + 
(c) 
CH3 
CH3 
NH2 
CH3 
CH3 
CH2CH3 
CH2CH3 (CH2)
3 
(CH2)
3 N 
N 
N 
N HN 
HN 
C 
C 
C 
N 
O
C 
C
O 
O 
O 
OH Anchor 
Anchor 
Anchor 
BDAC 
Ligand 
Ligand 
+ 
+ 
(d) 
R1 
R1 
R2 R2 
R2 
NH2 
N 
H 
NH 
NH 
C C
O 
O 
O
O 
OH 
CH 
Nalo4 
+ 
(e) 
R1 
R1 
R1 
R2 
R2 
NH2 
NH2 
CH2 CH2 CH2 
CH2 
CH2 CH2 CH2 CH2 CH2 
CH2 CH2 
C 
C C N 
N N 
C H 
H 
H 
H 
H 
H 
O 
O 
O 
+ 
+ 
Glutaraldehyde 
LIPOSOME STRUCTURE AND CHARACTERISTICS 461

462 LIPOSOMES AND DRUG DELIVERY 
in a number of studies since the interaction with the cells is much more favored 
[161] . Using Fab . fragments is more advantageous as the Fc part which mediates 
MPS activation through a receptor, is omitted [157, 160] . Moreover, the distance of 
the antibody fragment from the liposomal surface is another important factor which 
determines the drug delivery system uptake by the cells, as reported by Mamot 
et al. [167] . Besides the latter has been reported elsewhere even if the coupling 
reaction is different from thioether formation [168] . 
Examples of peptides attached on liposomal surface via thioether bonds were 
TAT - peptide and antagonist - G on Mal – PEG2000 – DSPE and PDP – PEG – DSPE, 
respectively [169, 170] . Both ligands exhibited signifi cant increase in cell uptake. 
Another possible way for ligand conjugation on liposomes is the formation of a 
disulfi de bridge, which is quite unstable in serum and thus it is not used as much 
[171] . 
However, an amide bond formed between the carboxylic acid group on the liposome 
surface (DSPE – PEG – COOH) and the primary amine of the ligand is favored 
as the ligand modifi cation is not necessary. According to this method, an acyl amino 
ether is produced in the presence of 1 - ethyl - 3 - (dimethylaminopropyl) carbodiimide 
(EDAC) and N - hydroxysulfosuccinimide (NHS), which eventually will react with 
the primary amine of the ligand [150] . For example, Wartchow et al. improved the 
effi cacy of a small integrin antagonist of the extracellular domain of the . . . 3 integrin 
by grafting it on dextran - coated liposomes [172] . DPPE - succinate was included 
in the liposomal bilayer and a 3 - amino - 2 - hydroxypropyl ether derivative of dextran 
was added to preformed liposomes in the presence of EDAC, while unreacted succinyl 
groups were converted to amides. The amino groups of dextran were succinylated 
and integrin antagonist (IA) was attached on the succinamidodextran liposomes 
in the presence of EDAC. The fi nal IA – dextran liposomes had a size of 110 nm, 
which is attributed to dextran coating, as the liposomes without dextran were 60 nm 
in diameter. The antiangiogenic mechanism of IA – dextran liposomes as well as the 
apoptotic potency was proved after a series of studies. 
Moreover, Voinea et al. attached antibodies against vascular cell adhesion 
molecule - 1 (VCAM - 1) overexpressed on activated human endothelial cells on liposomes 
with the intention of using them as drug carriers [162] . N - gluraryl - PE was 
used as membrane anchor for the antibody coupling via its free amino groups after 
its activation with carbodiimide. There is no necessity of antibody modifi cation 
before the coupling reaction. 
Also, transferrin was grafted onto liposomes containing N - glutaryl - PE activated 
with carboxidiimide with the fi nal plan to use those as carriers for inhalation therapy 
for lung cancer [173] . Tfr liposome uptake was signifi cantly higher from immortalized 
or cancer cell lines, but to reduce uptake from alveolar macrophages, PEG molecules 
are attached on the liposome surface. In general, transferrin is a glycoprotein 
that consists of a single chain of amino acids which has been coupled on the liposome 
surface for a number of applications because its receptor is overexpressed at malignant 
cells so a higher amount of transferrin binds on the cell membrane [174] . Its 
covalent attachment on liposomes takes place either by conjugation between transferrin 
- lipid and insertion of it on the preformed vesicles [174, 175] or preparation of 
liposomes with activated lipids and reaction with activated transferrin [176] . 
In addition, Torchilin et al. synthesized pNP ( p - nitrophenylcarbonyl) – PEG – 
DOPE to enable protein coupling via its amino group at the distal end of PEG mol

ecules on liposome surface in a quantitative manner at pH around 8.0 [177] . The only 
disadvantage mentioned is the hydrolysis of pNP groups from PEG – DOPE at pH 
higher than neutral (complete hydrolysis occurs in 1 – 2 h at pH around 8.0). Therefore, 
the coupling reaction between protein and pNP has to take place faster or at 
least at the same ratio. It was shown that 65% of pNP binds to ligand at pH 9, so the 
binding effi ciency and time are adequate at the conditions studied. Also, the amount 
of pNP – PEG – DOPE was critical for successful protein binding; it was shown that 
1 mol % of pNP – PEG – DOPE was enough to bind approximately 100 protein molecules. 
Incorporation of antibodies, lectins, avidin, and nucleosomes did not seem to 
alter the activity of those molecules even at high concentrations of pNP – PEG – 
DOPE. However, Savva et al. conjugated a genetically modifi ed tumor necrosis 
factor (TNF) at the free end of the PE 3500 molecule by reacting the latter with NHS 
and DCC ( N , N . - dicyclohexyl carbodiimide) to introduce the succinyl groups which 
will react with the phospholipid to produce DOPE – PEG – COOH [178] . Then, the 
carboxyl group of the derivatized PEGylated molecule located on the liposomal 
bilayer was activated with EDAC and NHS. Consequently, recombinant TNF was 
added in the liposome suspension for the fi nal conjugation to occur at 4 ° C where the 
overall coupling effi ciency was approximately 55%. However, it was shown that the 
biological activity of TNF was reduced when attached on the liposomes as the degree 
of PEG modifi cation increased irrespective of the PEG molecular weight (MW). 
Also, the formulation did not show the prolonged blood circulation expected due to 
the PEG presence. Those results were attributed to a number of reasons, including 
either damage of the protein during the coupling reaction or possible dissociation of 
the trimeric form of rKRKTNF to a monomeric less active form or cross - linking 
between PEG and rKRKTNF during the coupling reaction. 
A hydrazone bond is an alternative way of antibody coupling onto liposomal 
surface so as to avoid the use of essential (for the recognition) amino groups at the 
coupling reaction (by using the maleimido method) or the risk of rapid clearance 
due to Fc moiety/segment [150, 179] . According to this method, the carboxylic group 
of the heavy chain of the antibody undergoes mild oxidation by sodium periodate 
or galactose oxidase to aldehyde groups. The oxidized product can be coupled either 
on the hydrazide - hydrophobic anchor inserted in the lipid bilayer [180] or at the 
free end of PEG molecules [181, 182] . Hydrazide cross - linkers used are, for example, 
S - (2 - thiopyridyl) - l - cysteine hydrazide (TCPH), N - acetylmercaptobutyric hydrazide 
(AMBH), and 3 - (2 - pyridyldithio)propionic acid hydrazide (PDPH). TCPH is structurally 
closely related to PDPH and could be expected to behave in a similar manner 
[183] . Unprotected mercaptohydrazides such as AMBH are unsuitable since the free 
thiol function is susceptible to oxidation and may also reduce the disulfi de bonds 
in immunoglobulin IgG at the concentrations required for conjugation. The disadvantage 
of this method is the low coupling effi ciency (17% only) [166] . According 
to Ansell et al. [179] , the drawbacks reported was the possible damage of some 
amino acid residues, such as methionine, tyrosine, and tryptophan, due to periodate. 
Therefore, antibodies sensitive to periodate treatment would be unsuitable candidates 
for the PDPH protocol. Also, the hydrazone bond might undergo hydrolysis 
after six weeks of storage, which would be a potential problem if the conjugate 
would not be used soon after preparation. However, it is possible to stabilize the 
bond using sodium cyanoborohydride to reduce the hydrazone linkage if long - term 
storage is required [179] . 
LIPOSOME STRUCTURE AND CHARACTERISTICS 463

464 LIPOSOMES AND DRUG DELIVERY 
However, the labile nature of the hydrazone bond is used to formulate “ smart ” 
drug delivery systems where the basic idea is to introduce in parallel a pH - responsive 
ligand spacer in the lipid bilayer (which is PEG 5000 – Hz – PE), a temporarily 
shielded biotin or TATp and mAb (monoclonal antibody) attached to the surface 
of the drug delivery system via a noncleavable bond (TATp – PEG 2000 – PE) [183] . 
Such a system will be able to respond to environment stimuli such as pH changes, 
where, for example, at acidic pH (5.0 – 6.0) PEG5000 molecules will be detached from 
the carrier surface and biotin or TATp will be available to either bind to avidin or 
be internalized by the cells, respectively (Figure 8 ). The monoclonal antibody 
and biotin or TAT has been attached on pNP – PEG – PE. The produced mAb DDS 
(drug delivery system) demonstrated clear immunoreactivity toward the antigen. 
However, some affi nity decrease was observed for the antibodies modifi ed with the 
pNP – PEG – PE anchor and incorporated onto the immunoliposomes. Biotin binding 
to avidin was pH dependent with higher retention (75%) at pH 5.0, where the 
shielding PEG molecules were cleaved away. Signifi cant increase of DDS uptake 
by the cells was achieved when TAT was incorporated on the liposome surface at 
pH 5.0. 
An avidin – biotin system has been used to attach antibodies in the bilayer of 
DDSs. Xiao et al. developed a three - step strategy to improve the tumor - to - tissue 
ratio of anticancer agents [184] . Two antibodies specifi c for the CA - 125 antigen that 
is highly expressed on NIH:OVCAR - 3 cells were used. These cells were prelabeled 
with biotinylated anti - CA - 125 antibody and fl uoroscein isothiocyanate (FITC) – 
labeled streptavidin (SAv) prior to administration of biotinylated liposomes. Both 
antibodies were specifi cally bound to the cell surface of OVCAR - 3 cells but not to 
SK - OV - 3 cells, which do not express the specifi c antibody. Antibody biotinylation 
did not affect its immunoreactivity. 
Schnyder et al. explored the targetability of biotinylated immunoliposomes to 
skeletal muscle cell line in vitro [185] . OX26 mAb binds to transferrin receptor and 
is covalently attached to streptavidin by introducing thiol groups using 2 - iminothiolane 
(Traut ’ s reagent). Immunoliposomes consisted of DSPC (5.2 . mol), cholesterol 
a a a a a a a a 
a 
a 
a 
a 
a 
a 
a 
a 
c 
b 
b 
b b 
c 
Targeting by target specific antibody 
and/or long circulation 
Incubation at lowered pH 
Removal of PEG chains 
De-shielding of the “hidden” function 
FIGURE 8 Schematic for design of multifunctional drug delivery system (DDS) that 
includes pH - cleavable PEG - Hz - PE (a), temporarily “ shielded ” biotin or TATp (b), and 
monoclonal antibody (c) attached to surface of DDS via pH - noncleavable spacer. ( Reprinted 
with permission from ref. 183 . Copyright 2006 by the Americam Chemical Society .) 

(4.5 . mol), PEG – DSPE, (0.3 . mol), and linker lipid (bio - PEG – DSPE; 0.015 . mol). 
OX26 mAb – streptavidin was added to preformed liposomes in a 1 : 1 ratio. According 
to estimations, the average number of bio - PEG – DSPE molecules was 30, assuming 
that one 100 - nm liposome contains 100,000 phospholipid molecules. Uptake 
experiments with muscle cell line using the OX26 mAb, fl uorescence - labeled OX26 – 
streptavidin, or fl uorescent OX26 – immunoliposomes demonstrated cellular uptake 
and accumulation within an intracellular compartment of the OX26 mAb and its 
conjugates. 
All the methods described earlier consider that coupling of the ligand on the 
anchor already existed on the liposome surface. Another option is the ligand – 
anchor conjugate in the form of micelles to mix with the liposomes. According to 
that, anti - CD19 mAb was thiolated using Traut ’ s reagent and reacted with Mal - 
PEG – DSPE in a micellar form with PEG – DSPE and molar ratio 4 : 1 [186] . Antibodies 
were coupled at the end of the PEG – DSPE. Consequently, micelles were 
incubated with preformed liposomes at molar ratio 0.05 : 1, respectively, for 1 h 
at 60 ° C. 
In another study, mAb 2C5 with nucleosome - restricted specifi city, which recognizes 
specifi cally human brain tumor cells, was tested as a potential ligand candidate 
for liposome targeting to brain tumor cells [187] . The mAb was attached to pNP – 
PEG – DSPE and the formed micelles incubated with preformed liposomes. The 
100 – 200 mAbs bound per single liposome of approximately 200 nm in diameter. A 
slight reduction in immunoreactivity was observed for a single antibody molecule 
for a number of reasons; the overall evaluation was suffi cient target recognition and 
affi nity due to multipoint attachment of immunoliposomes to the target via several 
antibody molecules. Indeed, the immunoliposomes showed threefold higher accumulation 
in the tumors compared to nonspecifi c carriers. 
At this point it has to be emphasized that this method seems to be the most 
advantageous one because damaging chemical reactions are excluded as they happen 
at a different stage. Also, this method provides the fl exibility of attaching a large 
variety of ligands on liposomes of various compositions loaded with different drugs. 
Apparently, targeted liposomes produced with this last technique have shown similarities 
in the in vitro drug leakage, cell association, and therapeutic effi cacies to 
liposomes made by conventional coupling procedures. 
Noncovalent Binding of Ligands According to this procedure, the ligand is added 
to the lipid mixture during liposome preparation. Small molecules such as sugars 
have been attached on the liposome or lipoplex surface in this way. At fi rst galactose, 
mannose, and fucose were modifi ed to Gal – C4 – Chol, Man – C4 – Chol, and Fuc – C4 – 
Chol and they were added in the lipid mixture of DSPC/Chol/Sugar – C4 – Chol with 
a ratio 60 : 35 : 5. Chol was chosen due to the stability in the liposomal membrane 
while only one sugar was conjugated so the lipophilicity of the fi nal glycolipid would 
not be altered considerably, and thus the stability of the latter in the liposomal 
membrane would be more secure [188] . After in vivo administration of 0.5% Gal, 
Man, and Fuc liposomes it was found that the ratio of their uptake from parenchymal/
nonparenchymal liver cells was 15.1, 0.6, and 0.2, respectively. Also, in high 
doses, 5% Gal liposomes are taken up by nonparenchymal liver cells as well as the 
parenchymal ones, while they are capable of inhibiting the uptake of Fuc liposomes 
by nonparenchymal cells. 
LIPOSOME STRUCTURE AND CHARACTERISTICS 465

466 LIPOSOMES AND DRUG DELIVERY 
Even if this is a simple and mild technique, there is always a concern about ligand 
orientation, very low attachment effi ciency achieved (4 – 40%), and the liposome 
aggregation often observed. 
5.3.3 IN VIVO DISTRIBUTION 
Successful treatment depends not only on the formulation characteristics but also 
on the route of administration. For example, the schistosomicidal drug tartar emetic 
incorporated in PEGylated liposomes was delivered either intraperitoneally or 
subcutaneously (27 mg Sb/kg) to mice infected with Schisostoma mansoni [189] . 
Indeed, 82 and 67% reduction levels of worm were obtained, respectively. However, 
the effi cacy of the formulation given by either administrative route was not signifi - 
cantly different. The only difference was the slower liposome absorption by the 
subcutaneous route. 
Also, the therapeutic effect of liposomal adriamycin (PC/Chol, 120 nm) was 
enhanced signifi cantly after concurrent i.v. and s.c. administration to rabbits bearing 
VX2 tumors in the mammary gland [190] . The i.v. route signifi cantly inhibits breast 
tumor and metastasis, while the s.c. route acts on local - regional lymph nodes. That 
was proved by slowed growth rates, decreased messenger ribonucleic acid (mRNA) 
expression of proliferating cell nuclear antigen, and extensive necrosis and apoptosis 
of tumor cells. Even if allergic reactions have not been reported after s.c. injection 
of liposomes, there is more to be done on systemic toxicity. 
The therapeutic effi cacy of paclitaxel is stronger after drug incorporation in magnetoliposomes 
injected either i.v. or s.c to an EMT - 6 breast cancer mouse model 
[191] . The carrier manipulation due to the application of a magnetic fi eld led to their 
increased accumulation to the tumor site. However, paclitaxel accumulation is 
slightly lower after s.c. administration, probably attributed to time delay during the 
drug transportation process to the circulation. 
In another study by Wang et al., i.v. injection of liposomes carrying rat insulin 
promoter (RIK) – thymidine kinase (TK) was found to be less toxic to the liver than 
the i.p. injection of the same formulation to severe combined immunodefi cient mice 
(SCID) [192] . The direct injection of the liposomes to abdominal cavity probably 
leads to higher local absorption and, thus, higher liver toxicity. In contrast, the i.v. 
injected volume is smaller (70 . L to 100 . L of i.p. injection), which is diluted fast as 
soon as it enters the body. 
5.3.3.1 Conventional Liposomes 
Conventional liposomes are those that do not carry any sterically stabilizing or targeting 
moieties on their surface. Their biodistribution depends strongly on their 
physicochemical properties (size, . potential, composition) and physiological and 
pathological conditions of the body [193] . Thus, conventional liposomes comprise 
the passive targeting of drug molecules. 
Intravenous Administration Liposomes administered intravenously face barriers 
such as the endothelial lining of the vasculature and the blood – brain barrier. Extravasation 
of the liposomes occurs only in organs such as liver, spleen, and bone 

marrow (due to leaky fenestrae and loose junctions between the endothelial cells) 
and under certain pathological conditions (presence of tumors, infl ammation, infection). 
Besides, neutral (uncharged) liposomes of size smaller than 100 nm show slow 
blood clearance compared to others of larger size and/or positive or negative charge 
(due to presence of opsonins) [194, 195] . Also, lipid exchange between liposomal 
carriers and plasma lipoproteins contributes to liposomal membrane rupture and 
consequently loss of the therapeutic substance [196] . 
Therefore, conventional liposomes are used mostly in treating the RES system or 
to mask the toxic side effects of anticancer drugs. Many anticancer drugs entrapped 
in liposomes have shown altered biodistribution and reduced toxicity. Plain SUV 
liposomes consisting of DSPC/Chol (known as “ Stealth ” ) in a molar ratio 2 : 1 or 1 : 1 
have shown particularly promising vehicles as reported in a number of studies of 
animal models. They have undergone preclinical and clinical studies due to relatively 
low levels of RES uptake and the high level of tumor targeting exhibited [197 – 200] . 
Indeed, 111 In - labeled DSPC/Chol liposomes have proven capable of targeting a 
number of tumors [201 – 206] . Although the signifi cant RES uptake was a fact, about 
45 – 50% of liposomes remained in the blood circulation 4 h after i.v. injection. Positive 
images on gamma camera were reported. In patients with recurrent high - grade 
gliomas, 1% of the injected liposomal dose was taken up by the tumor [205] . 
Moreover, anthracyclines have been formulated in conventional liposomes and 
are commercially available. Doxorubicin for i.v. use is commercially available in the 
form of Myocet, which consists of egg PC and cholesterol [207] . It is recommended 
in combination with cyclophosphamide for metastatic breast cancer. Thus, drug 
entrapment in liposomal vesicles may reduce the incidence of cardiotoxicity and 
lower the potential for local necrosis, but infusion reactions, sometimes severe, may 
occur. Hand – foot syndrome (painful, macular reddening skin eruptions) occurs 
commonly with liposomal doxorubicin and may be dose limiting. Daunorubicin also 
has general properties similar to those of doxorubicin. A liposomal formulation 
(DSPC/Chol, 2 : 1, size 45 nm) for i.v. use is licensed for patients with AIDS - related 
Kaposi ’ s sarcoma [207, 208] . The plasma pharmacokinetics of DaunoXome differs 
signifi cantly from the results reported for free daunorubicin hydrochloride. 
DaunoXome has a small steady - state volume of distribution of 6.4 L (probably 
because it is confi ned to vascular fl uid volume) and clearance of 17 mL/min. These 
differences in the volume of distribution and clearance result in a higher daunorubicin 
exposure [in terms of plasma “ Area on the curve ” or “ bioavailabity ” (AUC )] 
from DaunoXome than with free daunorubicin hydrochloride. The apparent elimination 
half - life of DaunoXome (daunorubicin citrate liposome injection) is 4.4 h, 
far shorter than that of daunorubicin, and probably represents a distribution half - 
life. Although preclinical biodistribution data in animals suggest that DaunoXome 
crosses the normal blood – brain barrier, it is unknown whether DaunoXome crosses 
the blood – brain barrier in humans [207] . 
In addition, TAS - 103 (a novel quinoline derivative, topoisomerase inhibitor) 
incorporated in DPPC/Chol (2 : 1) liposomes (size < 80 nm) enhanced the survival 
time of mice with Lewis lung carcinoma to 42 days in comparison to the 38.6 days 
of those treated with free TAS - 103 [209] . The increases in lifetime were 45 and 58% 
for the free TAS - 103 and liposomal TAS - 103, respectively. 
In another study, the antibiotic cefoxitine was incorporated in DMPC/Chol (2 : 1) 
liposomes prepared using the reverse - phase evaporation technique in order to 
IN VIVO DISTRIBUTION 467

468 LIPOSOMES AND DRUG DELIVERY 
increase the effi cacy of the drug characterized by a short half - life and poor intracellular 
diffusion [210] . It was shown that the cefoxitin levels achieved in liver and spleen 
5 h postinjection were 6 - and 16 - fold higher than those observed after administration 
of free antibiotic. Also, the elimination rate through the kidney was slower. 
Intraperitoneal Administration Intraperitoneal administration has the biological 
and pharmacological advantage of creating direct exposure of the tumor, infection, 
or infl ammation to the therapeutic agent. This drug delivery method increases the 
dose intensity within the peritoneal cavity [211 – 213] . Intraperitoneal administration 
of liposomal formulations of anticancer drugs is preferred to the i.v. one, due to the 
higher drug accumulation in the tumors and lower drug plasma concentration minimizing 
drug toxicity [214 – 216] . 
Size, liposomal composition, charge, drug density in the liposomal membrane, 
and preparation method are some of the important parameters which need to be 
considered carefully to design an effi cient DDS. Sadzuka et al. assessed DOX - 
incorporating liposomes made by either DMPC or DSPC of a variety of sizes (150, 
600, and 4000 nm) and surface charge (positive and negative) on the therapy of solid 
tumors and peritoneal dissemination in Ehrilch ascites carcinoma - bearing mice 
[214] . When using small negative liposomes, lipid composition did not affect the 
clearance or stability of liposomes in the abdominal cavity. However, for neutral 
liposomes, DSPC ones were found more effective for the treatment of the solid 
tumor due to the higher stability of those liposomes in comparison to DMPC ones. 
Thus DSPC exhibited longer plasma circulation. As for the effect of surface charge, 
the positive vesicles were cleared faster from the abdominal cavity until 1 h postinjection 
and then showed a slower clearance rate until 48 h, in opposition to the 
negative ones. Larger particles were found in abundance in the peritoneal cavity 
and stayed longer there, inducing toxicity due to liposomal membrane disruption 
and release of the anticancer drug. Overall, it was concluded that the larger liposomes 
were effective against peritoneal dissemination and the smaller ones against 
the solid tumor. 
The same author evaluated the method of preparation by using DOX - 
encapsulating liposomes on the peritoneal dissemination of tumor in Ehrlich ascites 
carcinoma - bearing mice [217] . The liposomal carriers were made either with the 
method of Bangham et al. [1] (BLDOX), the pH gradient [144] (PLDOX), or the 
gelation method (GLDOX) [147] . It was shown that survival in the BLDOX group 
was signifi cantly prolonged compared to that in the DOXsol (DOX as solution) 
group, while there was no effect on survival of the GLDOX group. BLDOX liposomes 
appeared to be less stable and released DOX in a higher degree than the other 
formulations. The latter seems to be of high importance due to increased DOX level 
in the abdominal cavity and enhancement of drug effi ciency for the local therapy. 
Also, positive outcome was achieved after i.p. administration of the liposomal 
formulation of an l - dopa prodrug derivative to rats [218] . It was shown that the 
level of dopamine in rat striatum was 2.5 - fold higher to what was obtained after 
i.p. administration of l - dopa or the free prodrug itself. 
Subcutaneous Administration Liposomes given s.c. aim to target the lymphatic 
system for imaging, distribution of therapeutic agents, or vaccination [219, 220] . 
According to Oussoren and Storm, the determining factors infl uencing lymphatic 

absorption are liposome size and site of injection [219] . Liposome charge, composition, 
or PEG coating does not have a signifi cant effect on the fate of the liposome 
trip in the lymphatic system. 
Liposomes injected s.c. that do not enter the bloodstream either enter the lymphatic 
capillaries or stay at the site of injection. In the fi rst case, 1 – 2% of the injected 
liposomal formulation is captured by the lymphatic nodes 12 h postinjection. 
However, this depends on liposome size. Neutral vesicles smaller than 100 nm pass 
through the interstitium and then to the lymphatics a lot easier than the larger particles. 
Drug carriers remaining at the site of injection will release the entrapped 
molecule. Often, 40% of the injected dose of small liposomes (about 70 nm) is 
retained at the injection site. Therefore, liposome surface modifi cation was attempted 
using non specifi c human antibodies and saccharides. Only saccharide - modifi ed liposomes 
enhanced absorption from the injection site and enhanced lymph node uptake 
was in comparison to control liposomes [221] . Also, the specifi c site of injection is 
very important and species dependent. Taking the rat as animal model, s.c. injection 
in the dorsal foot or the footpad results in higher liposome uptake by the lymph 
nodes, in contrast to the fl ank as an injection point. 
As mentioned earlier, the s.c. route for delivery of anticancer agents could prevent 
the metastatic spread of tumors that occurs often through the lymphatic system. 
However, a number of limiting factors, such as incomplete absorption of drug - 
loaded liposomes, which would increase, for example , the toxicity of the released 
drug at the surrounding tissue and the development of tumors in the regional lymph 
nodes could limit the therapeutic potential of liposomes. 
In addition, for imaging studies, only liposome - encapsulated gadolinium was used 
successfully [222, 223] . In a more recent study, electron spin resonance (ESR) was 
applied successfully to investigate the integrity of MLV and the possibility of a depot 
effect after the s.c. injection in mice [224] . 
Also, Gregoriadis et al. evaluated the type and degree of immune response after 
s.c. injection of ovalbumin (OVA) – encoding plasmid DNA (2.5 or 10 . g) either 
alone or in liposomes, in mice [220] . Anti - OVA serum antibody titers were detected 
in animals immunized with 10 . g of liposomal DNA (after a single dose of antigen) 
and with both 2.5 and 10 . g of liposomal DNA (after two doses of antigen) [225] . 
However, the anti - OVA response was not detected using the DNA alone. 
Similarly, signifi cantly higher humoral responses were obtained after s.c. administration 
of either a lipid and/or a nonionic - based vesicle - entrapped plasmid for the 
nucleoprotein of H 3 N 2 infl uenza virus in comparison to the naked DNA alone 
[226] . 
5.3.3.2 Long - Circulating or PEG ylated Liposomes 
The liposome biodistribution profi le changes signifi cantly when the vesicle surface 
[227 – 229] is coated with polymers, usually PEG. Longer blood circulation, lower 
liver uptake, and higher accumulation in tumors have been reported. The presence 
of the hydrophilic groups of PEG on the liposome surface provides electrostatic 
and steric repulsion between PEG - grafted liposomes. PEG molecules neutralize the 
surface charge of vesicles and thus prevent their opsonization. Also, liposome opsonization 
is reduced due to inability of opsonins to bind to hydrophilic surfaces. 
Moreover, the thickness of the PEG layer infl uences the interaction of the liposomes 
IN VIVO DISTRIBUTION 469

470 LIPOSOMES AND DRUG DELIVERY 
with the macrophages. The thickness of the PEG layer depends on the PEG molecular 
weight and the amount (%) incorporated in the liposomal composition. For 
example (Figure 9 ), the fast clearance of positively charged stearylamine liposomes 
can be reversed by attachment of 6% mol of PEG with molecular weight of 750 or 
5000 kDa [230] . In the case of phosphatidic acid – containing liposomes, only PEG 
5000 can prolong blood circulation while phosphatidylserine - containing liposomes 
are eliminated fast due to the insuffi cient effect of 6% PEG 750 or 5000. 
However, an optimum level of PEGylation (PEG 2000 kDa) was estimated for 
PC (1.85 mol %)/Chol (1 mol %) liposomes as to the effect on blood circulation 
[231] . As reported, after 5 mol % of PEG incorporation the accumulation in the liver 
was signifi cantly decreased, while the minimum uptake by the spleen was achieved 
with 9.6 mol % of PEG insertion. The same authors showed that as the PEG amount 
grafted increased, the liposome accumulation in the heart increased. But above a 
9.6 mol % of PEG, the circulation time was slightly decreased in blood and was 
increased in the liver and spleen. The uptake of liposomes by RES was even higher 
when 13.7% of PEG was present on the vesicle surface. As shown by another group, 
the optimum PEG amount required for liposome stabilization and prolonged half - 
life was 5 – 10 mol % DSPE – PEG 2000 [232] . Higher amounts than that led to disruption 
of the vesicles. 
Intravenous Administration The effect of PEG on liposome biodistribution presented 
in the previous paragraph is for i.v. administration of liposomes. Several 
recent examples for the effect of PEG coating of liposomal drug formulations on 
the biodistribution (and thus pharmacological outcome) for different drugs follow. 
DOX concentrations were estimated in various organs after i.v. administration of 
DOX - loaded liposomes (PC/Chol/PEG2000, molar ratio 55 : 40 : 5) in xenograft 
tumor - bearing mice [233] . Obviously, the encapsulation of DOX in conventional or 
PEGylated liposomes reduced the drug concentration in liver, heart, kidney, and 
stomach compared to the drug solution and prolonged the circulation half - life to 
46.09 h, in contrast to 26.04 and 23.72 of conventional and free DOX, respectively. 
However, PEGylated DOX showed higher antitumor activity in comparison to that 
entrapped in conventional liposomes. In comparison with free DOX, the inhibition 
rate of both liposomal formulations was higher. Doxil is a commercially available 
formulation of DOX entrapped in HSPC/Chol/mPEG liposomes [234] . Thereafter, 
PEGylated liposomes are in common use in many applications. Covalent attachment 
of specifi c molecules (folate) at the free end of PEG molecules results in 
liposomes able to be recognized by specifi c receptors (folate receptors on cancer 
cells). There are a huge number of research papers on the active targeting of modi- 
fi ed PEGylated liposomes [235 – 237] with very promising results. 
Intraperitoneal Administration The potential of PEGylated liposomes administered 
intraperitoneally has been evaluated for cancers located in the peritoneal 
cavity. For example, Syrigos et al. [238] studied the biodistribution of indium ( 111 In) – 
labeled PEGylated liposomes [Hydrogenated soya PC (HSPC)/Chol/PEG – DSPE ] 
compared to free 111 In via either i.p. or i.v. route to non - tumor bearing mice . The 
AUC of In - PEG liposomes was 74 - fold higher than that of free indium. The relative 
ratio of the AUCs (RR - AUCs) for i.p. versus i.v. administration for peritoneum was 
1.36 [423.6 vs. 311.3% individual dose (ID) g/h]. The blood AUC values for i.p. and 

FIGURE 9 ( a ) Clearance from circulation and ( b ) accumulation in liver and spleen of 
liposomes composed of lecithin (LS) mixed or not with charged lipids (PS = phosphadityl 
serine, PA = phosphatidic acid, SA = stearylamine) and bearing or not, a surface coating with 
PEG molecules after IV administration (PEG - 750 and PEG - 5000 = polyethylene glycol with 
molecular weights 750 and 5000). ( Reproduced from ref. 230 with permission of Elsevier .) 
100 
80 
60 
40 
20
0 
100 
80 
60 
40 
20
0 
100 
80 
60 
40 
20
0 
100 
80 
60 
40 
20
0 
100 
80 
60 
40 
20
0 
100 
80 
60 
40 
20
0 
0 60 120 180 240 300 360 
0 60 120 180 240 300 360 0 60 120 180 240 300 360 
0 60 120 180 240 300 360 0 60 120 180 240 300 360 
0 60 120 180 240 300 360 
Time, min Time, min 
Time, min Time, min 
Time, min Time, min 
% injected dose/g 
% injected dose/g % injected dose/g 
% injected dose/g % injected dose/g 
% injected dose/g 
Plain LS 
Plain LS 
LS + PEG-750 
LS + SA + PEG-750 
LS + SA + PEG-5000 
LS + PEG-5000 
LS + PS 
LS + PA 
LS + PA 
Plain LS 
LS + SA + PEG-750 
LS + SA + PEG-5000 
LS + PA 
Plain LS 
LS + PA + PEG-750 
LS + PA + PEG-5000 
LS + PA 
Plain LS 
LS + PA + PEG-750 
LS + PA + PEG-5000 
LS + PA 
LS + SA 
Plain LS 
LS + PEG-750 
LS + PEG-5000 
LS + PS 
LS + PA 
LS + SA 
(a) (b) 
100 
80 
60 
40 
20
0 
100 
80 
60 
40 
20
0 
0 60 120 180 240 300 360 0 60 120 180 240 300 360 
Time, min Time, min 
% injected dose/g 
% injected dose/g 
Plain LS 
LS + PEG-750 
LS + PEG-5000 
LS + PS 
Plain LS 
LS + PS + PEG-750 
LS + PS + PEG-5000 
LS + PS 
IN VIVO DISTRIBUTION 471

472 LIPOSOMES AND DRUG DELIVERY 
i.v. administration were essentially the same (RR - AUC 1.03; 453.7 vs. 439.2% ID 
g/h) 18 h postinjection despite the delayed absorption of the liposomes from the 
peritoneal cavity. However, the relevant values for organs in the peritoneum were 
higher in case of i.p. administration. An increase in the range 1.2 – 5.1 was seen for 
organs such as stomach, pancreas, ileum, colon, gallbladder, ovary, and adrenal 
glands. This is an advantage compared to the i.v. administration because the drug 
can target both the primary site and any peritoneal deposits. The encapsulation of 
doxorubicin and cis - platin (small molecules with high toxicity and short half - lives) 
in PEGylated liposomes might increase the retention from the peritoneal cavity and 
reduce the drug toxicity. 
Another study points out the vesicle size rather than the presence of PEG as a 
more determining factor to successfully tackle peritoneal cancers [239] . According 
to this study, the synergistic effect of Doxil after coadministration of PEG – SUV – 
interleukin - 2 (IL - 2) or MLV – IL - 2 via either the i.p. or i.v. route to mice bearing 
M109 lung adenocarcinoma was studied. The cancer was inoculated i.p., resulting in 
multiple i.p. masses. Small PEGylated liposomes as vehicles for IL - 2 for systemic 
treatment of metastatic lung cancer boosted the antitumor effect of Doxil to the 
same level achieved with soluble IL - 2. In contrast, in the regional model, the most 
effective combination was Doxil with MLV – IL - 2 liposomes. This is attributed to the 
retention and slow release of IL - 2 in the peritoneal cavity due to the inability of 
MLVs to enter the circulatory system or the draining lymph vessels, whereas IL - 2 
in small liposomes or in soluble form escapes rapidly from the peritoneal cavity. 
Another possibility could be the enhanced immunostimulation results from the 
uptake of MLV – IL - 2 by peritoneal macrophages as opposed to the stealth properties 
of PEGylated SUV – IL - 2. 
Subcutaneous Administration As for the use of PEGylated liposomes subcutaneously, 
there is not much reported. In one paper the infl uence of the administration 
either s.c. or i.m. of mitoxantrone - loaded liposomes was studied. It was reported 
that mitoxantrone showed reduced irritation when the formulation was administered 
i.m. rather than s.c. However, when PEG was incorporated on the liposome 
surface, there was no apparent protective effect of the liposomes [240] . 
5.3.3.3 Other Routes of Administration 
In general, the administration route plays an important role on the impact of the 
therapeutic treatment, and it is chosen according to the kind or purpose of the 
treatment (local or systemic), toxicity, and accessibility of the diseased area. In this 
section specifi c characteristics of other routes of administration that are currently 
receiving attention will be emphasized and the most recent developments with 
respect to liposomal drug applications will be presented. 
Pulmonary Drug Delivery Pulmonary epithelium offers many advantages for 
drug delivery due to easy access and the large surface area provided by alveoli [241] . 
Also, macromolecules can penetrate the lungs much easier and faster than other 
noninvasive routes avoiding the fi rst - pass meabolism; therefore, many promising 
applications are being considered for the delivery of proteins and peptides. For a 
drug or drug delivery system to reach the lungs successfully, it has to be aerosolized 
with optimum aerodynamic particle diameter between 1 and 3 . m. The latter, in 

combination with how the patient inhales determines if the drug particles deposit 
primarily in the conducting ways or in the alveoli. 
Pulmonary delivery of liposomes has focused on the treatment of asthma, 
infectious diseases, genetic diseases (cystic fi brosis), and lung injury and lately on 
gene therapy. 
Special attention has been paid on the physical characterization of liposome 
aerosols [242 – 244] , including dry powder formulations [245] , and cationic liposome 
DNA complexes [246, 247] . Corticosteroid therapy using liposomal formulations has 
focused on the development of aerosols containing beclomethasone dipropionate, 
triamcinolone acetonide, and triamcinolone acetonide phosphates [248 – 251] . Aminoglycosides 
have been considered as good candidates for pulmonary delivery 
because of their potency and their ability to directly target the lungs. When amikacin 
was encapsulated in liposomes [fl uid or rigid state (chol containing)] and administered 
in sheep, the drug mean residence time (MRT) increased 5 times compared 
to the instilled solution (rigid liposomes gave 2 times higher MRT compared to fl uid 
ones) [252] . In a similar study, prolonged retention of liposome - encapsulated tobramycin 
was reported after administration in Pseudomonas aeruginosa – infected rat 
lungs [253] . Liposomal formulation of antioxidants has also been investigated for 
pulmonary delivery [254] . Liposomes containing . - tocopherol prolonged the residence 
time of radioactively labeled glutathione [255] . Paraquat poisoning, which 
causes extended damage to lung tissues, was attenuated after pretreatment with . - 
tocopherol - containing liposomes [256] . Liposomes containing CuZn/superoxide 
dismutase and catalase were found to protect the lungs of premature rabbits when 
exposed to hyperoxia [257] . 
Drugs for the treatment of infections such as aspergillosis, tuberculosis, and anticancer 
therapy have been formulated in liposomes and tested in vivo by administration 
via the respiratory system [258 – 260] . More specifi cally, rifampicin used to treat 
pulmonary tuberculosis associated with AIDS has been incorporated in MLV liposomes 
(PC/Chol, 7 : 3 molar ratio) with the aim of increasiing its effi cacy to macrophages 
and reduce side effects [258] . The rifampicin retention was found higher in 
PC/Chol/DCP (7 : 3 : 0.1) liposomes tested due to electrostatic interaction between 
DCP and the drug. Both liposomal formulations showed greater accumulation in 
the lungs when compared to the controls. In the case of free drug and after 0.5 h 
postadministration, only 39.12% of the administered dose was retained in the lungs 
and 29.84% of it was found in the serum. No drug was estimated in the lungs 24 h 
later. PC/Chol MLVs demonstrated higher lung accumulation (49.03%) after 0.5 h, 
but the overall distribution pattern was not much different to that of the free drug 
in solution with no drug estimation at the 24 - h time point. It seems that they were 
rapidly passed to the systemic circulation and to the RES organs (liver and spleen). 
The negatively charged MLVs showed even higher drug accumulation (53.86% of 
the administered dose) while 4.14% of it was still present after 24 h. This is attributed 
to the interaction of the negatively charged liposomes with the scavenger receptors 
on alveolar macrophages [261] . Also, it has to be mentioned here that the ligands 
maleylated bovine serum albumin (MBSA) and O - steroyl amylopectin (O - SAP) 
were incorporated on the liposome surface because they are recognized by the 
scavenger receptors of the macrophages. The lung accumulation levels were 61.49 
and 65.14% of the administered dose, respectively, after 0.5 h, while after 24 h they 
were 8.12 and 10.75%, respectively. The relative lung retention of the various 
formulations after 6 h of administration was 1.3 times for PC/Chol MLVs, 3.4 times 
IN VIVO DISTRIBUTION 473

474 LIPOSOMES AND DRUG DELIVERY 
for PC/Chol/DCP MLVs, 4.53 times for MBSA - PC/Chol, and 4.76 for O - SAP – PC/ 
Chol in comparison to plain drug solution administered by aerosolization. 
The same group (Vyas et al.) studied the impact of amphotericin B entrapped in 
liposomes (MLVs: PC/Chol, 7 : 3 molar ratio) in the absence or presence of ligands 
[ O - palmitoylated mannan (OPM) or O - palmitoylated pullulan (OPP)] on the vesicle 
surface as potential use for the treatment of aspergillosis [259] . Optimized formulation 
was the one of PC/Chol 7 : 3 molar ratio with the production of spherical MLVs 
of approximate mean vesicle size 2.56 . m and maximum entrapment effi ciency 
78.2%. Again the lung uptake of the ligand - appended liposomes was higher 
compared to the plain liposomes. The lung accumulation levels of OPM - and 
OPP - coated liposomes were 58.12 and 55.02% of the administered dose 5 h postadministration, 
respectively, while 24 h later the relevant lung retentions were 11.23 
and 9.86%, respectively. 
Methotrexate (MTX) (a folic acid antagonist) was entrapped into liposomes 
(PC/PI/Chol 2 : 1 : 1 molar ratio) so as to reduce nephrotoxicity and investigate the 
pharmacokinetics of the liposomal MTX [260] . Indeed, the latter showed increased 
MTX retention in the lungs while the biodistribution in spleen and kidney was less 
than that obtained with free MTX. Similar results were obtained by liposomalization 
of various anticancer drugs elsewhere [262 – 264] . For example, cytosine arabinoside 
was administered intratracheally to rats in the free or the liposomal form 
[262, 263] . Liposomal drug was effective into the lung but not other tissues, contrary 
to the free drug. Moreover, the antitumor properties of the anticancer 9 - 
nitrocamptothecin (9NC) after its liposomalization were tested in three different 
human cancers xenografted s.c. in mice as well as murine melanoma and human 
osteosarcoma pulmonary metastases [264] . The liposomal form of anticancer drug 
inhibited the growth of subcutaneous tumors and metastatic pulmonary cancers 
given via the respiratory system. Intramuscularly administered liposomal drug 
exhibited some anticancer activity, but that achieved using the aerosol was superior. 
Thus, the liposomal 9NC aerosol was proved to be of high potential for the treatment 
of cancers throughout the body. 
Recently the gene therapy of pulmonary diseases using liposomal formulations 
has attracted a lot of attention. Transfection of the lungs of animals with aerosolized 
cationic liposome – DNA complexes has been attempted [265, 266] . However, the 
transfection effi ciency was rather low despite the large amounts of DNA used in 
these studies. Cationic liposomes from 2, 3 - dioleyloxy - N - [2(sperminecarboxamido) 
ethyl] - N, N - dimethyl - 1 - propanaminium trifl uoracetate (DOSPA), ( ± ) N - (2 - hydroxyethyl) 
- N, N - dimethyl - 2, 3 - bis(tetradecyloxy) - 1 - propanaminium bromide (DMRIE) 
mixed with DOPE, and DNA at fi xed ratios of DNA/lipid 1 : 4 for DMRIE and 1 : 3 
for DOSPA were tested with two different types of jet aerosols (Aerotech II and 
Puritan - Bennett 1600) [246] . The decrease in transfection activity was gradual with 
Puritan - Bennett 1600, in contrast with Aerotech II, which rapidly lost transfection 
effi ciency. That was attributed to the increased throughput of the Aerotech II, 
resulting in more frequent cycling and therefore damage of the complex. 
The impact of the zeta potential of the formulation was emphasized by Eastman 
et al. [247] . The maximal aerosol transfer effi ciency of cationic lipid/DNA complexes 
was achieved in the presence of a salt concentration of 25 m M . The authors attributed 
that to the fact that the formulation kept its zeta potential between . 40 and 
. 50 mV. As a closing remark, one of the major problems and challenges of aerosol 

delivery is the duration to deliver therapeutic doses of DNA to the lungs. This possibly 
could be overcome with dry powders avoiding volume limitations of aqueous 
dispersions. 
Oral Delivery The destructive effects of the conditions in the gastrointestinal tract 
(GIT), especially due to the presence of bile salts, are known and well established 
[267, 268] . The lipid composition of liposomes determines to a large extent the possibility 
of remaining intact under such conditions [58] . Quadachi et al. attempted to 
provoke IgA response from the M cells of Peyer ’ s patches after oral administration 
of OVA - loaded MLV liposomes made by either soya PC or DSPC to a model of 
hypersensitivity to OVA Balb/C mouse [269] . Clearly, DSPC MLVs were much more 
stable as demonstrated in in vitro stability studies in simulating GIT media. However, 
liposome incorporation of OVA did not cause any signifi cant impact on the reduction 
of hypersensitivity to that obtained with the free allergen. Surprisingly, the 
empty liposomes show some immunoresponse which was attributed to nonspecifi c 
stimulation. 
The biodistribution of novel liposomal - like spherical carriers called Spherulites, 
consisting of PC/Chol/polyoxyethylene alcohol (43 : 4 : 3 w/w/w), was shown to be 
more promising [270] . These were prepared by shearing the phospholipid bilayer 
and labeling with 111 In. Their integrity was demonstrated by an increase in radioactivity 
in the blood 1 h after oral administration to fasted rats, while no increase 
was seen for free label. 
Skin Delivery Dermal delivery of phospholipid - based vesicles fi rst appeared in 
the literature in the early 1980s [271, 272] . Liposomes can play a dual role after their 
application to the skin: retention (and perhaps protection) of the active compound 
across the stratum corneum and acting as a penetration enhancer. The composition, 
size, and vesicle surface charge are parameters that can infl uence the transport rate 
of drugs contained in liposomal formulations across the skin [273] . Regarding their 
composition, liquid - state liposomes resulted in higher skin permeation rates compared 
with gel – liquid liposomes for progesterone and Triamcinolone (TRMA) [274, 
275] . Moreover skin lipid liposomes have provided higher drug disposition in the 
deeper layers of the skin for corticosteroids and acyclovir [276, 277] . Neverthless, in 
another study [278] higher amounts of acyclovir in the skin were delivered from 
conventional lipid liposomes compared with liposomes containing skin - based lipids. 
These studies emphasize that a careful design is needed to defi ne optimal compositions, 
depending also on the specifi c objective which may be either dermal (topical) 
delivery of the liposome - encapsulated drug (which is easier since only increased 
retention of the drug in the skin is desired) or transdermal delivery (systemic 
absorption of the drug), which is more complicated, and in general deeper penetration 
of the liposome carrier in the skin is required. What is also important as regards 
the fi nal outcome (especially when transdermal delivery is the objective) is the type 
of fi nal formulation with respect to the conditions applying, occlusive or 
nonocclusive. 
Amphotericin B encapsulated in charged liposomes demonstrated 10 - fold higher 
transport rates across the skin compared with neutral liposomes [279] . In a similar 
manner, the retention of acyclovir from positively charged vesicles was much higher 
compared with other formulations [277] . This could be attributed to the attraction 
IN VIVO DISTRIBUTION 475

476 LIPOSOMES AND DRUG DELIVERY 
between the positively charged liposomes and the negative charge of the skin [277] . 
The pore size of the skin is approximately 0.3 nm; however it can be opened up to 
40 nm without signifi cant damage to its structure. To some extent, vesicles can transport 
across the skin via the follicular and transcellular routes. Therefore the size of 
the vesicles used for transdermal delivery is a crucial parameter for the overall 
performance of the formulation [280 – 282] . Several studies have also emphasized 
the impact of the size of the liposomes on the transport rate of active compounds 
across the skin [277, 280, 283, 284] . Biologically active macromolecules, including 
superoxide dismutase [285] and interferon - . [286] , have been successfully applied 
to skin in liposomal formulations. Generally it is believed that the main pathway 
for transdermal delivery of active compounds is either intercellular or paracellular. 
However, the appendage transport (e.g., follicular route) has attracted a lot of interest 
lately. The pilocebaceous units (hair follicle, hair shaft, sebaceous glands) can be 
used for drug targeting. Combinations of liposomes with DNA [287, 288] and monoclonal 
antibodies [289] have demonstrated that liposome composition, hair structure, 
and hair cycle play signifi cant roles in the transfection of human hair follicles. 
Two different mechanisms have been suggested for the incorporation of active 
compounds to the hair shaft: (1) direct permeation of the vesicular formulations 
with the active compound to the hair shaft and (2) incorporation of the active compounds 
in the follicular matrix cells and then into the hair shaft as the matrix cells 
develop and differentiate into new hair shafts [290] . 
Recently, for the transdermal delivery of drugs using carrier systems, attention 
has been focused on the development of transformable [284, 285] or elastic vesicles 
[12] . These vesicles are liposomes that contain surfactants or in general “ edge activators 
” in addition to phospholipids in their lipid membranes (Figure 10 ), a fact that 
Double-chain nonionic surtactant or lipid 
Emphiphilic or lipophilic drug 
Charged hydrophilic drug 
Single-chain surfactant 
Hydrophilic drug 
FIGURE 10 Conventional liposomes/elastic vesicles. Charged hydrophilic, amphiphilic, 
and lipophilic drug molecules can be associated with the bilayers of the vesicles, whereas 
hydrophilic substances can also be entrapped in the vesicles. Rigid vesicles consist of double - 
chain nonionic surfactants or lipids in the presence or absence of cholesterol (left image). 
Elastic vesicles consist of double - chain surfactants or lipids and an edge activator. The edge 
is often a single - chain surfactant (right image). ( Reprinted from ref. 273 with permission of 
Elsevier .) 

increases their elasticity (ability to be deformed, without being disrupted, after 
applying pressure on them). Modifi ed liposomes called ethosomes (containing 
alcohol) have shown increased skin permeability. Because of their structure, ethosomes 
are able to encapsulate and deliver through the skin highly lipophilic molecules 
such as cannabinoids, testosterone, and minoxidil as well as cationic drugs such 
as propranolol and trihexyphenidil or even plasmids and insulin [291] . Although the 
mechanism of the increased transdermal delivery of drug molecules still remains a 
controversial issue, mainly with respect to the depth at which intact vesicles can 
travel in the skin, many interesting results are being generated. However, due to the 
limited permeability of the skin membrane, physical enhancement mechanisms, 
including iontophoresis, electroporation, and ultrasound, have been used in combination 
with chemical enhancers (liposomes) to increase skin permeability. One of 
the methods, application of an electric current to the skin, has been shown to 
promote the transdermal transport of drugs by an additional driving force, namely, 
an electrical potential gradient across the skin [292] . Transdermal iontophoretic 
transport of a liposomal formulation across human cadaver skin was fi rst reported 
for [Leu5] enkephalin [293] . Liposomes could penetrate into the skin. Enkephalin, 
when delivered iontophoretically from liposomes carrying positive or negative 
charge on their surface, was found to be the same or less than that of the controls; 
however, the degradation of enkephalin was less in liposome formulations as compared 
to controls, demonstrating that liposomes can protect peptides from the proteolytic 
environment of skin. However, when enoxacin was encapsulated in different 
liposome formulations and was transported (electrically assisted) across the skin, 
the drug transport results showed that the permeability of enoxacin released from 
liposomes was higher compared to that of free drug [294] . In vitro transdermal 
iontophoretic delivery of estradiol from ultradeformable liposomes, saturated 
aqueous solution [295] , and conventional liposomes [296] has demonstrated the 
superiority of ultradeformable liposomes (Transfersomes) due to their lipid composition. 
Liposomal formulations of . blockers were iontophoresced in vivo to hairless 
rats [297] . Skin irritation was signifi cantly reduced when a liposomal formulation of 
the propranolol base was used rather than the base itself, emphasizing another 
important role liposomes could play. When adriamycin was delivered via the hair 
follicles using various liposomes and iontophoresis combined with application of 
ionic liposomes, higher transport rates were obtained with the latter, emphasizing 
their synergistic effect [298] . Moreover skin electroporation was applied to enhance 
gene transfer into subcutaneous MC2 murine breast tumor skin in combination with 
cationic liposomes demonstrating signifi cant transfection improvement [299] . 
However, when electoporation combined with estradiol - loaded liposomes were 
applied to skin, the estradiol skin penetration was not affected signifi cantly. That 
was attributed to the antienhancer or retardant effect of liposomes [300] . In a 
mechanistic study, anionic phospholipids were found to enhance the transdermal 
transport of molecules by electroporation compared to cationic or neutral phospholipids, 
offering new insights to design better enhancers for transdermal drug and 
vaccine delivery [301] . Finally higher transport across the skin obtained after combined 
application of ultrasound and liposomal formulations of diclophenac demonstrated 
a synergistic effect [302] . 
Ocular Delivery Ocular drug delivery has evolved into a great challenge and a 
subject of interest for many scientists with different backgrounds, including medical, 
IN VIVO DISTRIBUTION 477

478 LIPOSOMES AND DRUG DELIVERY 
clinical, pharmaceutical, physical, chemical, biochemical, and toxicological sciences. 
For ailment of eye diseases, topical administration is preferred over systemic in 
order to avoid systemic toxicity, for rapid onset of action, and for decreasing the 
required dose. 
The main route for intraocular absorption is across the cornea [303] . In terms of 
drug delivery, the cornea presents an effective barrier to the absorption of both 
hydrophilic and lipophilic compounds. Actually, the main constraints in topical 
ocular delivery are (i) poor ocular retention of conventional dosage forms [304] and 
(ii) poor corneal absorption. Various approaches have been developed to increase 
the bioavailability and duration of therapeutic action of ocular drugs. One such 
approach is based on the use of drug delivery systems [305, 306] , which provide 
controlled and continuous delivery of drugs and can also provide improved 
(increased) residence time of the drug at the delivery site. Recently, intravitreal drug 
injection has evolved into a preferred administration method for therapy of disorders 
in the posterior segment of the eye [305] . The procedure is associated with a 
high risk of complications, particularly when frequent, repeated injections are 
required. Thus, sustained - release technologies are being proposed, and the benefi ts 
of using colloidal carriers in intravitreal injections are currently under investigation 
for posterior drug delivery. 
Between the different types of particulate drug delivery systems, liposomes offer 
additional advantages for ophthalmic delivery, since they are completely biodegradable 
and relatively nontoxic and thus are well tolerated by the eye [305, 306] . Indeed, 
when using other types of colloidal systems, for example , nanoparticles consisting 
of polyalkyl cyanoacrylate, infl ammation and damage of the corneal epithelium 
have been reported [307 – 309] . Another potential advantage of liposomes is their 
ability to come in intimate contact with the corneal and conjunctival surfaces. This 
results in increased probability of ocular drug absorption [310, 311] . 
The potential of liposomes in topical ocular drug delivery was fi rst exploited in 
the 1980s by a number of research groups [303, 310 – 315] . As an example, higher 
levels of inulin were found in the cornea when it was encapsulated in liposomes as 
compared to its aqueous solution [301, 314] , and this was attributed to the physical 
adsorption of lipid vesicles onto the epithelial surface of the membrane [315, 316] . 
More recently a number of liposomal applications for ocular delivery have been 
under investigation [305, 306] for anterior as well as posterior segment administration, 
as outlined in Table 3 . Indeed, a large number of ophthalmic drugs used in 
cases of ocular surface disorders (such as dry eye syndrome) [317] , keratitis and 
uveitis [318 – 327] , and keratoplasty [328 – 331] have been studied in liposomal form, 
and in most cases the results were promising in terms of drug penetration and retention 
in the various ocular tissues (cornea, sclera, retina, and choroids), following 
subconjunctival administration. In some cases, detectable levels of drugs were found 
in ocular tissues up to 7 days after administration [305, 306] . 
As mentioned above, the ability to adsorb to the cornea and an optimal drug 
release rate have been defi ned as the two liposomal attributes most responsible for 
increasing bioavailability after topical ocular administration. A number of factors, 
including drug encapsulation effi ciency, liposome size and charge, distribution of the 
drug within liposomes, stability of liposomes in the conjunctival sac and ocular 
tissues, their retention in the conjunctival sac, and most importantly their affi nity 
toward the corneal surface and the rate of release of the encapsulated drug, have 

been found to infl uence the effectiveness of liposomes in topical ocular drug delivery 
[310, 323, 332 – 335, 337] . Indeed, liposomal manipulation to increase corneal 
adherence has met with some success [310] . 
Positively charged liposomes seem to be preferentially captured at the negatively 
charged corneal surface as compared with neutral or negatively charged liposomes. 
Aditionally, cationic vehicles are expected to slow down drug elimination by the 
lacrymal fl ow both by increasing solution viscosity and by interacting with the negative 
charges of the mucus [334, 335] . Indeed, positively charged phospholipids 
yielded superior retention of liposomes at the corneal surface in rabbits [336] . 
Schaeffer et al. [310] worked with indoxole and penicillin G and reported that liposome 
uptake by the cornea is greatest for positively charged liposomes, less for 
negatively charged liposomes, and least for neutral liposomes. Positively charged 
unilamellar liposomes enhanced transcorneal fl ux of penicillin G across isolated 
rabbit cornea more than fourfold. Similar results were also obtained by others [336, 
323] . By observing the morphology of corneal surface treated with liposomes, it was 
suggested that positively charged liposomes formed a completely coated layer on 
the corneal surface [323] . These liposomes bind intimately on the corneal surface, 
TABLE 3 Experimental Liposomal Preparations of Drugs for Anterior and Posterior 
Segment Administration 
Drug Class Anterior Segment Drugs Posterior Segment Drugs 
Antibiotics Gentamicin 
Norfl oxacin 
Tobramycin 
Clindamycin 
Gentamicin 
Penicillin 
Antifungals Amphotericin B Amphotericin B 
Antivirals Acyclovir 
Idoxuridine 
Ganciclovir 
Trifl uorothymidine 
(Trifl uridine) 
Steroids Dexamethasone 
Immunosuppressives Cyclosporine 
FK506 (Tacrolimus) 
Cyclosporine 
Antimetabolites 5 - Fluorouracil (5 - FU) 
5 - Fluoroorotate 
5 - Fluorouracil (5 - FU) 
5 - Fluorouridine (5 - FUR) 
Bleomycin 
Cytosine arabinoside 
(Cytarabine) 
Daunomycin 
Daunorubicin 
Etoposide (VP - 16) 
Platelet - aggregating 
agents 
Adenosine diphosphate Adenosine diphosphate 
Photosensitive 
cytotoxic agents 
Verteporfi n (BPD - MA) 
Miscellaneous Dichloromethylene 
diphosphonate (Clodronate) 
Disulfi ram 
Immunoglobulins 
Dichloromethylene 
diphosphonate (Clodronate) 
Source : S. Ebrahim, G. Peyman, and P. J. Lee, Survey of Ophthalmology , 50, 167 – 182, 2005. 
IN VIVO DISTRIBUTION 479

480 LIPOSOMES AND DRUG DELIVERY 
leading to an increase in residence time and therefore to an increase in corneal 
absorption time. 
With respect to vesicle size, larger particles are more likely to be entrapped 
under the eyelids or in the inner canthus and can thus remain in contact with 
the corneal and conjunctival epithelia for extended periods. Indeed, larger liposomes 
have been found to resist drainage at the inner canthus and are more 
bioavailable at the ocular surface [337] . However, for patient comfort, it is considered 
that solid particles intended for ophthalmic use should not exceed 5 – 
10 . m diameter [338] . 
Assessment of ocular irritability of neutral or positively charged liposomes by 
the Draize test, histological examination, and the rabbit blinking test has also 
been reported in the literature [339] . The mean total score (MTS) of the Draize test 
was found to show a slight increase immediately following instillation of liposome 
preparations. However, it did not exceed the “ practically nonirritating level, ” and no 
corneal histological alteration was observed by optical microscopy. Neutral liposome 
preparations were confi rmed to be nonirritating; however, positively charged liposomes 
may cause initial pain or unpleasantness following instillation. Thus, althouth 
positive charge helps in improving the contact time with the cornea, at the same time 
it can lead to irritation. Additionally, the release rate of the drug is found to be more 
in neutral liposomes, while increased liposome size restricts solution drainage, thus 
prolonging contact time of the drug, but it can be increased within the limits of not 
inducing any irritancy. From all the above it is understood that in each case liposome 
properties have to be adjusted for best in vivo therapeutics results. As mentioned 
above, liposome stability is another important factor. Barber and Shek reported that 
increasing the cholesterol content of the liposomal membrane decreased the rate of 
tear - induced release of its contents [340] . 
The use of bioadhesive polymers (e.g., a polyacrylic acid, chitosan, hyaluronic 
acid) to prolong the residence time of an ocular preparation in the precorneal 
region, due to increased formulation viscosity, is another approach which can further 
improve liposomal drug delivery. In this respect “ collasomes ” (liposomes coupled 
to collagen matrices) increased the bioadhesive ability of liposomes, were well tolerated, 
and could be instilled safely and effectively by patients in the same fashion as 
ointments or drops [341] . It was also demonstrated that liposomes coated with collagen 
layer bound to cell monolayer with higher affi nity [342] . Other approaches 
were used to increase the contact time of ocular liposomes, such as the case in which 
prolonged retention of liposomal suspension of oligonucleotide was achieved 
by dispersing liposomes within an in situ gel - forming medium [343 – 345] . Novel 
measures to further enhance adsorption of liposomes and increase penetration 
of the cornea included application of a natural lectin which promoted binding of 
ganglioside - containing liposomes to the cornea [346] . 
Liposomes can also be used as promising dosage forms for topical administration 
of immunosuppressive compounds for the treatment of ocular immune - mediated 
diseases [327] . Indeed, it was found that liposomes containing immunosuppressive 
compound FK506 were effective in delivering signifi cantly higher drug concentrations 
to all ocular tissues and particularly aqueous humor and vitreous humor as 
compared to the oil formulation of the agent. Further, liposomes can be used to 
protect drug molecules from attack of metabolic enzymes present at the tear/corneal 
epithelium interface, as demonstrated for the O - palmitoyl prodrug of tilisolol 
[347] . 

As mentioned above, intravitreal injection of drugs should be used in many cases 
to achieve therapeutic intravitreal drug levels. This is especially true for cases of 
viral retinitis, such as cytomegalovirus (CMV) retinitis and acute retinal necrosis 
(ARN) which require intravitreal injection of antivirals, or for the treatment of 
bacterial and fungal endophthalmitis or proliferative vitreoretinopathy [305] . It still 
remains a controversial issue whether liposomes can reach the retina after intravitreal 
injections and which vesicle physicochemical characteristics should be preferred 
for such formulations. 
In addition to conventional drugs, liposomes were also used for intravitreal 
administration of oligonucleotides in order to treat ocular viral infections such as 
herpes simplex virus or CMV [344] . Antisense oligonucleotides are poorly stable in 
biological fl uids and their intracellular penetration is limited. Hence a system that 
is able to permit a protection of oligonucleotides against degradation and their slow 
delivery into the vitreous should be favorable for improving the therapeutic outcome 
in addition to patient compliance. It was found that lipid vesicles are able to protect 
oligonucleotides against degradation by nucleases [344, 345] . Furthermore, they 
increase the retention time of many drugs in the vitreous. Thereby, the use of liposomes 
for intravitreal administration is a very promising approach. 
Nasal Delivery Liposomes are able to decrease mucociliary clearance in the case 
of nasal administration due to their low viscosity. This is attributed to the incorporation 
of liposomal lipids in the membranes of the nasal epithelial cells, which results 
in the opening of pores in the paracellular tight junctions [348] . MLVs containing 
nifedipine administered via the nasal route could attain a constant plasma level 
[349] . Liposomal formulations of levonorgestrel containing carbopol or chitosan 
demonstrated prolonged contact time with the absorptive surfaces, resulting in 
increased bioavailability of the intranasally administered drug [350] . 
The nasal route may be highly promising for candidate vaccines against potential 
pathogens (and infection - related diseases such as cancers) that utilize this route of 
infection [351, 352] . In addition to the likelihood of increased patient compliance, 
immune responses elicited by nasal administration may be more predictable when 
compared with the vaginal route due to immunological changes in the female reproductive 
tract during the menstrual cycle [352] . Previous work with liposome - 
containing vaccines for nasal delivery demonstrated that liposomes can confer 
adjuvancy to the subunit infl uenza vaccine, but also empty liposomes administered 
48 h prior to immunization resulted in immune stimulation, emphasizing that the 
properties and composition of the liposomes play a signifi cant role facilitating the 
transport of the antigen across the membrane [353] . More recently, several promising 
liposome - based vaccines are being designed and investigated for delivery by the nasal 
route, such as the liposome - encapsulated plasmid DNA - encoding infl uenza virus 
hemagglutinin, which has been reported to elicit mucosal, cellular, and humoral 
immune responses after intranasal administration in Balb/C mice [354] . Additionally, 
nasal immunization studies using liposomes loaded with tetanus toxoid were perfomed, 
and it was found that intranasal administration of liposome - encapsulated 
vaccines can be an effective way for inducing mucosal immune responses [355] . Furthermore, 
intranasal immunization studies have been carried out with liposomes 
containing recombinant meningococcal opacity proteins [356] and with anthrax - 
protective antigen protein incorporated in liposome – protamine particles [357] , both 
with promising results. 
IN VIVO DISTRIBUTION 481

482 LIPOSOMES AND DRUG DELIVERY 
Vaginal Delivery The vaginal route has been under investigation in the last years, 
especially for the topical delivery of drugs that are intended to act in the vagina, as 
contraceptives, microbicides and antibiotics, as recently reviewed [358] . In such cases 
the main advantage of using liposomes would be the controlled and sustained 
release of the drug at the site, which would result in a less frequent drug administration 
and improved patient compliance. 
However, the major limitation of using liposomes topically and vaginally is the 
liquid nature of preparation. Nevertheless, several formulation characteristics should 
be optimized in order to achieve the needed rheological and mucoadhesice properties 
for maximum retention of the delivery system in the vagina. Research for the 
development of liposomal gels (gels that contain liposomes) with the required 
properties is currently ongoing [358 – 360] . 
It has been demonstrated that, by their incorporation in an adequate vehicle, such 
as carbopol resins, the original structure of vesicles is preserved [359] , while liposomes 
are fairly compatible with gels made from polymers derived from such resins. 
A previous study has suggested application of liposomes containing antimicrobial 
drugs for the local therapy of vaginitis [359] , while recently the design and in vitro 
evaluation of bioadhesive liposome gels containing clotrimazole and metronidazole, 
or acyclovir, were carried out [359, 360] . 
Some other applications are arising lately concerning liposomes and vaginal 
administration. As mentioned above (in the nasal administration section), mucosal 
surfaces serve as a gateway to disease. It was recently demonstrated that RNA 
interference can be used to manipulate mucosal gene expression in vivo. Using 
a murine model, it was shown by Zhang et al. [361] that direct application of 
liposome - complexed small interfering RNA (siRNA) mediates gene - specifi c 
silencing in cervicovaginal and rectal mucosa. A single vaginal or rectal administration 
of siRNA targeting hematopoietic or somatic cell gene products reduced 
corresponding messenger RNA (mRNA) levels by up to 90%. Additionaly, liposomal 
siRNA formulations proved nontoxic, did not elicit a nonspecifi c interferon 
response, and provided a means for genetic engineering of mucosal surfaces in 
vivo. 
In addition, it was recently found that when human immunodefi ciency virus 
type 2 (HIV - 2) DNA vaccine were formulated with cationic liposomes [362] , 
stronger immune responses in mice were observed compared with naked DNA 
alone. Using this knowledge, very recently a vaccine consisting of some HIV - 2 
genes ( tat, nef, gag , and env ) was formulated within cationic liposomes by Lochera 
et al. [363] . Baboons ( Papio cynocephalus hamadryas ) that were immunized by 
the intramuscular, intradermal, and intranasal routes with these expression constructs 
were challenged with HIV - 2 UC2 by the intravaginal route, and the results 
of this study demonstrate that partial protection against HIV - 2 vaginal challenge, 
as measured by reduced viral load, can be achieved using only a DNA vaccine 
formulation. 
5.3.4 APPLICATIONS OF LIPOSOMES IN THERAPEUTICS 
It is well known that liposomes have many applications in drug delivery. Initially, 
after the limitations of conventional liposomes were noticed, great effort was given 

toward the therapy of parasitic diseases, due to the fact that the targeting of RES 
macrophages, the place were parasites are mainly located, was considered to be very 
easy and fast (usually mentioned as passive targeting). Indeed, even now, research 
is ongoing, such as the treatment of drug - resistant visceral leishmaniasis with liposomal 
amphotericin - B [364] or with sterically stabilized liposomes containing camptothecin 
[371] . Nevertheless, possibly due to the high cost of liposome manufacturing, 
in relation to other drug formulations, and the fact that the main need for such 
medicaments would be for third world countries, such products have not been marketed 
for these diseases, despite the fact that they offer therapeutics advantages. In 
addition, again due to the same etiology, a very small part of recent research efforts 
and money are devoted to such diseases. 
A list of the marketed liposomal products is presented in Table 4 [364 – 366] . In 
addition, liposomes are currently being investigated for a variety of conventional 
and novel drugs: therapeutic agents, including antibiotics (as amikacin [367] , vancomycin, 
and ciprofl oxacin [368] ); anticancer agents (e.g., paclitaxel [369] and cisplatin 
[370] ; camptothecin and analogs [371 – 373] ), biologics such as antisense oligonucleotide 
[374] , DNA, and siRNA [375] ; and muramyl tripeptide [376] . A list of most 
liposome - based products currently under clinical investigation is presented in 
Table 5 . Additionally many products are currently being evaluated in preclinical 
studies. In many of the latest studies, the liposomes used have been surface modifi ed 
with active targeting ligands to improve delivery of therapeutics to target cells [238, 
377 – 379] . 
After investigating the recent literature, we have seen that most recent efforts 
connected with the use of liposome in therapeutics are mainly connected with the 
treatment of cancer. This is the reason why here we deal with the most common 
cancer types (brain, breast, lung, and ovarian cancer), and ongoing research and 
clinical treatments are discussed in more detail. However, we do not want to 
imply that the future of liposomes in drug delivery is limited to cancer therapy. 
Indeed, liposome structure, characteristics, and versatility are sure to fi nd, in 
TABLE 4 Currently Marketed Liposome - Based Products 
Active Agent a Application 
Daunorubicin (DaunoXome, Gilead Sciences, Inc.) Kaposi ’ s sarcoma 
Doxorubicin (Doxil/Caelyx, Ortho Biotech 
ProductsLP/Sequus Pharmaceuticals) 
Kaposi ’ s sarcoma 
Amphotericin B (Ambisome/Abelcet, Fujisawa 
Healthcare, Wyeth Pharmaceuticals) 
Fungal infections in 
immunocompromised patients 
Doxorubicin (Myocet/Evacet, Sopherion/ 
Liposome Company) 
Metastatic breast cancer 
Hepatitis A virus envelope proteins (Epaxal, 
Berna Biotech) 
Hepatitis A 
Infl uenza virus (Infl exal V, Berna Biotech) Infl uenza 
Verteporfi n (Visudyne, Novartis Ophthalmics) Age - related macular degeneration 
Source : D. Felnerova, J. F. Viret, R. Gluck, and C. Moser, Current Opinion in Biotechnology , 15, 518 – 529, 
2004. 
a Product names and companies given in parentheses. 
APPLICATIONS OF LIPOSOMES IN THERAPEUTICS 483

484 LIPOSOMES AND DRUG DELIVERY 
TABLE 5 Liposome - Based Products Currently under Clinical Testing 
Active Agent or Product a Application 
Company 
and Trial Phase/Reference 
Drug delivery 
Caelyx Bladder cancer Schering Plough; approved EU 
Doxil Multiple myeloma Schering Plough; III ALZA 
Pharm; III 
Bladder, liver cancer ALZA Pharm; II, III 
Pancreatic cancer ALZA Pharm; II, Sequuz; II 
Doxorubicin combined 
with ATB 
Prostate cancer Pharmacia; III, Neopharm; III 
Bladder cancer Neopharm; II, III Pharmacia; 
II, III 
Myocet combined with 
ATB 
Bladder cancer Liposome; III 
Liposomal ether lipid Bladder cancer Liposome; I 
(TLC ELL12) Lung, prostate, skin cancer Elan Pharm; I 
Platinum Cervical, ovarian, kidney Aronex Pharm; II 
(Aroplatin) Lung, pancreatic cancer Aronex Pharm; I, II 
Annamycin Leukemia 
Paclitaxel Head and neck cancer Pharmacia; II, III 
Lung cancer Neopharm; II, III 
Vincristine (Onco - TCS) Lung cancer Inex Pharm; II 
Topoisomerase inhibitor 
(OSI 211) 
Lung, ovarian cancer OSI Pharm; II 
All - trans retinoic acid 
(ATRA - IV) 
Lung cancer Antigenics; II 
Mitoxanthrone Other cancers Neopharm; II 
Nystatin (Nyotran) Leukemia (antifungal) Pharmacia; II 
Lung (antifungal) Aronex Pharm; II 
Prostate cancer (antifungal) Abbott Laboratory; II 
DNA delivery 
Human leucocyte 
antigen (HLA) B and 
. 2 microglobulin 
plasmid DNA 
(Allovectin) 
Head and neck cancer Vical; II 
Interleukin - 2 plasmid 
DNA 
Kidney, prostate cancer Vical; II 
Antisense toraf - 1 
(LerafON) 
Leukemia Neopharm; I 
Antigen delivery: 
MUC - 1 peptide: BLP25 
(human epithelial 
mucin peptide) 
Lung cancer Biomira; II 
Merck; III 
Source : D. Felnerova, J. F. Viret, R. Gluck, and C. Moser, Current Opinion in Biotechnology , 15, 518 – 529, 
2004. 
a Commercial names are given in parentheses. ATB, antibiotika. 

addition to those existing already, numberous applications in drug delivery in the 
future. 
Recently, a multicomponent liposomal drug delivery system consisting of doxorubicin 
and antisense oligonucleotides targeted to MRP1 mRNA and BCL2 mRNA 
to suppress pump resistance and non – pump resistance, respectively, has been developed 
[379] . This liposomal system successfully delivered the antisense oligonucleotides 
and doxorubicin to cell nuclei, inhibited MRP1 and BCL2 protein synthesis, 
and substantially potentiated the anticancer action of doxorubicin by stimulating 
the caspase - dependent pathway of apoptosis in multidrug resistant human lung 
cancer cells. 
5.3.4.1 Anticancer Drug Delivery 
Brain Tumors Brain tumors are classifi ed as gliomas (astrocytomas, oligodendrogliomas, 
ependymomas) and primitive neuroectodermal tumors (PNET) (medulloblastoma 
and supratentorial PNETs). Approximately half of all primary brain 
tumors are gliomas, while 80% of those are astrocytomas and glioblastomas. Most 
chemotherapeutic drugs are toxic to the healthy tissue and have damaging side 
effects due to their nonspecifi c nature. Incorporation of those drugs in liposomes 
can enhance the therapeutic effi cacy and reduce the toxicity. Drug delivery to the 
brain via the intravenous route has been a very challenging task due to the strict 
selectivity of the blood – brain barrier (BBB) as to the number and kind of molecules 
able to pass through. 
The BBB is the tight junction formed between the cerebral endothelial cells 
(Figure 11 ). These cells are in close contact with astrocytes and pericytes connected 
over a basal membrane. Only small (MW < 400 – 600) lipophilic molecules can diffuse 
FIGURE 11 Schematic of neurovascular unit/cell forming BBB (brain – blood barrier). 
( Reproduced from D. J. Begley, Pharmacology & Therapeutics , 104, 29 – 45, 2004, with 
permission by Elsevier .) 
Axonal 
ending 
Pericyte 
Endothelium 
Extraccllular matrix 
Astrocytic 
foot process 
Microglial 
cell 
Tight 
junction 
APPLICATIONS OF LIPOSOMES IN THERAPEUTICS 485

486 LIPOSOMES AND DRUG DELIVERY 
through the BBB, while the majority of the circulating drugs cannot access the brain. 
Nutrients and peptides pass through the BBB via receptor - mediated or carrier - 
mediated transport systems. These mechanisms are exploited in an attempt to deliver 
drug - loaded liposomes into the central nervous system (CNS) [380] . The most 
common are low - density lipoprotein (LDL) receptors, insulin receptors, and transferrin 
receptors. 
Thus, mAb against the transferrin receptor OX26 mAb has been conjugated via 
a stable thioether bond to the end of the PEG chain inserted on the liposome 
surface. Tritiated daunomycin was incorporated in OX26 mAb – PEG liposomes and 
the formulation was given intravenously to rats. The brain volume of distribution 
of daunomycin increased with time and exceeded 200 . L/g 24 h after injection [381] . 
In contrast, the pharmacokinetics of the free drug and the drug loaded in conventional 
liposomes was much lower [382] . 
In another study, the OX26 mAb has been grafted on the PEG chains by the 
biotin streptavidin coupling [383] . Brain tissue distribution obtained using biotinylated 
immunoliposomes was the same with that reported in the previous work where 
the mAb was chemically linked on the distal end of PEG. Therefore, the coupling 
method has not had a great impact on the brain accumulation of immunoliposomes. 
However, accumulation of the biotinylated PEG immunoliposomes was quite high 
in tissues such as liver, spleen, heart, muscle, and kidney. The latter was attributed 
to either the fact that the OX26 – biotinylated PEG immunoliposomes could pass 
through the BBB by an active transport system or the biotinidase activity, which 
could mediate cleavage of the targeting antibody from PEG and interfere with the 
tissue distribution of the formulation. 
An increase in therapeutic effi cacy and lower toxicity was reported with liposomes 
where the bradykinin analogue RMP - 7 was chemically attached at the end 
of PEG molecules of PEGylated liposomes (approximate size 70 nm) [384] . RMP - 7 
exhibits high selectivity for the B2 receptor of the BBB endothelial cells, which 
“ shrunk ” and let the RMP - 7 – PEG liposomes to pass into the brain. Actually the 
mechanism used in that study was based on opening the tight junctions of the BBB. 
Liposome - incorporated nerve growth factor (NGF) concentration increased 10 
times in comparison to free NGF, while they accumulated mainly in striatum, 
hippocampus, and cortex. 
A different type of immunoliposome was developed using antinuclear autoantibodies 
with nucleosome (NS) – restricted specifi city [187] . Anti - NS mAb 2C5 
specifi cally recognizes human brain tumor cells. Evaluation of immunoliposomes 
2C5 – PEG – PC/Chol was carried out in nude mice bearing subcutaneous brain tumor 
(U - 87 astrocytoma) and exhibited a threefold higher accumulation in the tumor in 
comparison to control (IgG – PEG liposomes). 
Moreover, disialoganglioside (GD 2 ) is expressed in abundance by neuroectodermal 
cancer cells and in low levels by normal cells located mainly in cerebellum and 
peripheral nerves. The Fab . fragment of the monoclonal antibody anti - GD 2 was 
grafted on PEG chains of sterically stabilized liposomes loaded with doxorubicin 
and their potential in treating neuroblastoma was assessed in nude mice with 
HTLA - 230 xenografts [160] . Mice receiving intravenously the immunoliposomes 
showed signifi cant improvement in long - term survival compared with other mice 
that received free DOX, freeGD 2 Fab . , Fab . – PEG liposomes, PEG liposomes – DOX. 
The control mice died from metastatic disease, while the immunoliposome - treated 

group lived 4 months longer. The same ligand, anti - GD 2 – Fab . , was used with liposomes 
carrying the antisense oligonucleotide c - myb [385] . After intravenous injection 
of the targeted liposomes into nude mice with HTLA - 230 xenografts, signifi cant 
prolonged survival times were obtained in comparison to controls (Figure 12 ). The 
suggested mechanism was downregulation of c - myb proto - oncogene expression. 
Although all the active targeting liposomes mentioned earlier have not left the 
laboratory, nonspecifi c sterically stabilized liposomes are being tested in clinical 
trials. Doxorubicin is the anticancer agent which is used as standard therapy, and it 
has the most serious side effects (mucositis, cardiotoxicity), so its incorporation in 
liposomes and bioavailability enhancement are under scrutiny [386 – 388] . 
It has also been demonstrated that PEGylated liposomal doxorubicin, Caelyx, 
can cross the BBB with a consequent accumulation in primary and secondary brain 
lesions [389] . In 10 patients with metastatic brain tumors treated with radiolabeled 
Caelyx concurrent with radiotherapy, the accumulation of the liposomal doxorubicin 
was 7 – 13 times higher in the metastatic lesions compared to the normal 
brain. 
Liposomal formulations, type of tumor, anticancer agent, delivery pathway, day 
of treatment, and general conclusions are given in Table 6 . DaunoXome (liposomal 
daunorubicin) and Doxil (liposomal doxorubicin) have been proved to have good 
response in clinical trials, in the range of approximately 40%. 
Also, according to Arnold et al. [390] , the dose scheme of doxorubicin - loaded 
liposomes affects the drug accumulation in various tissues as well as the tumor. The 
authors reported that after repetitive doses of sterically stabilized liposomes (SSL) - 
DOX every week, the plasma half - life of the drug increased, the deposition in liver 
and spleen decreased, and peak concentrations of DOX in the heart were threefold 
+ 
+ 
+ 
+ 
+ 
+ 
+ 
+
+ 
+ 
+ 
+ 
+ 
+ 
+ 
+ 
+ 
+ 
+ 
+ 
Control 
Free- CpG-myb-as 
CCL-CPG-myb-scr 
CCL-CpG-myb-as 
Targeted-CCL-CpG-myb-scr 
Targeted-CCL-CpG-myb-as 
100 
75 
50 
25
0 
0 40 80 120 160 
Time (days) 
Survival (%) 
FIGURE 12 Survival of NB - bearing nude mice after injection of oligonucleotides 
(free or encapsulated within liposomal formulations). Nude mice were injected intravenously 
with 3.5 . 10 6 HTLA - 230 neuroblastoma cells. After 4 h each mouse received 50 . g of 
oligonucleotides either free or encapsulated in targeted or nontargeted liposomes. 
Control mice received HEPES - buffered saline. ( Reprinted from ref. 385 with permission of 
Elsevier .) 
APPLICATIONS OF LIPOSOMES IN THERAPEUTICS 487

488 LIPOSOMES AND DRUG DELIVERY 
TABLE 6 Recently Completed and Ongoing Clinical Trials of Low - Molecular - Weight Drug - 
Carrying Liposomes for Brain Tumors 
Tumor 
Number 
of 
Patients Treatment Delivery 
Day of 
Treatment General Conclusions 
Recurrent 
tumor 
14 Daunorubicin IV Once every 4 
weeks 
6/14: Patients showed 
positive response 
GBM 8 Daunorubicin IV 24 h before 
surgery 
Concentration similar 
in tumor mass and 
peripheral regions 
Pediatric 
glioma 
7 Daunorubicin 
+ free 
carboplatin 
and 
etoposide 
IV Daunorubicin: 
day 1 and 2; 
carboplatin 
and etoposide: 
day 1 
5/7: Showed positive 
response with 
monthly treatment 
GBM 15 Doxorubicin + 
radiation 
IV Doxorubicin: 
days 1, 21; 
radiation: days 
1 – 21, 21 – 23 
4/10: Patients 
completely 
responded 
Solid tumor 22 Doxorubicin IV Once every 4 
weeks 
Phase I: dose - limiting 
toxicity: 70 mg/m 2 
Glioma 40 Daunorubicin 
+ free 
tamoxifen 
IV PEG - dox: day 4 
every 14 days 
Phase II: response 
(including 
stabilization) 40%; 
tamoxifen: day 1 
every 4 days 
Glioma 8 111 In labeled IV Contrast only Tumor uptake: 1.1% 
max tumor/brain 
contrast 7:5 
Glioma 3 Bleomycin IT Twice weekly 
for up to 6 
weeks 
All patients 
deteriorated, no 
toxicity 
Recurrent 
meningeal 
malignancies 
15 Cytarabine IT Once every 2 
weeks for 2 
courses; in 
positive 
response, 
second 
induction in 2 
weeks after 
fi rst dose 
Meningeal 
neoplasms 
100 Cytarabine 
against free 
methotrexate 
IT Once every 2 
weeks 
Ongoing 
Source : From G. H. Huynh, D. F. Deen, and F. C. Szoka, Jr., Journal of Controlled Release , 110, 236 – 259, 2006. 
Reprinted with permission of Elsevier. 
Note : GBM: glioblastoma; IV, intravenous; IT: intratumor; PEG: poly(ethylene glycol). See http://www. 
clinicaltrials.gov . 

lower. These results were not obtained using free DOX. In addition, a signifi cant 
increase in survival was achieved in animals treated weekly with SSL - DOX, while 
animals treated with free drug did not survive longer than the untreated controls. 
In phase I clinical trials children with recurrent or refractory tumors previously 
treated with free doxorubicin were administered i.v. Doxil in various doses (40 – 
70 mg/m 2 ) in order to determine the best tolerated one [391] . It was concluded that 
the maximum tolerated dose was 60 mg/m 2 because at the highest, 70 mg/m 2 , some 
patients developed mucositis, so dose adjustment was necessary. 
Administration of a combination of liposomal anthracyclines in parallel with 
other anticancer agents has been found of great advantage as they prolong 
the patients ’ survival. Caraglia et al. evaluated in a phase II study the use of 
combination Doxil with temozolomide in the treatment of brain metastases from 
brain tumors [392] . 
It is worth mentioning that temozolomide accumulates signifi cantly in the brain 
after oral administration. It is well tolerated and therefore is considered a potential 
candidate for combination chemotherapy. Administration of 200 mg/m 2 of temozolomide 
for 5 days and liposomal doxorubicin 35 mg/m 2 on day 1 was performed 
on 19 patients. The overall response rate was 36.8% and the median overall survival 
was 10.0 months. 
Moreover, Fiorillo et al. [393] studied the effect of a combination of liposomal 
daunorubicin, etoposide, and carboplatin administered to seven children with recurrent 
malignant supratentorial brain tumors as a second - line therapy. Chemotherapy 
consisted of infusion of liposomal daunorubicin on days 1 and 2 and infusion of 
etoposide and carboplatin on day 1 whereas courses were repeated every 3 – 4 weeks. 
After a total of eight courses, fi ve of seven children evaluated were alive 12 – 64 
months after diagnosis and 8 – 29 months from the start of the second - line chemotherapy. 
Of the seven children, three showed complete response, two partial 
responses, one stable disease, and one progressive disease. The time to the best 
response was 3 – 10 months, while the median time to progression was 23 months. 
The toxicity observed was minimum. 
Boron neutron capture therapy (BNCT) is also of high interest in treating brain 
tumors, especially glioblastoma multiforme, due to the high degree of normal brain 
infi ltration, the high histological complexity, and the heterogeneity of the cellular 
composition of the latter. This method is based on the nuclear reaction which 
occurs when boron - 10 is irradiated with low - energy thermal neutrons, producing 
high linear energy transfer of . particles and lithium - 7 nuclei [394] . Development 
of BNCT has been ongoing over the last 50 years and the greatest challenge is to 
achieve selective tumor targeting at a suffi cient therapeutic dose with minimal 
toxicity. Various compounds are currently being used or have been used in BNCT 
(shown in Figure 1 of [394] ), such as BPA [( l ) - 4 - dihydroxy - borylphenylalanine] 
and BSH (sodium mercaptoundecahydro - closo - dodecaborate), which are the fi rst 
most successfully used chemical compounds (so - called second - generation compounds) 
due to low toxicity, longer retention to the tumor site and tumor/brain, 
and tumor/blood ratios higher than 1. However, these drugs are not ideal, but they 
are safe after i.v. administration, so they are being used in clinical trials in Europe, 
the United States, Japan, and Argentina. The next group of advanced boron molecules 
(third generation) consists of stable boron or a cluster attached to a tumor - 
APPLICATIONS OF LIPOSOMES IN THERAPEUTICS 489

490 LIPOSOMES AND DRUG DELIVERY 
targeting moiety, such as monoclonal antibodies or low - MW biomolecules with 
amphiphilic properties. Other anionic compounds show little specifi city, so their 
potency increases when they are incorporated in either targeted or nontargeted 
liposomes [395, 396] . 
According to Feakes et al. [396] , boron - loaded DSPC/Chol liposomes of 40 nm 
average size were prepared and injected i.v. in murine mice carrying EMT6 tumors. 
Those liposomes showed high tumor retention, while boron amount was at therapeutic 
levels through the entire course of the experiment (more than 15 . g B/g 
tumor). However, targeted liposomes would, in theory, assure higher boron accumulation 
than the nontargeted ones [397] . 
Due to the overexpression by glioma cells, the most potent ligands for glioma 
treatment are endothelial growth factor receptor (EGFR) [398] , the vIII mutation 
of EGFR [399] , platelet - derived growth factor (PDGFR) [400] , and tenascin epitopes 
[401] . 
Liposomes are also used as carriers for gene delivery to gliomas while the cationic 
ones have demonstrated better interaction with cells in comparison to other types 
of liposomes [402, 403] . However, cationic liposomes suffer from toxicity, which 
varies according to cell type, duration of exposure, and density of cell culture. Antisense 
genes have been incorporated into liposomal carriers. For example, the EGFR 
antisense gene was packaged in PEGylated immunoliposomes [carrying human 
insulin receptor antibody (HIR)] [404] . It was reported [405] that the liposomes 
could cause 70 – 80% inhibition in human glioma cell growth. The same authors 
reported a 100% increase in survival time of mice with intracranial human brain 
cancer with weekly i.v. injections of antisense gene therapy directed at the human 
EGFR [405] . 
Moreover, double immunoliposomes were developed in order to treat intracranial 
human brain cancer in mice [406] . The mAbs used were the rat 8D3 mAb to 
the mouse transferrin receptor and the mAb against the HIR. RNAi (intereference 
RNA) is a new antisense gene therapy, where an expression plasmid encodes for a 
shRNA (short hairpin RNA). The shRNA is processed in the cell to an RNA duplex. 
The latter mediated RNAi. Indeed, weekly i.v. RNAi gene therapy reduced tumor 
expression of immunoreactive EGFR and caused an 88% increase in survival time 
of mice with advanced intracranial brain cancer. 
In another study, the herpes simplex virus thymidine kinase (HSVtk) gene was 
evaluated as to its potency to increase the sensitization of ganciclovir (GCV) to 
glioma cell lines when that gene was incorporated in liposomes [407] . 
Although the effi ciency of transfection was 18.6% in vivo after intratumoral 
injection of DNA liposomal complexes, the sensitivity to ganciclovir was improved 
as tumor weight induction was observed. In 2001, the FDA approved a clinical protocol 
relevant to liposomal gene therapy with the HSVtk/GCV system for the treatment 
of glioblastoma multiforme [408] . 
Hybrid vectors consisting of adeno - associated virus (AAV) vectors enclosed in 
liposomes lead to a10 - fold increase in transduction effi ciency compared to liposomes 
containing plasmid DNA and 6 - fold increase compared to AAV vector alone 
[409] . 
However, alternative routes are currently used in the clinic according to the 
site/location and type of tumor to achieve higher therapeutic effi ciency [410] because 
systemically administred drugs are not able to pass to cerebrospinal fl uid (CSF), 

whereas there is direct tumor exposure to the drug, characterized with increased 
drug concentration and half - life [410] . Thus, drugs can be administered in CNS via 
intrathecal, intraventricular, and intraparenchymal routes. Intrathecal and intraventricular 
routes result in a high drug concentration in the bulk CSF but with limited 
penetration to parenchyma. Thus, they are suited for treating meningeal and ventricular 
diseases. The intraparenchymal route assures the delivery in a local region 
in the parenchyma and so is useful for solid tumors and degenerative diseases that 
are surgically accessible. 
DepoCyt, a slow - release liposomal cytosine – arabinoside, undergoes clinical trials, 
as in some previous preclinical and clinical tests it was shown of good potency for 
the treatment of meningeal malignancies. For example, DepoCyt was administered 
to children with refractory neoplastic meningitis via lumbar puncture using an 
Ommaya reservoir or intraventricularly [411] . That study demonstrated the safety 
and feasibility of using liposomal cytarabine in children older than three years at 
the recommended dose of 35 mg with concomitant administration of dexamethasone. 
The latter is given to reduce the side effects of liposomal cytarabine, which is 
mild headache and back or neck pain. In a relevant study on adults with lymphomatous 
meningitis, DepoCyt given once every two weeks at a dose of 50 mg yielded 
a response rate of 71%, whereas free cytarabine at a dose of 50 mg twice a week 
produced a response rate of only 15% [412] . In patients with solid tumor neoplastic 
meningitis, 50 mg DepoCyt given once every two weeks yielded a response rate in 
evaluable patients of 36%, whereas free methotrexate 10 mg given twice a week 
produced a response rate of 21%. However, a problem accompanies the direct 
delivery to the brain parenchyma: the limited diffusion coeffi cient of particles in 
general (liposomes, nanoparticles, viral vectors) from the injection site in the brain 
tissue [410] . Thus, intraparenchymal delivery of liposomal carriers is facilitated using 
convection - enhanced delivery (CED) to distribute the drug through a larger region 
in the tissue. CED utilizes a bulk fl ow mechanism to deliver and distribute macromolecules 
to clinically signifi cant volumes of solid tissues providing a larger volume 
of distribution. A range of parameters affecting CED are connected with the physicochemical 
properties of liposomes, that is, size, surface charge, and steric stabilization 
[413] . 
According to investigations by MacKay et al. [413] , the ideal liposome (or in 
general nanoparticle) for CED will be PEGylated and of less than 100 nm in diameter, 
have neutral or negative surface charge, and need a targeting ligand to direct 
the particle to the target cell. For example, mannosylated liposomes containing clodronate 
infused directly to the fourth ventricle of the rat brain were selectively 
taken up by macrophages but not from microglia [414] . Also, the lipid dose is important 
as it was shown that infusing a high total lipid concentration reduced the fraction 
of the dose taken up by perivascular cells in the brain. 
However, MacKay and co - workers [413] carried out the previously mentioned 
study using healthy animals. Mamot et al. administered liposomes of either 40 or 
90 nm with 25% mannitol in U - 87 glioma xenograft animals [415] . In contrast to 
MacKay et al. [413] , the small 40 - nm liposomes infused via CED without mannitol 
achieved distribution over nearly all the tumor tissue. The addition of mannitol to 
CED further enhanced distribution, as liposomes accumulated throughout all sections 
of tumor and further penetrated beyond the tumor boundary into adjacent 
normal brain tissue. For the 90 - nm liposomes, CED with mannitol resulted in more 
APPLICATIONS OF LIPOSOMES IN THERAPEUTICS 491

492 LIPOSOMES AND DRUG DELIVERY 
than 50% tumor penetration of the total area evaluated , which also included extension 
into surrounding normal brain. Distribution was somewhat less extensive than 
with the 40 - nm liposomes, though. 
The effi cacy of liposome administration with CED was demonstrated using real - 
time magnetic resonance imaging (MRI) of rat brain tumors [416] . Two types of 
liposomes were used: doxorubicin - loaded liposomes and gadolinium (Gd) – loaded 
(MRI contrast agent) liposomes. According to the authors, MRI facilitated the distinction 
of distribution between different tumor models, such as C6 gliomas and 
9L - 2 gliomas. Also, after CED of Doxil, the drug presence was identifi ed in the tissue 
several weeks after a single administration, while the therapeutic response achieved 
was greater compared to systemic administration of Doxil. The fact that therapeutic 
liposomes can be coinfused with liposomal Gd to successfully monitor the distribution 
of the drug carrier in the nonhuman primate brain was shown by Saito and 
co - workers [417] . A 2 : 1 ratio between the volume of distribution and the volume 
of liposomes infused was found while liposomal Gd was still detectable 48 h postinfusion, 
confi rming the previous fi nding of prolonged retention of liposomal Gd in 
the tissues and negligible toxicity in rat. With this study, the authors showed that 
CED is a technique enabling safe and extensive liposome distribution. Also, real - 
time MRI is a potentially useful tool to estimate the concentration and tissue half - 
lives of liposome - loaded drugs within target tissues. 
However, CED might not always be successful as good technical skills are 
required (if the catheter is not placed properly, the liposomes will escape within the 
CSF). Also CED is an invasive technique and could cause infl ammation and neurotoxicity 
and it is determined by many formulation characteristics and diffusional 
properties of the latter in brain tissue [418] . 
Breast Cancer Breast cancer is the second leading cause of cancer deaths in 
women today (after lung cancer) and is the most common cancer among women, 
excluding nonmelanoma skin cancers [419] . According to the World Health Organization, 
more than 1.2 million people will be diagnosed with breast cancer this year 
worldwide. Breast cancer stages range from stage 0 (very early form of cancer) to 
stage IV (advanced, metastatic breast cancer). Each patient ’ s individual tumor 
characteristics, state of health, genetic background, and so on, will impact her survival. 
In addition, levels of stress, immune function, will to live, and other unmeasurable 
factors also play a signifi cant role in a patient ’ s survival. The majority of women 
with breast cancer will undergo surgery as part of their cancer treatment (lumpectomy 
and mastectomy). In addition to surgery, some women will receive adjuvant 
(additional) treatment (chemotherapy, radiation therapy, and drug treatments) to 
stop cancer growth, spread, or recurrence. Occasionally women may be treated with 
chemotherapy, radiation, or drugs without having breast surgery. 
From a clinical point of view and with relevance to liposomes, administration of 
either PEGylated (Doxil) or non - PEGylated liposomal doxorubicin (Myocet) has 
improved the safety and tolerance of patients with breast cancer by signifi cantly 
reducing the main side effect of those drugs, cardiotoxicity. However, drug effi cacy 
remains the same either incorporated or not in the liposomes as this is reported 
after conducting various clinical studies [420, 421] . For example, Myocet (M) received 
European approval for use in patients with metastatic breast cancer at a dose of 

60 mg/m 2 in combination with cyclophosphamide (C) having shown equivalent effi - 
cacy at a phase III study [422] . Also, doxorubicin is given at a dose of 60 or 75 mg/m 2 
[423] . Chan et al. [424] studied the effi cacy and tolerability of Myocet in a dose of 
75 mg/m 2 in combination with cyclophosphamide in 160 patients. A high incidence 
of neutropenia was obtained. Thus, that group confi rmed that the use of 60 mg/m 2 
is an appropriate choice. However, administering a combination of either Myocet 
and cyclophosphamide or epirubicin and cyclophosphamide to patients with metastatic 
breast cancer, the response rate was not signifi cantly different, which showed 
that the use of the liposomal formulation just reduced the cardiotoxicity of 
doxorubicin. 
In a phase I study, Myocet was administered in combination with docetaxel in 21 
metastatic breast cancer patients with the aim of estimating the safety and maximum 
tolerated dose of Myocet in parallel with docetaxel [425] . The latter is proved highly 
active in metastatic breast cancer as well as doxorubicin. The maximum tolerated 
dose was 50 mg/m 2 of Myocet and 25 mg/m 2 of docetaxel. As reported in the previous 
study, neutropenia was the most common toxicity effect while some incidents of 
congestive heart failure were observed after a total doxorubicin dose of 540 mg/m 2 . 
Moreover, a combination of Myocet (75 mg/m 2 ) with gemcitabine (350 mg/m 2 ) and 
docetaxel (75 mg/m 2 ) was injected intravenously in 44 patients with early breast 
cancer every three weeks for six cycles [426] . The overall clinical response rate was 
80% and the pathological complete response was 17.5%. The toxicity of the regimen 
was moderate and, as expected, neutropenia and leukopenia were the most prominent 
side effects. The latter effects were well managed and treatment discontinuation 
was not required. 
Using PEGylated liposomal doxorubicin (Caelyx), Keller et al. [427] compared 
the effi cacy of the liposomal formulation with that of a common regimen in patients 
with taxane - refractory advanced breast cancer. The regimen scheme was Caelyx 
(50 mg/m 2 every 28 weeks) or vinorelbine (30 mg/m 2 ) or mitomycin C (10 mg/m 2 
every 28 days) plus vinblastine (5 mg/m 2 at days 1, 14, 28, 42). Finally, progression 
free survival and overall survival were similar for Caelyx and the comparative 
regimen. 
The effi cacy and toxicity of Caelyx in combination with paclitaxel (Taxol) were 
investigated as a fi rst - line therapy in 34 patients with advanced breast cancer in a 
multicentric phase II study [428] . Paclitaxel at a dose of 175 mg/m 2 and Caelyx 
(30 mg/m 2 ) were administered intravenously every 3 weeks. It was shown that the 
response rates of the combination were over 70% while the median time to treatment 
failure was 45 weeks. No signifi cant clinical cardiotoxicity was observed and 
the usual side effects (mucositis, stomatitis, hand – foot syndrome) were treated 
accordingly. 
In another phase II study, Caelyx was used in combination with cyclophosphamide 
(CP) as a fi rst - line therapy in patients with metastatic or recurrent breast 
cancer [429] . Three different schemes were given in groups of patients: Caelyx 
50 mg/m 2 intavenously on day 1 and CP 100 mg/m 2 (orally on days 1 – 14) every 
28 days, 30 mg/m 2 Caelyx and 600 mg/m 2 i.v. on day 1 every 21 days, and 35 mg/m 2 
Caelyx plus 600 mg/m 2 CP i.v on day 1 every 21 days. The responses were similar 
among the different groups (51%) of patients, but less side effects were observed 
in group 2, making that regimen the one of choice as a fi rst - line therapy for patients 
APPLICATIONS OF LIPOSOMES IN THERAPEUTICS 493

494 LIPOSOMES AND DRUG DELIVERY 
with metastatic or recurrent breast cancer. The median duration of response was 
35.1 weeks. 
Coleman et al. [430] demonstrated that using Caelyx in a dose format of 
50 mg/m 2 every four weeks in patients with metastatic breast cancer is quite effective 
(objective partial responses 31%) while the main side effect was hand – foot 
syndrome. 
From a research point of view, liposomes have been used in order to facilitate 
or alter the pharmacokinetics/bioavailability of various molecules. For example, 
ceramide is an antiproliferative and proapoptotic molecule produced after sphingomyelin 
metabolism [431] . Ceramide C6 – loaded PEG(750) - C8 liposomes were 
injected intravenously in mice bearing syngeneic or human xenografts of breast 
adenocarcinoma. Administration of 36 mg/kg of liposomal C6 over a three - week 
period caused a sixfold decrease of tumor size in the case of syngeneic tumor – 
bearing mice. Liposomal C6 accumulated in caveolae and mitochondria, while a 
marked increase of apoptotic cells was observed. The PEGylated liposomal ceramide 
followed fi rst - order kinetics in the blood and a steady - state concentration was 
achieved in tumor tissue (Figure 13 ). Also, a decrease in tumor size was obtained 
in the human xenograft model. Minimal toxicities were observed in tumor - bearing 
mice, suggesting that the bioactive concentration of C6 achieved in the tumor tissue 
is not active in normal tissues. Besides, contortrostatin (CN), a molecule isolated 
from snake venom, has been proved to interact with integrins on tumor cells and 
with newly growing vascular endothelial cells via its two Arg – Gly – Asp sites [432] . 
Thus, interactions between CN and integrins disrupt several steps critical to tumor 
growth: angiogenesis and metastasis. CN was incorporated in PEGylated liposomes 
with an encapsulation effi ciency of approximately 80% while retaining full biological 
activity. Intravenous injection of approximately 100 . g of liposomal CN twice a 
week caused a signifi cant reduction of 94% of microvascular density and hindered 
the tumor growth in the MDA - MB - 435 xenograft model. Also, liposomal CN demonstrated 
prolonged circulation ( t 1/2 values were 19 and 0.5 h for liposomal and 
nonencapsulated CN, respectively). 
In another study, the authors modifi ed the liposome composition by including 
polyoxy - ethylenedodecyl ether [C 12 (EO) n ] in the DMPC - made liposomes [433] . 
These vesicles caused activation of caspases 3, 8, 9 and eventually induction of 
apoptosis in MDA - MB - 453 cells. Another idea is based on using methods to 
direct the drug loading carriers to the cancer site. More specifi cally, the paclitaxel 
is successfully used for the treatment of breast cancer among other types of 
cancers but has side effects such as neutropenia, peripheral neuropathy, and 
hypersensitivity reactions. Also, it is formulated with Cremophor EL due to its 
high lipophilicity. In order to decrease the drug toxicity and enhance the therapeutic 
potential of that drug, paclitaxel was incorporated in negatively charged 
magnetic liposomes and its effi cacy was evaluated in vivo [191] . A magnetic fi eld 
of suitable strength was used to direct the magnetoliposomes to the desired site. 
Indeed, after intravenous injection the AUC obtained for the magnetic carriers 
and the Cremophor EL/ethanol were 20.7 and 6.8 h · . g/mL, respectively. The liposomal 
paclitaxel concentration was much higher in the tumor in comparison to 
other organs while the concentration peak was reached sooner (19.85 . g/g at 
0.25 h). Even after 8 h, paclitaxel concentration in the plasma was 2.71 – 0.33 . g/mL 
for the Cremophor EL formulation. The antitumor effi cacy of magnetoliposomes 

FIGURE 13 Blood and tumor concentrations of bioactive ceramide – lipid C6 in tumor - 
bearing mice were maintained over a 48 - h period: ( a ) 10 - and 40 - mg/kg doses of liposomal - C6 
followed fi rst - order kinetics with blood concentration exceeding in vitro IC 50 sustaibed at 
48 h; ( b ) at these doses steady - state concentration of C6 in tumor tissue achieved at . 60 min. 
The 40 - mg/kg dose maintained a concentration above the desired IC 50 up to 48 h. ( Reprinted 
from ref. 431 with permission of American Association for Cancer Research .) 
40 mg/kg 
10 mg/kg 
IC50 (–5.M) 
40 mg/kg 
10 mg/kg 
IC50 (–5.M) 
(a) 
(b) 
40 
30 
20 
10
0
0
5 
10 
15 
20
0.0 0.5 1.0 1.5 2 12 22 32 42 52 
0.0 0.5 1.0 1.5 2 12 22 32 42 52 
Hours 
Hours 
.g C6 / MI Blood Ng C6 / mg Tumor Tissue 
(expressed as change rate of tumor weight) after subcutaneous injection of 10 mg/ 
kg drug was equivalent to that achieved with Cremophor EL 50.4 and 51.9%. 
When the administered dose was 20 mg/kg, the change rate of tumor weight was 
the highest, 60.5%. 
APPLICATIONS OF LIPOSOMES IN THERAPEUTICS 495

496 LIPOSOMES AND DRUG DELIVERY 
Pulsed high - intensity focused ultrasound (HIFU) is another suggested method 
to enhance the delivery and therapeutic effect of Doxil in a murine breast cancer 
tumor model with the fi nal aim of reducing the Doxil dose given during therapy 
[434] . However, the results were not particularly encouraging as there was not any 
signifi cant difference between free and liposomal Doxil distribution in the tumor 
after HIFU exposure. The latter was possibly attributed to the ability of liposomes 
to extravasate easily through the leaky vasculature of the tumor, so the use of HIFU 
did not add any therapeutic or other advantage. 
In an attempt to achieve active targeting, sigma receptor ligands were incorporated 
on to the liposomal surface. Sigma receptors are overexpressed in various 
human tumors, including breast cancer cells [435] . Haloperidol has shown high affi nity 
for sigma receptors; thus it has been attached at the end of PEG molecules, which 
are protruding from the surface of cationic lipoplexes. Indeed, haloperidol – PEG 
lipoplexes showed more than 10 - fold higher gene expression in MCF - 7 (breast 
carcinoma) cells than control lipoplexes, while the presence of haloperidol or 1,3 - 
ditolylguanidine suppressed the expression of the reporter gene [435] . 
Transferrin is another ligand attached on the lipoplex surface so to cause higher 
transfection effi ciency, as has been reported [436] . Thus, Basma et al. [437] studied 
the effect of cis - diaminedichloro platinum (CDDP) in combination with bcl - 2 antisense 
treatment on p53(+)MCF - 7 and p53( . )MCF - 7/E6 breast cancer cells using 
transferrin bcl - 2 lipoplexes. The median inhibitory concentration (IC 50 ) for bcl - 2 
antisense delivered with lipoplexes plus transferrin was approximately 1.4 . M for 
MCF - 7 and 1.2 . M for MCF - 7/E6 cells (Figure 2 of ref. 437 ). In CDDP - treated cells, 
the IC 50 was approximately 5 . M for both cell lines. In general, bcl - 2 antisense 
delivered in the form of transferrin bcl - 2 lipoplexes in combination with cisplatin 
induced cell death and apoptosis in a higher degree in MCF - 7/E6 cells rather than 
MCF - 7 cells. Cisplatin demonstrated higher caspase - 8 activation compared to the 
targeted lipoplexes, which suggested that possibly caspase - 8 is the major pathway 
for cancer cell death. G3139 is another oligonucleotide (ODN) capable of downregulating 
bcl - 2 and, its effi cient delivery to breast cancer cells would potentially 
make it a successful candidate for antisense therapy [438] . Thus, the effi ciency of 
different liposomal formulations for the lipid composition (DOTAP, DC - Chol, 
CCS) 1 with or without helper lipids DOPC, DOPE, and Chol and liposomal size 
[approximately 100 nm LUV or unsized heterolamellar vesicles (UHV)] on MCF - 7 
breast cancer cells was examined [438] . Out of 18 tested formulations, only the CCS - 
bcl - 2 lipoplexes (UHV derived) have been effective, causing a larger than 50% 
decrease of cell growth in comparison to free ODN. The possible mechanism of 
action of the particular formulation is attributed to the presence of one primary and 
two secondary amines on the spermine moiety of the CCS molecule. Because of 
this, CCS lipoplexes are taken up by cells via adsorptive endocytosis and the secondary 
amines cause a “ proton sponge effect ” due to which ODN escapes from the 
endosomes and so has the ability to interact with the bcl - 2 neutralizing it. Of course, 
the effect of the L + /DNA . ratio is a determining factor along with the lipid composition 
and size. 
1 DOTAP: N - (1 - (2,3 - dioleoyloxy)propyl) - N , N , N - trimethylammonium chloride; DC - Chol: 3 . [ N - ( N . N . - di 
methylaminoethane)carbamoyl] - cholesterol; CCS: ceramide carbomoyl spermine. 

Diagnosis is always the fi rst aim in effectively treating breast cancer, so the need 
to develop or reveal more tumor markers at an early stage of the cancer is absolutely 
critical [439] . Such are the circulating epithelial cells, the cyclins, and the urokinase - 
type plasminogen activator and plasminogen activator inhibitor, which indicate 
breast cancer or metastatic spread apart from the already existing markers estrogen 
receptor, progesterone receptor, and human epidermal growth factor receptor - 2 
(HER - 2). Liposomal formulations have facilitated anticancer activity of highly toxic 
anticancer drugs as well as altered their biodistribution, while targeted drug - loaded 
liposomes have shown some promising results in the laboratory. Development of 
new drugs and use of particulate carriers to increase bioavailability could be one 
way forward in the battle for improvement of quality of life for patients suffering 
from breast cancer. 
Lung Cancer Lung cancer might be the most common form of cancer and the 
most common cause of death in both men and women, although it affects more men 
than women [440] . There are three main types of lung cancer, based on their appearance 
when examined by a pathologist: small cell carcinoma, squamous cell carcinoma, 
and adenocarcinoma. The latter two consist of non – small cell lung cancer. It 
is important to know which type of cancer a patient has because small cell cancers 
respond best to chemotherapy (anticancer medicines) whereas the other types 
(often referred to collectively as non – small cell cancer) are better treated with 
surgery or radiotherapy (X - ray treatment). 
Surgery can cure lung cancer, but only one in fi ve patients are suitable for this 
treatment. If the tumor has not spread outside the chest and does not involve vital 
structures such as the liver, then surgical removal may be possible, but only if the 
patient does not also have severe bronchitis, heart disease, or other illnesses. Small 
cell lung cancer is treated with chemotherapy. Non – small cell cancer may be treated 
with radiotherapy and chemotherapy (as part of a research trial) or with supportive 
care. Radiotherapy is either “ radical ” or “ palliative. ” 
Regarding drug treatment of lung cancer, Merck KGaA and Biomira will soon 
start phase III trial on their BLP - 25 liposomal vaccine for patients with non – small 
cell lung carcinoma [441] . L - BLP25 is a synthetic MUC1 peptide vaccine. MUC1 is 
a mucinous protein expressed on the apical borders of normal epithelial cells. 
It is overexpressed and glycosylated on tumor cells, where it appears to be antigenically 
distinct from the normal protein. The liposomal vaccine can induce a MUC1 - 
specifi c T - cell response. Median survival for patients with stage IIIB locoregional 
non – small cell lung cancer who received L - BLP25 in a phase IIb study was 30.6 
months in the vaccinated group compared with 13.3 months for the unvaccinated 
group. 
Caelyx is liposomal doxorubicin very well used as a treatment of choice for a 
number of cancers with good tolerability and antitumor activity, as has been demonstrated 
in many phase I or II clinical trials. One such example is the conduct of 
phase I study of Caelyx (PEGylated liposomal doxorubicin, 25 – 40 mg/m 2 ) in combination 
with cyclophosphamide (750 – 1000 mg/m 2 ) and vincristine (1.2 mg/m 2 ) every 
21 days in patients with relapsed or refractory small cell lung cancer [442] . The suggested 
doses were CaelyxTM 35 mg/m 2 , cyclophosphamide 750 mg/m 2 , and vincristine 
1.2 mg/m 2 intravenously every 21 days. This combination was well tolerated 
APPLICATIONS OF LIPOSOMES IN THERAPEUTICS 497

498 LIPOSOMES AND DRUG DELIVERY 
while antitumor activity was observed for patients with relapsed small cell lung 
carcinoma with survival duration of 5 months. The latter is similar to what is achieved 
using only camptothecin analogues taxanes or vinorelbine (survival duration . 6 
months). 
Also, paclitaxel is the most widely used anticancer agent for non – small cell 
lung cancer [420] . Liposomal encapsulated paclitaxel faces the problem of formulation 
due to the drug ’ s high hydrophobicity. However, a phase I trial with liposomal 
paclitaxel reported dose - limiting toxicity at the dose of 150 mg/m 2 /week. Besides, 
the whole blood clearance of paclitaxel was similar for liposomal and free 
paclitaxel. 
Promising results that were referred to liposomal encapsulated paclitaxel easy to 
use (LEP - ETU; NeoPharm) consisted of DSPC/Chol/cardiolipin molar ratio 90 : 5 : 5, 
lipid/drug molar ratio 33 : 1, and paclitaxel concentration 2 mg/mL (mean liposome 
size 150 nm). In a phase I study of increasing doses of liposomal paclitaxel (135 – 
375 mg/m 2 ) in 25 patients, the enhanced effectiveness was proved with much better 
tolerability and with 3 partial remissions and 11 patients with stable disease. Lurtotecan, 
a camptothecin analogue, was incorporated in PC/Chol (2 : 1 molar ratio) 
liposomes (size 50 – 100 nm) with a lipid/drug molar ratio 20 : 1. A remarkable 1500 - 
fold AUC increase was obtained after administration in nude mice and several 
xenograft models. 
9 - Nitro - 20( S ) - camptothecin (9NC), another lipophilic camptothecin analogue, 
showed antitumor effects in mice and milder effect in humans after oral administration. 
Thus, the potency of 9NC - loaded di-lauryl-PC (DLPC) liposomes was investigated 
in patients with primary or metastatic lung cancer after aerosol administration 
(aerosol droplet size 1 – 3 . m) for fi ve consecutive days per week [373] . Indeed, the 
most serious side effect of 9NC, hematological toxicity, did not appear in any of the 
patients, while the dose - limiting toxicity, chemical pharyngitis, was observed at a 
dose of 26.6 . g/kg/day. 9NC plasma levels, after aerosol administration (13.3 . g/kg/ 
day), were similar to that given orally (2 mg/m 2 ), despite the lower dose administered 
in the fi rst case. More specifi cally, C max and AUC were 76.7 ± 39.1 ng/mL and 275 ± 
149 ng - h/mL, respectively, after aerosol administration and 111 ng/mL and 194.4 ± 
108.4 ng - h/mL, respectively, after oral administration. The recommended dose for 
phase II studies is 13.3 . g/kg/day on a 1 - h exposure for fi ve consecutive days per 
week and for eight weeks, with 0.4 mg/mL 9NC concentration in the nebulizer. 
Moreover, targeting moieties have been grafted on the vesicle surface, exploiting 
the fact that antigen/receptor overexpression on cancer cell membranes increases 
the specifi city of the active substance. For example, antagonist - G is a hexapeptide 
which blocks the action of multiple mitogenic neuropeptides by interacting with 
their receptors [170] . Antagonist - G was chemically attached at the distal end of 
PEG molecules of stealth liposomes (HSPC/Chol/mPEG – DSPE/PDP – PEG – DSPE, 
2 : 1 : 0.08 : 0.02 molar ratio) (SLG) loaded with doxorubicin. The antiproliferative 
activity of doxorubicin was estimated on the human variant small cell lung carcinoma 
(SCLC) H82 cell line. Indeed, 20 - to 30 - fold increase of both, binding, and 
internalization took place after SLG incubation on cells in comparison to stealth 
liposomes only, or PEGylated liposomes without the antagonist - G. The 0.03 . g of 
antagonist - G on the liposome surface was enough to cause 50% cell liposome association. 
Doxorubicin accumulation in the whole cell was 20 - fold higher with SLG. 
Eventually, there was the suggestion that the main antagonist - G mechanism of 

action is to bind to vasopressin receptor, which is expressed abundantly on the 
SCLC cells. 
Hyaluronan (HA) is another potential candidate for tumor targeting because it 
has been proven that hyaluronan receptors CD44 and RHAMM are overexpressed 
on several tumor types [443] . In this case, hyaluronan is used not only as a targeting 
moiety but also to prolong the circulation half - life of the vesicles in question; in 
other words, it replaces PEG molecules. HA was chemically attached on the vehicle 
surface (57 . g/ . mol lipid), which was comprised of PC/PE/Chol (3 : 1 : 1) (HA – LIP). 
Doxorubicin was entrapped in 78 ± 5% encapsulation effi ciency, while the . potential 
and size of the targeted liposomes were . 13.1 ± 3.9 mV and 81 ± 13. Pharmacokinetic/
biodistribution studies were carried out with Doxil (PEGylated liposomal 
doxorubicin) as a comparison to the new hyaluronan liposomes. Those studies plus 
therapeutic responses were tested on three mice models, one of which was C57Bl/6 - 
bearing B16F10.9 lung metastasis. Clearly, ha-liposomal (HA–LIP)–DXR exhibited 
prolonged blood circulation similar to Doxil (approved formulation in the market), 
which indicated that the amount of attached HA on the vehicle surface is enough 
to stabilize and offer the steric stabilization required (Figure 14 ). Also, accumulation 
of HA – LIP – DXR was much more reduced in liver of C57Bl/6 - bearing B16F10.9 
lung metastasis mice, while DXR accumulation in the tumor site was threefold 
higher to that achieved using Doxil. Signifi cant improvement was obtained in both 
metastatic burden and survival with Doxil and HA – LIP – DXR (Figure 15 ). Besides, 
the HA – LIP – DXR had positive results on all tumor types tested. 
Manipulating the genetic material by injecting tumor - suppressing genes has been 
an alternative way in cancer treatment. The 3p FUS1 gene is a tumor suppressor 
FIGURE 14 Doxorubicin (DXR) plasma concentration ( . g/mL) as function of time from 
dosing: ( a ) C57BL/6 mice inoculated (by intraveneous injection) with B16F10.9 cells; 
( b ) healthy C57BL/6 mice; ( c ) BALB/c mice inoculated with C - 26 cells (injected into right - 
hind footpad). A single dose of the selected formulation was injected into the tail vein. DXR 
formulations and doses are specifi ed. ( Reprinted from ref. 443 with permission of Neoplasia 
Press, Inc .) 
Free DXR (10 mg/kg) 
nt-LIP-DXR (10 mg/kg) 
Doxil (10 mg/kg) 
tHA-LIP-DXR (10 mg/kg) 
C57BL/6 
B16F10.9 
Time (h) 
DXR plasma concentration (.g/mL) 
100 
10
1 
0.1 
0.01 
0.001
0 20 40 60 80 100 
(a) 
APPLICATIONS OF LIPOSOMES IN THERAPEUTICS 499

500 LIPOSOMES AND DRUG DELIVERY 
gene which belongs to a 120 - kb region of the 3p chromosome on the region 3p21.3. 
That 120 - kb region is missing so injection of DOTAP/Chol lipoplexes with the FUS1 
gene could induce apoptosis and inhibit tumor growth [444] . A signifi cant inhibition 
of lung metastatic tumor was obtained in animals (bearing subcutaneous lung tumor 
xenografts) treated with liposome – FUS1 DNA complex (total of six doses). Also, 
intravenous treatment of lung tumor - bearing animals with DOTAP/Chol – FUS1 
complex prolonged the animal survival; 40% of the animals were still alive after 
approximately 100 days. Also, the antitumor potency of FUS1 was demonstrated to 
be superior to the one obtained with p53 as the same therapeutic effect was achieved 
by using less amount of FUS1 gene in the lipoplexes in comparison to p53. This 
would be an advantage because lowering the DNA dose would result in much lower 
toxicity of DNA due to infl ammatory response. 
FIGURE 15 Therapeutic responses of mice bearing B16F10.9 - originating lung metastatic 
disease. Doxobubicin (DXR) formulations and doses are specifi ed. Treatments were on days 
1, 5, and 9 by injection of selected formulation to the tail vein. ( a ) Lung metastatic burden. 
Light - shaded bars are data for increase in lung weight; dark - shaded bars are data for number 
of lung metastasis: ( * * * ) P < 0.001 compared with free drug. ( b ) Survival ( n = 5). Each line 
connects the symbols representing the daily survival state of the group. ( Reprinted from 
ref. 443 with permission of Neoplasia Press, Inc .) 
400 
300 
200 
100
0 
100 
80 
60 
40 
20
0 
Increase in lung weight (%)
Number of lung metastases 
160 
120 
80 
40 
0 
Saline Free DXR nt-LIP-DXR Doxil tHA-LIP-DXR 
Saline 
Free DXR(10 
mg/kg) 
nt-LIP-DXR 
(10 mg/kg) 
Doxil (10 
mg/kg) 
tHA-LIP-DXR 
(10 mg/kg) 
Survival (%) 
0 20 40 60 80 100 
Time from tumor inoculation (days) 
(a) 
(b)

Moreover, the effi ciency of gene therapy depends very much on the vector used 
to achieve the gene delivery, the properties and stability of the fi nal lipoplexes in 
the presence or absence of serum, and the pharmacokinetics and biodistribution. 
On this basis, Li et al. studied the effect of lipoplex size on the lipofection effi ciency 
of TFL - 3/pDNA (plasmid - encoding luciferase) liposomes in the absence and presence 
of serum and investigated the correlation between in vitro and in vivo results 
using either B16BL6 murine melanoma cell line or the same cells injected i.v. in 
C57BL/6 mice in order to produce pulmonary metastases [445] . The authors incubated 
a range of different ratios of pDNA/TFL - 3 (P/L) ranging from 8 to 120 P/L 
on the previously mentioned cell line in the absence and presence of serum. Serum 
had a dramatic effect on lipoplex size; in the absence of serum, the size increased 
as the pDNA amount increased and it reached a maximum value at the ratio 80 g/ 
mol P/L, while the . potential decreased as the P/L ratio increased and at 80 g/mol 
P/L was approximately +2 mV. Maximum luciferase activity was observed with 
lipoplexes of 80 g/mol. In contrast, in the presence of serum, the size increased from 
8 to 20 g/mol and decreased from 40 to 120 g/mol as the P/L increased. Maximum 
luciferase activity was obtained with 40 g/mol in the presence of serum. However, 
the in vitro transfection effi ciency of the lipoplexes in the presence of serum at the 
highest point of 40 g/mol was twofold less than the one achieved at the peak of 
transfection effi ciency at 80 g/mol in the absence of serum. However, the plasmid 
expression was higher with 8 and 80 g/mol lipoplexes in tumor - bearing animals in 
contrast to the in vitro fi ndings in the presence of serum. This was attributed to less 
aggregate formation in the blood due to the lipoplex/serum ratio (50 – 100 . L serum/ 
200 . L lipoplex dispersion) so there is no complete interaction between lipoplexes 
and serum proteins. Also, pulmonary gene expression was dependent on the time 
after cell inoculation. It was shown that gene expression takes place at different 
parts of the lung; for example, 8 g/mol P/L lipoplexes expressed luciferase in the 
cells surrounding the tumor, while 80 g/mol lipoplexes expressed the gene in 
the entire lung without any specifi city. Possible explanations for the latter are the 
extended lung capillary bed and increased lipoplex uptake by tumor cells compared 
to the normal cells. Of course many other factors could contribute to the different 
patterns of gene expression, such as endosomal release, nuclear uptake, increased 
translation, and transcription. 
All - trans retinoic acid (ATRA) has shown anticancer activity in a number of 
types of cancer cells [446] . However, some non – small cell lung carcinoma is resistant 
to ATRA probably due to the high lipophilicity of the molecule, which makes it 
unable to pass through the cellular membrane. Therefore, cationic liposomes 
(DOTAP/Chol 1 : 1 molar ratio) were used to incorporate and facilitate ATRA ’ s 
action in the resistant NSCLC in A549 human lung cancer cells. The produced 
ATRA – DOTAP/Chol lipoplexes were 125 nm in diameter with 50 mV of . potential, 
while for the DSPC/Chol liposomes used as control these were 110 nm and . 3 mV, 
respectively. The former lipoplexes exhibited higher uptake by the cells in comparison 
to DSPC/Chol due probably to the positive surface charge, which interacts to a 
higher degree with the negatively charged cell membrane. Thus, the apoptosis 
induced on these cells in the presence of ATRA lipoplexes was much higher than 
the one caused by ATRA only or ATRA – DSPC/Chol (Figure 16 ). 
Lastly, although antisense therapy is considered a simple and effi cient procedure, 
its products have never reached the market. This is due to the instability of phos- 
APPLICATIONS OF LIPOSOMES IN THERAPEUTICS 501

502 LIPOSOMES AND DRUG DELIVERY 
phodiester oligonucleotides (PO - ONs) in the cytoplasm, while the phosphothioate 
ONs demonstrate unwanted side effects [447] . In order to design better delivery 
systems for PO - ONs, the intracellular fate of the PO - ON and PS - ON lipoplexes 
with DOTAP/DOPE was investigated after their application on A549 cells. After 
endocytosis, endosomal localization, and endosomal escape of the lipoplexes, the 
ONs were localized in the nuclei. However, PO - ONs were degraded in the nucleus 
(degradation products diffuse out of the cells after 2 h) whereas PS - ONs were still 
intact and remained in the nucleus for 8 h. It is worth mentioning that PS - ONs were 
eliminated from the nucleus and were found in cytoplasmic granules, which indicated 
that the cell has a mechanism of elimination of intact PS - ONs. Also, it seemed 
that the amount of PO - ONs was quite important for the ONs ’ stability as the 
degradation was reduced after injecting 22 . M , in contrast to complete degradation 
with injection of 2 . M . From the study it was concluded that ON degradation 
happened after their release from the lipoplexes. Thus, for the successful delivery 
of ONs, they have to remain complexed with their carrier. Polyethylenimine (PEI) 
was suggested as a possible carrier due to its proton sponge effect, leading to 
endosomal rupture without releasing the ONs as well as graft - pDMAEMA – PEG 
[poly(2 - dimethylamino)ethyl methacrylate - co - aminoethyl methacrylate – bearing 
polyethylene glycol chains]. According to the authors, probably cationic polymers 
are more effi cient than cationic lipids, but lots of work still needs to be done on the 
matter. 
Ovarian Cancer Ovarian cancers start at the ovaries [448] . They can be either 
benign, and so never spread from the ovary, or malignant, in which case they can 
Percent of cells 
Viable cells 
Apoptosis cells 
Necrotic cells 
Control ATRA Bare 
DOTAP/ 
cholesterol 
liposome 
ATRA/ 
DOTAR/ 
cholesterol 
liposome 
H2O2 
0 
20 
40 
60 
80 
100 
120 
FIGURE 16 Flow cytometric analysis of A549 cells treated with 1.0 . M ATRA, bare 
DOTAP/cholesterol liposomes, or ATRA incorporated in DOTAP/cholesterol liposomes for 
48 h in A549 cells. As a control. Cells were incubated with 1% DMSO and 400 . M H 2 O 2 . 
Signifi cant differences: ( * ) P < 0.05 vs. control; (#) P < 0.05 vs. ATRA; ( ‡ ) P < 0.05 vs. DOTAP/ 
cholesterol liposomes. ( Reprinted from ref. 446 after permission of Elsevier .) 

metastasize to other parts of the body. Malignant cancers are classifi ed into germ 
cell, stromal, and epithelial tumors. Germ cells produce eggs, stromal cells produce 
progesterone and estrogen, and epithelial cells cover the ovary. The majority of the 
malignant cases are epithelial cancer (almost 90%). As with most tumors, the usual 
treatment is surgery, chemotherapy, and radiotherapy. 
Chemotherapy refers to drug administration with highly serious side effects, such 
as nausea, hand and foot rashes, mouth sores, and increased risk of infection, easy 
bruising, and so on. Therefore, liposomal carriers have been used in order to improve 
the drug ’ s biodistribution and protect the patient from those side effects. The 
main anticancer drugs used to treat ovarian cancer are carboplatin and cisplatin, 
paclitaxel, topotecan, and lurtotecan. PEGylated liposomal doxorubicin has been 
approved as a regimen for patients with metastatic ovarian cancer refractory to both 
paclitaxel and platinum based - therapy [449] . 
A number of clinical trials have been reported and many refer to a combined 
therapeutic regimen of liposomes with other anticancer drugs. PEGylated liposomal 
doxorubicin was used in a phase II trial on patients with platinum/paclitaxel pretreated 
ovarian cancer with or without topotecan. Approximately 36% of patients 
showed stable disease [420] . A phase III study followed up the one already mentioned 
in 474 patients with pretreated epithelial ovarian cancer that failed or 
recurred upon platinum - based combination chemotherapy. The patients received 
either PEGylated liposomal doxorubicin (50 mg/m 2 for 1 day every 4 weeks) or 
topotecan (1.5 mg/m 2 for 1 – 5 days every 3 weeks). The response rates and the overall 
progression - free survival (PFS) were similar for the two formulations. The platinum - 
sensitive patients showed longer PFS and overall survival of 108 weeks against the 
72 weeks obtained using topotecan. The interesting result of the study was the 
reduced cytotoxicity recorded for PEG – liposomal DXR compared to topotecan, 
proving that the former treatment improves patient quality of life. 
A phase III study was conducted to compare PEG – liposomal DXR (50 mg/m 2 
every 4 weeks) with paclitaxel (175 mg/m 2 every 3 weeks) using 214 patients 
with relapse after fi rst - line platinum - based chemotherapy [420] . As previously, 
the response rates and PFS were not signifi cantly different, but again the liposomal 
formulation was notably less toxic as fewer patients recorded with grade 
4 adverse effects (17% compared to 71% for topotecan) and thus was more 
tolerable. 
The most common toxic effects associated with liposomal DXR treatment were 
hand - and - foot syndrome and stomatitis, which can be handled by modifying the 
dose so there is no need for the regimen to cease. For example, administration of 
40 mg/m 2 liposomal DXR every 4 weeks reduces the incidences of hand - and - foot 
syndrome and stomatitis compared to 50 mg/m 2 liposomal DXR every 4 weeks 
without loss of drug potency [450] . A series of clinical trials using patients with a 
variety of ovarian cancers in terms of characteristics (relapsed, refractory, platinum, 
paclitaxel resistant) were carried out. The optimized dose regimen for PEGylated 
liposomal doxorubicin was 10 – 12.5 mg/m 2 per week when it is used as a single 
therapy. Combining liposomal DXR with other anticancer drugs is an equally or 
more effective strategy due to the lower dosage regimen of the two formulations 
avoiding relevant toxicities. 
While platinum compounds and paclitaxel comprise the fi rst - line treatment in 
combination with PEGylated liposomal doxorubicin for patients with ovarian 
APPLICATIONS OF LIPOSOMES IN THERAPEUTICS 503

504 LIPOSOMES AND DRUG DELIVERY 
cancer, vinorelbine has been used in combination with liposomes as a pallia - 
tive second - line therapy for patients with refractory/resistant ovarian cancer to 
platinum – paclitaxel [451] . The best MTD (mean therapeutic dose) was 25 mg/m 2 
at days 1 and 8 for vinorelbine and 30 mg/m 2 at day 1 for liposomal DXR of every 
21 days, which was well tolerated with moderate hematologic and mild nonhematologic 
toxicities. A phase II study relevant to the clinical effi cacy, toxicity, and 
pharmacokinetics of that combined therapeutic regimen was carried out by 
Katsaros et al. in 30 patients with platinum – paclitaxel pretreated recurrent ovarian 
cancer [452] . Caelyx (30 mg/m 2 ) and vinorelbine (30 mg/m 2 ) were administered every 
3 weeks for six cycles. The regimen was proved of signifi cant activity for patients 
pretreated with paclitaxel – platinum fi rst - line therapy. The overall response rate was 
37% with 10% of patients demonstrating stable disease. Vinorelbine bioavailability 
was higher under the current regimen. The overall survival was 9 months while the 
toxicity was mild and reversible. There were no treatment - related deaths and there 
were only 2 patients, one reported with grade 4 and the other with grade 3 hand – foot 
syndrome. Also, the toxicity due to liposomal formulation was much lower compared 
to that reported from a phase III study with the drug given as a single agent 
[453] . In another phase II study, the combination of liposomal doxorubicin and 
infused topotecan was studied in 27 patients with platinum - resistant ovarian cancer 
in two cohorts [454] . Liposomal DXR (30 mg/m 2 at day 1) and topotecan (1 mg/m 2 
for 5 days) were infused and the cycle was repeated every 21 days. The overall 
response rate of the regimen was 28% and the median overall survival was 40 weeks. 
However, neutropenia and thrombopenia were observed at 70 and 41% of the 
patients. Therefore, the topotecan dose was reduced to 0.75 mg/m 2 and liposomal 
DXR was increased to 40 mg/m 2 in order to reduce the toxicity. After that, the cytotoxic 
effect due to liposomal doxorubicin increased and the bone marrow cytotoxicity 
remained the same despite the effectiveness of the regimen. 
Lurtotecan is a more advanced camptothecin analogue with probably greater 
potency with regard to topotecan and therefore was encapsulated in liposomes to 
investigate the toxicity and pharmacokinetics [455] . In a multi - institutional open - 
label phase II study, 22 patients with topotecan - resistant ovarian cancer were administered 
liposomal lurtotecan in a dose of 2.4 mg/m 2 on days 1 and 8 every 21 days. 
Although the toxicity profi le of the drug was lower, no response was observed. 
Others used a regimen of liposomal delivery at days 1 and 3 with more promising 
therapeutic results, but higher toxicity, too [456] . 
A variety of new molecules either in combination with liposomal doxorubicin or 
not are in development at the moment [457] . For example, a phase III study will be 
conducted to test the effi cacy and safety of pattupilone versus PEG – liposomal DXR 
in taxane/platinum refractory/resistant patients with recurrent epithelial ovarian, 
primary fallopian, or primary peritoneal cancer. A phase III randomized study of 
Telcyta with Doxil/Caelyx versus Doxil/Caelyx has been planned in patients with 
platinum - refractory or platinum - resistant ovarian cancer. A phase II study relevant 
to side effects and best dose of ixabepilone combined with liposomal DXR will be 
assessed in patients with advanced ovarian epithelial, peritoneal cavity, or fallopian 
tube cancer or metastatic breast cancer. 
Gemcitabine is a clinically active antineoplastic drug in platinum - refractory 
ovarian cancer. The effi cacy and tolerability of the particular drug in combination 
with liposomal DXR were investigated in athymic mice bearing cisplatin - resistant 
human ovarian carcinoma [458] . 

Using two therapeutic regimens, either 80 mg/kg of gemcitabine and 15 mg/kg of 
liposomes or 20 mg/kg of gemcitabine and 6 mg/kg of liposomes, the same trend of 
response was observed, with some of the animals having complete tumor regression 
at the end of the study. The lack of toxicity and therapeutic effi cacy observed makes 
that regimen promising for clinical trials. 
In terms of drug development, a novel analogue of vitamin E assembled into 
liposomes was evaluated as a potent anticancer agent in combination with cisplatin 
in mice bearing human ovarian cancer xenografts [459] . The analogue is the 2,5,7,8 - 
tetramethyl - 2 R - (4 R , 8 R , 12 - trimethyltridecyl)chimoran - 6 - yloxyacetic acid ( . - TEA), 
which has apoptotic properties to cancer and not the normal cells in a dose - and 
time - dependent manner. Liposomes were administered in the form of aerosol while 
cisplatin was injected intraperitoneally to mice with either small or large tumor 
volume. It was shown that the combined therapeutic scheme demonstrated the 
highest antitumor activity compared to . - TEA itself in both cases. Also, the current 
regimen signifi cantly reduced micrometastases observed in lungs and lymph nodes, 
while the ratio of proliferating to apoptotic cells in the tumor was decreased due to 
induction of apoptosis. However, the specifi c apoptotic mechanism of the particular 
molecule needs to be elucidated. 
By grafting folate molecules on the liposome surface, their accumulation is highly 
increased in macrophages located in tumor ascites fl uid but not in solid tumors, 
which have been tested and the existence of functional folate receptors has been 
confi rmed [460] . Folate receptors are overexpressed on epithelial tumor cells, and 
thus folate was attached on PEG molecules via cysteine (folate – cysteine – PEG3400 – 
PE), which, consequently, was one of the components of the liposomal bilayer (PC/ 
Chol/PEG – PE). The ligand - bearing liposomes (diameter of 65 – 90 nm) were injected 
i.p. in tumor - bearing mice. Signifi cant folate - bearing liposome accumulation was 
obtained in the ascites fl uid and more specifi cally by macrophages, which indicates 
macrophage activation. A reduced targeted liposome accumulation in the tumor 
could occur for a number of reasons, such as poor liposome penetration into solid 
tumor mass. Activated macrophages secrete immunosuppressive cytokines and 
angiogenic factors, so liposome targeting could possibly eliminate them during 
malignancy therapy. 
Gene delivery is another approach trying to tackle the problem of cancer. Mutations 
of p53 tumor suppressor gene contribute to genetic abnormalities in ovarian 
cancer. Kim et al. developed a nonviral vector for delivery of p53 in ovarian cancer 
cells (OVCAR - 3) [461] . The nonviral (liposomal) vector consisted of DOTAP, 
DOPE, and Chol in the molar ratio 1 : 0.7 : 0.3. High expression of p53 mRNA and 
proteins in OVCAR - 3 cells indicated successful transfection of the lipoplexes to the 
cells used. The latter was indicated with the inhibition of cell growth obtained in 
OVCAR - 3 cells due to apoptosis caused by that protein. Intratumoral injection of 
DDC/pp53 - GFAP lipoplexes into mice bearing the OVCAR - 3 cells clearly showed 
the tumor growth inhibition, which suggests the therapeutic effi ciency of the particular 
lipoplexes. 
The neutral 1,2 - dioleoyl - sn - glycerol - 3 - phosphatidylcholine (DOPC) was used to 
make complexes with the siRNA targeting the oncoprotein EphA2 and then injected 
i.v. into ovarian tumor - bearing mice [462] . According to the authors, the signifi cance 
of the particular study is the formulation of siRNA in liposomes with successful 
outcome after delivery of the loaded carrier, which showed signifi cant reduced 
protein expression and tumor growth (Figure 17 ). 
APPLICATIONS OF LIPOSOMES IN THERAPEUTICS 505

506 LIPOSOMES AND DRUG DELIVERY 
REFERENCES 
1. Bangham , A. D. , Standish , M. M. , and Watkins , J. C. ( 1965 ), Diffusion of univalent ions 
across the lamellae of swollen phospholipids , J. Mol. Biol. , 13 , 238 – 252 . 
2. Papahadjopoulos , D. ( 1978 ), Liposomes and their uses in biology and medicine , Ann. 
N.Y. Acad. Sci. , 408 , 1 . 
3. Gregoriadis , G. ( 1978 ), Liposomes in therapeutic and preventive medicine: The development 
of the drug - carrier concept , Ann. N.Y. Acad. Sci. , 308 , 343 – 370 . 
4. Kirby , C. , Clarke , J. , and Gregoriadis , G. ( 1980 ), Cholesterol content of small unilamellar 
liposomes controls phospholipid loss to high density lipoproteins in the presence of 
serum , FEBS Lett. , 111 , 324 – 328 . 
5. Pain , D. , Das , P. K. , Ghosh , P. C. , and Bachhawat , B. K. ( 1984 ), Increased circulatory 
half - life of liposomes after conjugation with dextran , J. Biosci. , 6 , 811 – 816 . 
FIGURE 17 Therapeutic effi cacy of siRNA - mediated EphA2 down regulation. A and B 
nude mice were injected i.p. with ( a ) 2.5 . 105 HeyA8 cells or ( b ) 1.0 . 106 SKOV3ip1 cells 
and randomly allocated to one of fi ve groups, with therapy beginning 1 week after cell injection. 
(a) Empty DOPC liposomes, (b) control siRNA in DOPC, (c) EphA2 - targeting siRNA 
in DOPC, (d) paclitaxel + control siRNA in DOPC, or (e) paclitaxel + EphA2 siRNA in 
DOPC. siRNA liposomes were injected twice weekly at a dose of 150 . g/kg siRNA. Paclitaxel 
(100 . g) or vehicle (fi rst three groups) was injected i.p. once weekly. ( Reprinted from ref. 462 
and with permission of the American Association for Cancer Research .) 
* 
+++++++++ 
+++ 
§ 
§ 
†
* 
* 
*
* 
†
† * 
(a) 
(b) 
HeyA8: Mean weights Individual weights 
SKOV3ip1: Mean weights Individual weights 
Tumor weight (g) 
Empty liposomes 
Control siRNA 
EphA2 siRNA 
Paclitaxel +control siRNA 
Paclitaxel +EphA2 siRNA 
Empty liposomes 
Control siRNA 
EphA2 siRNA 
Paclitaxel +control siRNA 
Paclitaxel +EphA2 siRNA 
4.0 
3.0 
2.0 
1.0 
0.0 
5.0 
4.0 
3.0 
2.0 
1.0 
0.0 
1.5 
1.0 
0.5 
0.0 
1.5 
1.0 
0.5 
0.0 
p<0.05 compared to control siRNA 
p<0.05 compared to empty liposomes 
p<0.05 compared to control siRNA plus paclitaxel 
2.40 
0.81 0.35 
0.70 0.22 
0.04 
1.51 
0.98 0.84 
0.21

REFERENCES 507 
6. Allen , T. M. , and Chonn , A. ( 1987 ), Large unilamellar liposomes with low uptake into 
the reticuloendothelial system , FEBS Lett. , 223 , 42 – 46 . 
7. Papahadjopoulos , D. , Allen , T. M. , Gabizon , A. , Mayhew , E. , Matthay , K. , Huang , S. K. , 
et al . ( 1991 ), Sterically stabilized liposomes: Improvements in pharmacokinetics and 
antitumor therapeutic effi cacy , Proc. Natl. Acad. Sci. USA , 88 , 11460 – 11464 . 
8. Lasic , D. D. , Martin , F. J. , Gabizon , A. , Huang , S. K. , and Papahadjopoulos , D. ( 1991 ), 
Sterically stabilized liposomes: A hypothesis on the molecular origin of the extended 
circulation times , Biochim. Biophys. Acta , 1070 , 187 – 192 . 
9. Torchilin , V. P. , Levchenko , T. S. , Whiteman , K. R. , Yaroslavov , A. A. , Tsatsakis , A. M. , 
Rizos , A. K. , et al . ( 2001 ), Amphiphilic poly - N - vinylpyrrolidones: Synthesis, properties 
and liposome surface modifi cation , Biomaterials , 22 , 3035 – 3044 . 
10. Takeuchi , H. , Kojima , H. , Yamamoto , H. , and Kawashima , Y. ( 2001 ), Evaluation of circulation 
profi les of liposomes coated with hydrophilic polymers having different molecular 
weights in rats , J. Controlled Release , 75 , 83 – 91 . 
11. Haran , G. , Cohen , R. , Bar , L. , and Barenholz , Y. ( 1993 ), Transmembrane ammonium 
sulphate gradients in liposomes produce effi cient and stable entrapment of amphipathis 
weak bases , Biochim. Biophys. Acta , 1151 , 201 – 215 . 
12. Loan Honeywell - Nguyen , P. , Wouter , H. W. , Groenink , A. , de Graaff , M. , and Bouwstra , 
J. A. ( 2003 ), The in vivo transport of elastic vesicles into human skin: Effects of occlusion, 
volume and duration of application , J. Controlled Release , 90 , 243 – 255 . 
13. Lasic , D. ( 1993 ), Liposomes from Physics to Applications , Elsevier , London . 
14. Hauser , H. ( 2000 ), Short - chain phospholipids as detergents , Biochim. Biophys. Acta , 
1508 , 164 – 181 . 
15. Eibl , H. ( 1980 ), Synthesis of glycerophospholipids , Chem. Phys. Lipids , 26 , 405 – 429 . 
16. Barenholz , Y. , Thompson , T. E. ( 1999 ), Sphingomyelin: Biophysical aspects , Chem. Phys. 
Lipids , 102 , 29 – 34 . 
17. Barenholz , Y. ( 2004 ), Sphingomyelin and cholesterol: From membrane biophysics and 
rafts to potential medical applications , Subcell. Biochem. , 37 , 167 – 215 . 
18. Patel , G. B. , and Sprott , G. D. ( 1999 ), Archaeobacterial ether lipid liposomes 
(archaeosomes) as novel vaccine and drug delivery systems , Crit. Rev. Biotechnol. , 19 , 
317 – 357 . 
19. Fatouros , D. , Ioannou , P. V. , and Antimisiaris , S. G. ( 2006 ), Novel nanosized arsenic 
containing vesicles for drug delivery: Arsonoliposomes , J. Nanosci. Nanotechnol. , 6 , 
2618 – 2687 . 
20. Biltonen , R. L. , and Lichtenberg , D. ( 1993 ), The use of differential scanning calorimetry 
as a tool to characterize liposome preparations , Chem. Phys. Lipids , 64 , 129 – 142 . 
21. Champan , D. , Urbina , J. , and Keough , K. M. ( 1974 ) , Studies of lipid water systems using 
differential scanning calorimetry , J. Biol. Chem. , 249 ( 8 ), 2512 – 2521 . 
22. van Osdol , W. W. , Johnson , M. L. , Ye , Q. , and Biltonen M. ( 1991 ), Relaxation dynamics 
of the gel to liquid - crystalline transition of phosphatidylcholine bilayers. Effects of chain 
length and vesicle size , Biophys. J. , 59 , 775 – 785 . 
23. van Osdol , W. W. , Ye , Q. , Johnson , M. L. , and Biltonen , M. ( 1992 ), Effects of the anesthetic 
dibucaine on the kinetics of the gel - liquid crystalline transition of dipalmitoylphosphatidylcholine 
multilamellar vesicles , Biophys. J. , 63 , 1011 – 1017 . 
24. Momo , F. , Fabris , S. , Bindoli , A. , Scutari , G. , and Stevanato , R. ( 2002 ), Different effects 
of propofol and nitrosopropofol on DMPC multilamellar liposomes , Biophys. Chem. , 95 , 
145 – 155 . 

508 LIPOSOMES AND DRUG DELIVERY 
25. Zhang , F. , and Rowe , E. S. ( 1994 ), Calorimetric studies of the interactions of cytochrome 
c with dioleoylphosphatidylglycerol extruded vesicles: Ionic strength effects , Biochim. 
Biophys. Acta , 1193 , 219 – 225 . 
26. Lo , Y. L. , and Rahman , Y. E. ( 1995 ), Protein location in liposomes, a drug carrier: A 
prediction by differential scanning calorimetry , J. Pharm. Sci. , 84 , 805 – 814 . 
27. Parente , R. A. , and Lentz , B. R. ( 1985 ), Advantages and limitations of 1 - palmitoyl - 2 - [[2 - 
[4 - (6 - phenyl - trans - 1,3,5 - hexatrienyl)phenyl]ethyl]carbonyl] - 3 - sn - phosphatidylcholine 
as a fl uorescent membrane probe , Biochemistry , 24 , 6178 – 6185 . 
28. Metso , A. J. , Zhao , H. , Tuunainen , I. , and Kinnnunen , P. K. ( 2004 ), Characterization of 
the main transition of dinervonoylphosphocholine liposomes by fl uorescence spectroscopy 
, Biochim. Biophys. Acta , 1663 , 222 – 231 . 
29. van Langen , H. , Van Ginkel , G. , Shaw , D. , and Levine , Y. K. ( 1989 ), The fi delity of 
response by 1 - [4 - (trimethylammonio)phenyl] - 6 - phenyl - 1,3,5 - hexatriene in time - resolved 
fl uorescence anisotropy measurements on lipid vesicles. Effects of unsaturation, headgroup 
and cholesterol on orientational order and reorientational dynamics , Eur. Biophys. 
J. , 17 , 37 – 48 . 
30. Davenport , L. , and Targowski , P. ( 1996 ), Submicrosecond phospholipid dynamics using 
a long - lived fl uorescence emission anisotropy probe , Biophys. J. , 71 , 1837 – 1852 . 
31. de Kruijff , B. , and Cullis , P. R. ( 1980 ), The infl uence of poly( l - lysine) on phospholipid 
polymorphism. Evidence that electrostatic polypeptide - phospholipid interactions can 
modulate bilayer/non - bilayer transitions , Biochim. Biophys. Acta , 601 , 235 – 240 . 
32. Vasilenko , I. , de Kruijff , B. , and Verkleij , A. J. ( 1982 ), The synthesis and use of thionphospholipids 
in 31P - NRM studies of lipid polymorphism , Biochim. Biophys. Acta , 685 , 
144 – 152 . 
33. Perkins , W. R. , Mincey , S. R. , Ostro , M. J. , Taraschi , T. F. , and Janoff , A. S. ( 1988 ), The 
captured volume of multilamellar vesicles , Biochim. Biophys. Acta , 943 , 103 – 107 . 
34. Davis , J. H. ( 1983 ), The description of membrane lipid conformation, order and dynamics 
by 2H - NMR , Biochim. Biophys. Acta , 737 , 117 – 171 . 
35. Wu , W. G. , Dowd , S. R. , Simplaceanu , V. , Peng , Z. Y. , and Ho , C. ( 1985 ), 19F NMR 
investigation of molecular motion and packing in sonicated phospholipid vesicles , 
Biochemistry , 24 , 7153 – 7161 . 
36. Bertoli , E. , Masserini , M. , Sonnino , S. , Ghidoni , R. , Cestaro , B. , and Tettamanti , G. ( 1981 ), 
Electron paramagnetic resonance studies on the fl uidity and surface dynamics of egg 
phosphatidylcholine vesicles containing gangliosides , Biochim. Biophys. Acta , 647 , 
196 – 202 . 
37. Shin , Y. K. , and Freed , J. H. ( 1989 ), Dynamic imaging of lateral diffusion by electron spin 
resonance and study of rotational dynamics in model membranes. Effect of cholesterol , 
Biophys. J. , 55 , 537 – 550 . 
38. Lai , C. S. , Joseph , J. , and Shih , C. C. ( 1989 ), Molecular dynamics of antitumor ether - linked 
phospholipids in model membranes: A spin - label study , Biochem. Biophys. Res. Commun. , 
160 , 1189 – 1195 . 
39. Ondrias , K. , Stasko , A. , Marko , V. , and Nosal , R. ( 1989 ), Infl uence of beta - adrenoceptor 
blocking drugs on lipid - protein interaction in synaptosomal membranes. An ESR study , 
Chem. Biol. Interact. , 69 , 87 – 97 . 
40. Hoekstra , D. ( 1982 ), Fluorescence method for measuring the kinetics of Ca 2+ - induced 
phase separations in phosphatidylserine - containing lipid vesicles , Biochemistry , 21 , 
1055 – 1061 . 
41. Hoekstra , D. ( 1990 ), Fluorescence assays to monitor membrane fusion: Potential 
application in biliary lipid secretion and vesicle interactions , Hepatology , 12 , 61S – 
66S . 

REFERENCES 509 
42. LeNeveu, D. M. , Rand, R. P. , Parsegian, V. A. , and Gingel, D. (1976), Apparent modifi cation 
of forces between lecithin bilayers , Science , 191 , 399 – 400 . 
43. Franks , N. P. ( 1976 ), Structural analysis of hydrated egg lecithin and cholesterol bilayers. 
I. X - ray diffraction , J. Mol. Biol. , 100 , 345 – 358 . 
44. Worcester , D. L. , and Franks , N. P. ( 1976 ), Structural analysis of hydrated egg lecithin 
and cholesterol bilayers. II. Neutron diffraction , J. Mol. Biol. , 100 , 359 – 378 . 
45. LeNeveu , D. M. , and Rand , R. P. ( 1977 ), Measurement and modifi cation of forces 
between lecithin bilayers , Biophys. J. , 18 , 209 – 230 . 
46. Hobbs , S. K. , Monsky , W. L. , Yuan , F. , Roberts , W. G. , Griffi th , L. , Torchillin , V. P. , et al . 
( 1998 ), Regulation of transport pathways in tumor vessels: Role of tumor type and 
microenvironment , Proc. Natl. Acad. Sci. USA , 95 , 4607 – 4612 . 
47. Drummond, D. C. , Meyer , O. , Hong , K. , Kirpotin, D. B. , and Papahadjopoulos , D. (1999 ), 
Optimizing liposomes for delivery of chemotherapeutic agents to solid tumour , 
Pharmacol. Rev. , 51 , 691 – 743 . 
48. Devine , D. V. , Wong , K. , Serrano , K. , Chonn A. , and Cullis , P. R. ( 1994 ), Liposome - 
complement interactions in rat serum: Implications for liposome survival studies , 
Biochim. Biophys. Acta , 1191 , 43 – 51 . 
49. Kolchens , S. , Ramaswami , V. , Birgenheier , J. , Nett , L. , and O ’ Brien , D. F. ( 1993 ), Quasi - 
elastic light scattering determination of the size distribution of extruded vesicles , Chem. 
Phys. Lipids , 65 , 1 – 10 . 
50. Lesieur , S. , Grabielle - Madelmont , C. , Paternostre , M. T. , and Ollivon , M. ( 1991 ), 
Size analysis and stability study of lipid vesicles by high - performance gel exclusion 
chromatography, turbidity, and dynamic light scattering , Anal. Biochem. , 1 , 192 , 334 – 
343 . 
51. Matsuzaki , K. , Murase , O. , Sugishita , K. , Yoneyama , S. , Akada , K. , Ueha , M. , Nakamura , 
A. , and Kobayashi , S. ( 2000 ), Optical characterization of liposomes by right angle light 
scattering and turbidity measurement , Biochim. Biophys. Acta , 31 , 219 – 226 . 
52. Moon, M. H. , and Giddings , J. C. (1993), Size distribution of liposomes by fl ow fi eld - fl ow 
fractionation , J. Pharm. Biomed. Anal. , 11 , 911 – 920 . 
53. Korgel , B. A. , van Zanten , J. H. , and Monbouquette , H. G. ( 1998 ), Vesicle size distributions 
measured by fl ow fi eld - fl ow fractionation coupled with multiangle light scattering , 
Biophys. J. , 74 , 3264 – 3272 . 
54. Lee , H. , Williams , S. K. , Allison , S. D. , and Anchordoquy , T. J. ( 2001 ), Analysis of self - 
assembled cationic lipid - DNA gene carrier complexes using fl ow fi eld - fl ow fractionation 
and light scattering , Anal. Chem. , 73 , 837 – 843 . 
55. Grabielle - Madelmont , C. , Lesieur , S. , and Ollivon , M. ( 2003 ), Characterization of 
loaded liposomes by size exclusion chromatography , J. Biochem. Biophys. Methods , 56 , 
189 – 217 . 
56. Goni , F. M. , and Alonso , A. ( 2000 ), Spectroscopic techniques in the study of membrane 
solubilization, reconstitution and permeabilization by detergents , Biochim. Biophys. 
Acta , 1508 , 51 – 68 . 
57. Alonso , A. , Villena , A. , and Go n i , F. M. ( 1981 ), Lysis and reassembly of sonicated lecithin 
vesicles in the presence of Triton X - 100 , FEBS Lett. , 123 , 200 – 204 . 
58. Kokona , M. , Kallinteri , P. , Fatouros , D. , and Antimisiaris , S. G. ( 2000 ), Stability of SUV 
liposomes in the presence of cholate salts and pancreatic lipases: Effect of lipid composition 
, Eur. J. Pharm. Sci. , 9 , 245 – 252 . 
59. Almog , S. , Litman , B. J. , Wimley , W. , Cohen , J. , Wachtel , E. J. , Barenholz , Y. , Ben - Saul , 
A. , and Lichtenberg , D. ( 1990 ), States of aggregation and phase transformations in 
mixtures of phosphatidylcholine and octyl glucoside , Biochemistry , 29 , 4582 – 4592 . 

510 LIPOSOMES AND DRUG DELIVERY 
60. Lopez , O. , de la Maza , A. , Coderch , L. , Lopez - Iglesias , C. , Wehrli E. , and Parra , 
J. L. ( 1998 ), Direct formation of mixed micelles in the solubilization of phospholipid 
liposomes by Triton X - 100 , FEBS Lett. , 426 , 314 – 318 . 
61. Fatouros , D. G. , Piperoudi , S. , Gortzi , O. , Ioannou , P. V. , and Antimisiaris , S. G. ( 2005 ), 
Physical stability of sonicated arsonoliposomes: Effect of calcium ions , J. Pharm. Sci. US , 
94 , 46 – 55 . 
62. Akashi , K. , Miyata , H. , Itoh . H. , and Kinoshita , K. , Jr. ( 1996 ), Preparation of giant liposomes 
in physiological conditions and their characterization under an optical microscope 
, Biophys. J. , 71 , 3242 – 3250 . 
63. Shohda , K. , Toyota , T. , Yomo , T. , and Saguwara , T. ( 2003 ), Direct visualization of DNA 
duplex formation on the surface of a giant liposome , Chembiochem. , 4 , 778 – 781 . 
64. Henriksen , J. R. , and Ipsen , J. H. ( 2004 ), Measurement of membrane elasticity by micro - 
pipette aspiration , Eur. Phys. J. E. Soft Matter , 14 , 149 – 167 . 
65. Olson , F. , Hunt , C. A. , Szoka , F. C. , Vail , W. J. , and Papahadjopoulos , D. ( 1979 ), Preparation 
of liposomes of defi ned size distribution by extrusion through polycarbonate membranes 
, Biochim. Biophys. Acta , 557 , 9 – 23 . 
66. Weibull , C. , Christiansson , A. , and Carlemalm , E. ( 1983 ), Extraction of membrane lipids 
during fi xation, dehydration and embedding of Acholeplasma laidlawii - cells for electron 
microscopy , J. Microsc. , 129 , 201 – 207 . 
67. Dermer , G. B. ( 1968 ), An autoradiographic and biochemical study of oleic acid absorption 
by intestinal slices including determinations of lipid loss during preparation of 
electron microscopy , J. Ultrastruct. Res. , 22 , 312 – 325 . 
68. Hope , M. J. , Walker , D. C. , and Cullis , P. R. ( 1983 ), Ca 2+ and pH induced fusion of small 
unilamellar vesicles consisting of phosphatidylethanolamine and negatively charged 
phospholipids: A freeze fracture study , Biochem. Biophys. Res. Commun. , 110 , 15 – 22 . 
69. Hope , M. J. , Wong , K. F. , and Cullis , P. R. ( 1989 ), Freeze - fracture of lipids and model 
membrane systems , J. Electron. Microsc. Tech. , 13 , 277 – 287 . 
70. Burger , K. N. , Nieva , J. L. , Alonso , A. , and Verkleij , A. J. ( 1991 ), Phospholipase C activity 
- induced fusion of pure lipid model membranes. A freeze fracture study , Biochim. 
Biophys. Acta , 1068 , 249 – 253 . 
71. Meyer , H. W. , and Richter , W. ( 2001 ), Freeze - fracture studies on lipids and membranes , 
Micron. , 32 , 615 – 644 . 
72. Taggar , A. S. , Alnajim , J. , Anantha , M. , Thomas , A. , Webb , M. , Ramsay , E. , and Bally , 
M. B. ( 2006 ), Copper - topotecan complexation mediates drug accumulation into liposomes 
, J. Controlled Release , 114 , 78 – 88 . 
73. Anabousi , S. , Laue , M. , Lehr , C. M. , Bakowsky , U. , and Ehrhardt . C. ( 2005 ), Assessing 
transferring modifi cation of liposomes by atomic force microscopy and tramsmission 
electron microscopy , Eur. J. Pharm. Biopharm , 60 , 295 – 303 . 
74. Bellare , J. R. , Davis , H. T. , Scriven , L. E. , and Talmon , Y. ( 1988 ), Controlled environment 
vitrifi cation system. An improved sample preparation technique , J. Elec. Micr. Tech. , 10 , 
87 – 111 . 
75. Dubochet , J. , Adrian , M. , Chang , J. , Homo , J. C. , Lepault , J. , McDowell , A. W. , and 
Schultz , P. ( 1988 ), Cryo - electron microscopy of vitrifi ed specimens , Q. Rev. Biophys. , 21 , 
129 – 228 . 
76. Frederik , P. M. , Stuart , M. C. , and Verklej , A. J. ( 1989 ), Intermediary structures during 
membrane fusion as observed by cryo - electron microscopy , Biochim. Biophys. Acta , 979 , 
275 – 278 . 
77. Siegel , D. P. , Green , W. , and Talmon , J. ( 1989 ), Intermediates in membrane fusion and 
bilayer/nonbilayer phase transitions imaged by time - resolved cryo - transmission electron 
microscopy , Biophys J. , 56 , 161 – 169 . 

REFERENCES 511 
78. Frederik , P. M. , Burger , K. N. , Stuart , M. C. , and Verklej , A. J. ( 1991 ), Lipid polymorphism 
as observed by cryo - electron microscopy , Biochim. Biophys. Acta , 1062 , 133 – 141 . 
79. Siegel , D. P. , Green , W. , and Talmon , J. ( 1994 ), The mechanism of lamellar - to - inverted 
hexagonal phase transitions: A study using temperature - jump cryo - electron microscopy , 
Biophys. J. , 66 , 402 – 414 . 
80. Mozafari , M. R. , Reed , C. J. , Rostron , C. , and Hasirci , V. ( 2005 ), A review of scanning 
probe microscopy investigations of liposome - DNA complexes , J. Liposome Res. , 15 , 
93 – 107 . 
81. Papadimitriou , E. , and Antimisiaris , S. G. ( 2000 ), Interactions of PC/Chol and PS/Chol 
liposomes with human cells in vitro , J. Drug Target. , 5 , 335 – 351 . 
82. Haywood , A. M. , and Boyer , B. P. ( 1986 ), Ficoll and dextran enhance adhesion of 
Sendai virus to liposomes containing receptor (ganglioside GD1a) , Biochemistry , 25 , 
3925 – 3929 . 
83. Shichijo , S. , and Alving , C. R. ( 1986 ), Inhibitory effects of gangliosides on immune reactions 
of antibodies to neutral glycolipids in liposomes , Biochim. Biophys. Acta , 858 , 
118 – 124 . 
84. Ho , R. J. , Rouse , B. T. , and Huang , L. ( 1986 ), Target - sensitive immunoliposomes: Preparation 
and characterization , Biochemistry , 25 , 5500 – 5506 . 
85. Bogdanov , A. A. , Gordeeva , L. V. , Torchilin , V. P. , and Margolis , L. B. ( 1989 ), Lectin - 
bearing liposomes: Differential binding to normal and to transformed mouse fi broblasts , 
Exp. Cell Res. , 181 , 362 – 374 . 
86. Woodle , M. C. , and Lasic , D. D. ( 1992 ), Sterically stabilized liposomes , Biochim. Biophys. 
Acta , 1113 , 171 – 199 . 
87. Woodle , M. C. , Collins , L. R. , Sponsler , E. , Kossovsky , N. , Papahadjopoulos , D. , and 
Martin , F. J. ( 1992 ), Sterically stabilized liposomes. Reduction in electrophoretic mobility 
but not electrostatic surface potential , Biophys. J. , 61 , 902 – 910 . 
88. Mayhew , E. , Lazo , R. , and Vail , W. J. ( 1989 ), Preparation of liposomes entrapping cancer 
chemotherapeutic agents for experimental in vivo and in vitro studies , in Gregoriadis , 
G. , Ed., Liposome Technology , Vol. II , CRC Press , Boca Raton, FL , Chapter 2. 
89. Fatouros , D. G. , and Antimisiaris , S. G. ( 2001 ), Physicochemical properties of liposomes 
incorporating hydrochlorothiazide and chlorothiazide , J. Drug Target. , 9 , 61 – 74 . 
90. Fatouros , D. G. , and Antimisiaris , S. G. ( 2002 ), Effect of amphiphilic drugs on the stability 
and zeta - potential of their liposome formulations: A study with prednisolone, diazepam 
and griseofulvin , J. Coll. Interf. Sci. , 251 , 271 – 277 . 
91. Szoka , F. C. , and Papahadjopoulos , D. ( 1980 ), Comparative properties and methods of 
preparation of lipid vesicles (liposomes) , Ann. Rev. Biophys. Bioeng. , 75 , 4194 – 4199 . 
92. Kulkarni , S. B. , Betageri , G. V. , and Singh , M. ( 1995 ), Factors affecting microencapsulation 
of drugs in liposomes , J. Microencapsul. , 12 , 229 – 246 . 
93. Amselem , S. , Gabizon , A. , and Barenholz , Y. ( 1990 ), Optimization and up scaling of 
doxorubicin - containing liposomes for clinical use , J. Pharm. Sci. , 79 , 1045 – 1052 . 
94. Mezei , M. , and Nugent , F. J. ( 1984 ), Method of encapsulating biologically active materials 
in multilmellar lipid vesicles, U.S. Patent 4, 485,054 . 
95. Ma , L. , Ramachandran , C. , and Weiner , N. D. ( 1991 ), Partitioning of a homologous series 
of alkyl p - amino benzoates in dipalmitoyl phosphatidyl choline liposomes: Effect of 
liposomes type , Int. J. Pharm. , 70 , 209 – 218 . 
96. Betageri , G. V. , and Parsons , D. L. ( 1992 ), Drug encapsulation and release from multilamellar 
and unilamellar liposomes , Int. J. Pharm. , 81 , 235 – 241 . 

512 LIPOSOMES AND DRUG DELIVERY 
97. Puglisi , G. , Fresta , M. , La Rosa , C. , Ventura , C. A. , Panic , A. M. , and Mazzonne , A. ( 1992 ), 
Liposomes as potential drug carrier for citicoline (CDP - choline) and the effect of formulation 
conditions on encapsulation effi ciency , Pharmazie , 47 , 211 – 215 . 
98. Betageri , G. V. ( 1993 ), Liposomal encapsulation and stability of dideoxynosine triphosphate 
, Drug Dev. Ind. Pharm. , 19 , 531 – 539 . 
99. Fresta , M. , Villari , A. , Puglisi , G. , and Cavallaro , G. ( 1993 ), 5 - Fluorouracil: Various kinds 
of loaded liposomes: Encapsulation effi ciency, storage stability and fusogenic properties , 
Int. J. Pharm. , 99 , 145 – 156 . 
100. Elorza , B. , Elorza , M. A. , Frutos , G. , and Chantres , J. R. ( 1993 ), Characterization 
of 5 - fl ourouracil loaded liposomes prepared by reverse phases evaporation or 
freezing thawing extrusion methods: Study of release , Biochim. Biophys. Acta , 1153 , 
135 – 142 . 
101. Law , S. L. , Chang , P. , and Lin , C. H. ( 1991 ), Characteristics of mitoxantrone loading on 
liposomes , Int. J. Pharm. , 70 , 1 – 7 . 
102. Alpar , O. H. , Bamford , J. B. , and Walters , V. ( 1981 ), In vitro incorporation and release 
of hydroxycobalamin by liposomes , Int. J. Pharm. , 7 , 349 – 351 . 
103. Crommelin , D. J. A. , and Van Bloois , L. ( 1983 ), Preparation and characterization of 
doxorubicin containing liposomes. Part 2. Loading capacity, long - term stability and 
doxorubicin bilayer interaction mechanism , Int. J. Pharm. , 17 , 135 – 144 . 
104. Arien , A. , Coigoux , C. , Baquey , C. , and Dupuy , B. ( 1993 ), Study of in vitro and in vivo 
stability of liposomes loading with calcitonin or indium in GIT , Life Sci. , 53 , 
1279 – 1290 . 
105. D ’ Silva , J. B. , and Notari , R. E. ( 1982 ), Drug stability in liposomal suspensions: Hydrolysis 
of indomethacin, cyclocytidine and p - nitrophenyl acetate , J. Pharm. Sci. , 71 , 1394 – 
1398 . 
106. Kirby , C. , Clarke , J. , and Gregoriadis , G. ( 1980 ), Effect of the cholesterol content of 
small unilamellar liposomes on their stability in vivo and in vitro , Biochem. J. , 186 , 
591 – 595 . 
107. Kirby , C. , and Gregoriadis , G. ( 1980 ), The effect of the cholesterol content of small 
unillamellar liposomes on the fate of their lipid components in vivo , Life Sci. , 27 , 
2223 – 2230 . 
108. Layton , D. , and Trouet , A. ( 1980 ), A comparison of the therapeutic effects of free and 
liposomally encapsulated vincristine in leucemic mice , Eur. J. Cancer , 16 , 945 – 951 . 
109. McCormack , B. , and Gregoriadis , G. ( 1994 ), Drugs - in - cylcodextrins - liposomes: A novel 
concept in drug delivery , Int. J. Pharm. , 112 , 249 – 258 . 
110. McCormack , B. , and Gregoriadis , G. ( 1996 ), Comparative studies of the fate of free and 
liposome - entrapped hydroxypropyl - . - cyclodextrin - drug complexes after intravenous 
injection into rats: Implications in drug delivery , Biochim. Biophys. Acta , 1291 , 237 – 
244 . 
111. Fatouros , D. G. , Hatzidimitirou , K. , and Antimisiaris , S. G. ( 2001 ), Liposomes encapsulating 
prednisolone and prednisolone - cyclodextrin complexes: Comparison of membrane 
integrity and drug release , Eur. J. Pharm. Sci. , 13 , 287 – 296 . 
112. Grit , M. , and Crommelin , D. J. A. ( 1993 ), Analysis and hydrolysis kinetics of phospholipids 
in aqueous liposome dispersions , in Gregoriadis G. , Ed., Liposome Technology , 
Vol. I , CRC Press , Boca Raton, FL , pp. 455 – 487 . 
113. Grit , M. , and Crommelin , D. J. A. ( 1993 ), Chemical stability of liposomes: Implications 
for their stability , Chem. Phys. Lipids , 64 , 3 – 18 . 
114. Zuidam , N. J. , and Crommelin , D. J. A. ( 1995 ), Chemical hydrolysis of phospholipids , 
J. Pharm. Sci. , 84 , 1113 – 1119 . 

REFERENCES 513 
115. Grit , M. , de Smidt , J. , Struijke , H. A. , and Crommelin , D. J. A. ( 1989 ), Hydrolysis of 
natural soybean phosphatidylcholine in aqueous liposome dispersions , Int. J. Pharm. , 50 , 
1 – 6 . 
116. Hunt , C. A. , and Tsang , S. ( 1981 ), . - Tocopherol retards autoxidation and prolongs the 
shelf life of liposomes , Int. J. Pharm. , 8 , 101 – 110 . 
117. Fukuzawa , K. , Chida , H. , Akira , T. , and Tsukatani , H. ( 1981 ), Autooxidative effect of . - 
tocopherol incorporation into lecithin liposomes on ascorbic acid Fe ++ - induced lipid 
peroxidation , Arch. Biochem. Biophys. , 206 , 173 – 180 . 
118. Talsma , H. , van Steenbergen , M. J. , and Crommelin , D. J. A. ( 1992 ), The cryopreservation 
of liposomes. 2. Effect of particle size on crystallization behavior and marker retention , 
Cryobiology , 29 , 80 – 86 . 
119. Talsma , H. , van Steenbergen , M. J. , and Crommelin , D. J. A. ( 1991 ), The cryopreservation 
of liposomes. 3. Almost complete retention of a water - soluble marker in small liposomes 
in a cryoprotectant containing dispersion after a freezing/thawing cycle , Int. J. Pharm. , 
77 , 119 – 126 . 
120. Kristiansen , J. ( 1992 ), Leakage of a trapped fl uorescent marker from liposomes: Effects 
of eutectic crystallization of NaCl and internal freezing , Cryobiology , 29 , 575 – 584 . 
121. Crowe , L. M. , and Crowe , J. H. ( 1988 ), Trehalose and dry dipalmitoloylphosphatidylcholine 
revisited , Biochim. Biophys. Acta , 946 , 193 – 201 . 
122. Crowe , J. H. , Leslie , S. B. , and Crowe , L. M. ( 1994 ), Is vitrifi cation suffi cient to preserve 
liposomes during freeze - drying? Cryobiology , 31 , 355 – 366 . 
123. Piel , G. , Piette , M. , Barillaro , V. , Castagne , D. , Evrard , B. , and Delattre , L. ( 2006 ), Betamethasone 
- in - cyclodextrin - in - liposome: The effect of cyclodextrins on encapsulation 
effi ciency and release kinetics , Int. J. Pharm. , 312 , 75 – 82 . 
124. Woodle , M. C. , and Papahadjopoulos , D. ( 1989 ), Liposome preparation and size characterization 
, Meth. Enzym. , 171 , 193 – 217 . 
125. New , R. R. C. , Ed. ( 1989 ), Liposomes. A Practical Approach , IRL Press , Oxford. UK . 
126. Bangham , A. D. , and Horne , R. W. ( 1964 ), Negative staining of phospholipids and their 
structured modifi cation by surface active agents as observed in the electron microscope . 
J. Mol. Biol. , 8 , 660 – 668 . 
127. Papahadjopoulos , D. , and Watkins , J. C. ( 1967 ), Phospholipid model membrane 2: Permeability 
properties of hydrated liquid crystals , Biochim. Biophys. Acta , 135 , 639 – 652 . 
128. Batzri , S. , and Korn , E. D. ( 1973 ), Single bilayer vesicles prepared without sonication , 
Biochm. Biophys. Acta , 298 , 1015 – 1019 . 
129. Deamer , D. , and Bangham , A. D. ( 1976 ), Large volume liposomes by an ether vaporization 
method , Biochm. Biophys. Acta , 443 , 629 – 634 . 
130. Barenholz , Y. , Amselem , S. , and Lichtenberg , D. ( 1979 ), A new method for preparation 
of phospholipid vesicles , FEBS Lett. , 99 , 210 – 214 . 
131. Mayhew , E. , Lazo , R. , Vail , W. J. , King , J. , and Green , A. M. ( 1984 ), Characterization of 
liposomes prepared using microfl uidizer , Biochim. Biophys. Acta , 775 , 169 – 174 . 
132. Szoka , F. C. , and Papahadjopoulos , D. ( 1978 ), Procedure for preparation of liposomes 
with large internal aqueous space and high capture by reverse phase evaporation , Proc. 
Natl. Acad. Sci. USA , 75 , 4194 – 4198 . 
133. Gregoriadis , G. , Da Silva , H. , and Florence A. T. ( 1990 ), A procedure for the effi cient 
entrapment of drugs in dehydration - rehydration liposomes , Int. J. Pharm. , 65 , 235 – 242 . 
134. Zadi , B. , and Gregoriadis , G. ( 2000 ), A novel method for high - yield entrapment of 
solutes into small liposomes , J. Lipos. Res. , 10 , 73 – 80 . 
135. Reeves , J. P. , and Dowben R. M. ( 1969 ), Formation and properties of thin phospholipids 
vesicles , J. Cell. Phys. , 73 , 49 – 60 . 

514 LIPOSOMES AND DRUG DELIVERY 
136. Antimisiaris , S. G. , Jayesekera , P. , and Gregoriadis , G. ( 1993 ), Liposomes as vaccine carriers: 
Incorporation of soluble and particulate antigens in giant vesicles , J. Immunol. 
Methods , 166 , 271 – 280 . 
137. Mimms , L. T. , Zampighi , G. , Nozaki , Y. , Tanford , C. , and Reynolds , J. A. ( 1981 ), Phospholipid 
vesicles formation and transmembrane protein incorporation using octyl glucoside 
, Biochemistry , 20 , 833 – 840 . 
138. Hauser , H. , Brunner , J. , and Skrabal , P. ( 1976 ), Single bilayer vesicles prepared without 
sonication , Biochim. Biophys. Acta , 455 , 322 – 331 . 
139. Papahadjopoulos , D. , Vail , W. J. , Jacobson , K. , and Poste , G. ( 1975 ), Cochleate lipid cylinders 
formation by fusion of unilamellar lipid vesicles , Biochim. Biophys. Acta , 394 , 
483 – 491 . 
140. Talsma , H. , Van Steenbergen , M. J. , Borchert , J. C. H. , Crommelin , D. J. A. ( 1994 ), A 
novel technique for the one - step preparation of liposomes and nonionic surfactant vesicles 
without the use of organic solvents. Liposome formation in a continuous gas stream: 
The “ bubble ” method , J. Pharm. Sci. , 83 , 276 – 280 . 
141. Avestin, Inc., Ottawa, Canada , http://www.avestin.com/products.html , accessed Jan. 1, 
2004 . 
142. Mayer , L. D. , Bally , M. B. , and Cullis , P. R. ( 1990 ), Characterization of liposomal systems 
containing doxorubicin entrapped in response to pH gradients , Biochim. Biophys. Acta , 
1025 , 143 – 151 . 
143. Nichols , J. W. , and Deamer , D. W. ( 1990 ), Catecholamine uptake and concentration of 
liposomes maintaining pH gradients , Biochim. Biophys. Acta , 455 , 269 – 278 . 
144. Harrigan , P. R. , Wong , K. F. , Redelmeier , T. E. , Wheller , J. J. , and Cullis , P. R. ( 1993 ), 
Accumulation of doxorubicin and other lipophilic amines into LUVs in response to 
transmembrane pH gradient , Biochim. Biophys. Acta , 1149 , 237 – 244 . 
145. Boman , N. L. , Mayer , L. D. , and Cullis , P. R. ( 1993 ), Optimization of the retention and 
properties of vincristine in liposomal systems , Biochim. Biophys. Acta , 1152 , 253 – 258 . 
146. Gabizon , A. , Shiota , R. , and Papahadjopoulos . D. ( 1989 ), Pharmacokinetics and tissue 
distribution of doxorubicin encapsulated in stable liposomes with long circulation times , 
J. Nat. Can. Inst. , 81 , 1484 – 1488 . 
147. Gabizon , A. , Barenholz , Y. , and Bialer , M. ( 1993 ), Prolongation of the circulation time 
of doxorubicin encapsulated in liposomes containing a polyethylene glycol derivatized 
phospholipid: Pharmacokinetic studies in rodents and dogs , Pharm. Res. , 10 , 703 – 708 . 
148. Cheung , B. C. , Sun , T. H. , Leenhouts , J. M. , and Cullis , P. R. ( 1998 ), Loading of doxorubicin 
into liposomes by forming Mn 2+ – drug complexes , Biochim. Biophys. Acta , 1414 , 
205 – 216 . 
149. Abraham , S. A. , Edwards , K. , Karlsson , G. , MacIntosh , S. , Mayer , L. D. , McKenzie , C. , 
and Bally , M. B. ( 2002 ), Formation of transition metal – doxorubicin complexes inside 
liposomes , Biochim. Biophys. Acta , 1565 , 41 – 54 . 
150. Nobs , L. , Buchegger , F. , Gurny , R. , and Allemann , E. ( 2004 ), Current methods for attaching 
targeting ligands to liposomes and nanoparticles , J. Pharm. Sci. , 93 , 1980 – 1991 . 
151. Sapra , P. , and Allen , T. M. ( 2003 ), Ligand - targeted liposomal anticancer drugs , Prog. 
Lipid Res. , 42 , 439 – 462 . 
152. Klibanov , A. L. , Maruyama , K. , Beckerleg , A. M. , Torchilin , V. P. , and Huang , L. ( 1991 ), 
Activity of amphipathic poly(ethylene glycol) 5000 to prolong the circulation time of 
liposomes depends on the liposome size and is unfavorable for immunoliposome binding 
to target , Biochim. Biophys. Acta , 1062 , 142 – 148 . 
153. Maruyama , K. , Takizawa , T. , Takahashi , N. , Nagaike , K. , and Iwatsuru , M. ( 1997 ), Targeting 
effi ciency of PEG - immunoliposome - conjugated antibodies at PEG terminals , Adv. 
Drug Deliv. Rev. , 24 , 235 – 242 . 

REFERENCES 515 
154. Maruyama , K. , Takizawa , T. , Yuda , T. , Kennel , S. J. , Huang , L. , and Iwatsuru , M. ( 1995 ), 
Targetability of novel immunoliposomes modifi ed with amphipathic poly(ethylene 
glycol)s conjugated at their distal terminals to monoclonal antibodies , Biochim. Biophys. 
Acta , 1234 , 74 – 80 . 
155. Park , J. W. , Benz , C. C. , and Martin , F. J. ( 2004 ), Future directions of liposome - and 
immunoliposome - based cancer therapeutics , Semin. Oncol. , 31 ( 6 Suppl 13 ), 196 – 205 . 
156. Ishida , T. , Iden , D. L. , and Allen , T. M. ( 1999 ), A combinatorial approach to producing 
sterically stabilized (Stealth) immunoliposomal drugs , FEBS Lett. , 460 , 129 – 133 . 
157. Park , C. G. , Thiex , N. W. , Lee , K. M. , Szot , G. L. , Bluestone , J. A. , and Lee , K. D. ( 2003 ), 
Targeting and blocking B7 costimulatory molecules on antigen - presenting cells using 
CTLA4Ig - conjugated liposomes: In vitro characterization and in vivo factors affecting 
biodistribuiton , Pharm. Res. , 20 , 1239 – 1248 . 
158. Bartsch , M. , Weeke - Klimp , A. H. , Meijer , D. K. F. , Scherphof , G. L. , and Kamps , J. A. A. 
( 2002 ), Massive and selective delivery of lipid - coated cationic lipoplexes of oligonucleotides 
targeted in vivo to hepatic endothelial cells , Pharm. Res. , 19 , 676 – 680 . 
159. Chekhonin , V. P. , Zhirkov , Y. A. , Gurina , O. I. , Ryabukhin , I. A. , Lebedev , S. V. , 
Kashparov , I. A. , and Dmitriyeva , T. B. ( 2005 ), PEGylated Immunoliposomes directed 
against brain astrocytes , Drug Deliv. , 12 , 1 – 6 . 
160. Brignole , C. , Marimpietri , D. , Gambini , C. , Allen , T. M. , Ponzoni , M. , and Pastorino , F. 
( 2003 ), Development of Fab . fragments of anti - GD 2 immunoliposomes entrapping 
doxorubicin for experimental therapy of human neuroblastoma , Cancer Lett. , 197 , 
199 – 204 . 
161. Demirovic , A. M. , Marty , C. , Console , S. , Zeisberger , S. M. , Ruch , C. , Jaussi , R. , 
Schwendener , R. A. , and Ballmer - Hofer , K. ( 2005 ), Targeting human cancer cells with 
VEGF receptor - 2 - directed liposomes , Oncol. Rept. , 13 , 319 – 324 . 
162. Voinea , M. , Manduteanu , I. , Dragomir , E. , Capraru , M. , and Simionescu , M. ( 2005 ), 
Immunoliposomes directed toward VCAM - 1 interact specifi cally with activated endothelial 
cells — a potential tool for specifi c drug delivery , Pharm. Res. , 22 , 1906 – 1917 . 
163. Park , J. W. , Hong , K. , Kiprotin , D. B. , Meyer , O. , Papahadjopoulos , D. , and Benz , C. C. 
( 1997 ), Anti - HER2 immunoliposomes for targeted therapy of human tumors , Cancer 
Lett. , 118 , 153 – 160 . 
164. Mercadal , M. , Domingo , J. C. , Petriz , J. , Garcia , J. , and de Madariaga , M. A. ( 1999 ), A 
novel strategy affords high - yield coupling of antibody to extremities of liposomal 
surface - grafted PEG chains , Biochim. Biophys. Acta , 1418 , 232 – 238 . 
165. Longmuir , K. J. , Robertson , R. T. , Haynes , S. M. , Baratta , J. L. , and Waring , A. J. ( 2006 ), 
Effective targeting of liposomes to liver and hepatocytes in vivo by incorporation of a 
Plasmodium amino acid sequence , Pharm. Res. , 23 , 759 – 769 . 
166. Hansen , C. B. , Kao , G. Y. , Moase , E. H. , Zalipsky , S. , and Allen , T. M. ( 1995 ), Attachment 
of antibodies to sterically stabilized liposomes: Evaluation, comparison and optimization 
of coupling procedures , Biochim. Biophys. Acta , 1239 , 133 – 144 . 
167. Mamot , C. , Ritschard , R. , Kung , W. , Park , J. W. , Herrmann , R. , and Rochlitz , C. F. ( 2006 ), 
EGFR - targeted immunoliposomes derived from the monoclonal antibody EMD72000 
mediate specifi c and effi cient drug delivery to a variety of colorectal cancer cells , J. Drug 
Targe. , 14 , 215 – 223 . 
168. Kallinteri , P. , Papadimitriou , E. , and Antimisiaris , S. G. , ( 2001 ), Uptake of liposomes 
which incorporate a glycopeptide fraction of asialofetuin by HepG2 cells , J. Lipos. Res. , 
11 , 175 – 193 . 
169. Fretz , M. M. , Koning , G. A. , Mastrobattista , E. , Jiskoot , W. , and Storm , G. ( 2004 ), 
OVCAR - 3 cells internalize TAT - peptide modifi ed liposomes by endocytosis , Biochim. 
Biophys. Acta , 1665 , 48 – 56 . 

516 LIPOSOMES AND DRUG DELIVERY 
170. Moreira , J. N. , and Gaspar , R. ( 2004 ), Antagonist G - mediated targeting and cytotoxicity 
of liposomal doxorubicin in NCI - H82 variant small cell lung cancer , Braz. J. Med. Biol. 
Res. , 37 , 1185 – 1192 . 
171. Martin , F. J. , Hubbell , W. L. , and Papahadjopoulos , D. ( 1981 ), Immunospecifi c targeting 
of liposomes to cells: A novel and effi cient method for covalent attachment of Fab . 
fragments via disulfi de bonds , Biochemistry , 20 , 4229 – 4238 . 
172. Wartchow , C. A. , Alters , S. E. , Garzone , P. D. , Li , L. , Choi , S. , DeChene , N. E. , Doede , T. , 
Huang , L. , Pease , J. S. , Shen , Z. , Knox , S. J. , and Cleland , J. L. ( 2004 ), Enhancement of 
the effi cacy of an antagonist of an extracellular receptor by attachment to the surface 
of a biocompatible carrier , Pharm. Res. , 21 , 1880 – 1885 . 
173. Anabousi , S. , Bakowsky , U. , Schneider , M. , Huwer , H. , Lehr , C. M. , and Ehrhardt , C. 
( 2006 ), In vitro assessment of transferrin - conjugated liposomes as drug delivery 
systems for inhalation therapy of lung cancer , Eur. J. Pharm. Sci. , Jul 22 , 29 , 367 – 
374 . 
174. Lopez - Barcons , L. A. , Polo , D. , Llorens , A. , Reig , F. , and Fabra , A. ( 2005 ), Targeted 
adriamycin delivery to MXT - B2 metastatic mammary carcinoma cells by transferring 
liposomes: Effect of adriamycin ADR - to - lipid ratio , Oncol. Rept. , 14 , 1337 – 1343 . 
175. Afzelius , P. , Demant , E. J. F. , Hansen , G. H. , and Jensen , P. B. ( 1989 ), Covalent modifi cation 
of serum transferrin with phospholipids and incorporation into liposomal membranes 
, Biochim. Biophys. Acta , 979 , 231 – 238 . 
176. Stavridis , J. C. , Deliconstantinos , G. , Psallidopoulos , M. C. , Armenakas , N. A. , Hadjiminas , 
D. J. , and Hadjiminas , J. ( 1986 ), Construction of transferrin - coated liposomes for on vivo 
transport of exogenous DNA to bone marrow erythroblasts in rabbits , Exp. Cell Res. , 
164 , 568 – 572 . 
177. Torchilin , V. P. , Levchenko , T. S. , Lukyanov , A. N. , Khaw , B. A. , Klibanov , A. L. , 
Rammohan , R. , Samokhin , G. P. , and Whiteman , K. R. ( 2001 ), p - Nitrophenylcarbonyl - 
PEG - PE - liposomes: Fast and simple attachmentof specifi c ligands, including monoclonal 
antibodies, to distal ends of PEG chains via p - nitrophenylcarbonyl groups , Biochim. 
Biophys. Acta , 1511 , 397 – 411 . 
178. Savva , M. , Duda , E. , and Huang , L. ( 1999 ), A genetically modifi ed recombinant tumor 
necrosis factor - . conjugated to the distal terminals of liposomal surface grafted polyethyleneglycol 
chains , Int. J. Pharm. , 184 , 45 – 51 . 
179. Ansell , S. M. , Tardi , P. G. , and Buchkowsky , S. S. ( 1996 ), 3 - (2 - Pyridyldithio)propionic acid 
hydrazide as a cross - linker in the formation of liposome - antibody conjugates , Bioconjugate 
Chem. , 7 , 490 – 496 . 
180. Chua , M. M. , Fan , S. T. , and Karush , F. ( 1984 ), Attachment of immunoglobulin to liposomal 
membrane via protein carbohydrate , Biochim. Biophys. Acta , 800 , 291 – 300 . 
181. Koning , G. A. , Morselt , H. W. , Velinova , M. J. , Donga , J. , Gorter , A. , Allen , T. M. , Zalipsky , 
S. , Kamps , J. A. , and Scherphof , G. L. ( 1999 ), Selective transfer of a lipophilic prodrug 
of 5 - fl uorodeoxyuridine from immunoliposomes to colon cancer , Biochim. Biophys. 
Acta , 1420 , 153 – 167 . 
182. Harding , J. A. , Engbers , C. M. , Newman , M. S. , Goldstein , N. I. , and Zalipsky , S. ( 1997 ), 
Immunogenicity and pharmacokinetic attributes of poly(ethylene glycol) - grafted immunoliposomes 
, Biochim. Biophys. Acta , 1327 , 181 – 192 . 
183. Sawant , R. M. , Hurley , J. P. , Salmaso , S. , Kale , A. , Tolcheva , E. , Levchenko , T. S. , and 
Torchilin , V. P. ( 2006 ), “ SMART ” drug delivery systems: Double - targeted pH - responsive. 
Pharmaceutical nanocarriers , Bioconjugate Chem. , 17 , 943 – 949 . 
184. Xiao , Z. , McQuarrie , S. A. , Suresh , M. R. , Mercer , J. R. , Gupta , S. , and Miller , G. G. ( 2002 ), 
A three - step strategy for targeting drug carriers to human ovarian carcinoma cells in 
vitro , J. Biotechnol. , 94 , 171 – 184 . 

REFERENCES 517 
185. Schnyder , A. , Krahenbuhl , S. , Torok , M. , Drewe , J. , and Huwyler , J. ( 2004 ), Targeting 
of skeletal muscle in vitro using biotinylated immunoliposomes , Biochem. J. , 377 , 
61 – 67 . 
186. Allen , T. M. , Mumbengegwi , D. R. , and Charrois , J. R. ( 2005 ), Anti - CD19 - targeted doxorubicin 
improves the therapeutic effi cacy in murine B - cell lymphoma and ameliorates 
the toxicity of liposomes with varying drug release rates , Clin. Cancer Res. , 11 , 
3567 – 3573 . 
187. Gupta , B. , Levchenko , T. S. , Mongayt , D. A. , and Torchilin , V. P. ( 2005 ), Monoclonal 
antibody 2C5 - mediated binding of liposomes to brain tumor cells in vitro and in subcutaneous 
tumor model in vivo , J. Drug Target. , 13 , 337 – 343 . 
188. Kawakami , S. , Wong , J. , Sato , A. , Hattori , Y. , Yamashita , F. , and Hashida , M. ( 2000 ), 
Biodistribution characteristics of mannosylated, fucosylated, and galactosylated liposomes 
in mice , Biochim. Biophys. Acta , 1524 , 258 – 265 . 
189. de Melo , A. L. , Silva - Barcellos , N. M. , Demicheli , C. , and Frezard , F. ( 2003 ), Enhanced 
schistosomicidal effi cacy of tartar emetic encapsulated in pegylated liposomes , Int. J. 
Pharm. , 255 , 227 – 230 . 
190. Chen , J - H. , Ling , R. , Yao , Q. , Li , Y. , Chen , T. Wang , Z. , and Li , K - Z. ( 2005 ), Effect of 
small - sized liposomal adriamycin administered by various routes on a metastatic breast 
cancer model , Endocr. - Related Cancer , 12 , 93 – 100 . 
191. Zhang , J. Q. , Zhang , Z. R. , Yang , Q. Y. , Qin , S. R. , and Qiu , X. L. ( 2005 ), Lyophilized 
paclitaxel magnetoliposomes as a potential drug delivery system for breast carcinoma 
via parenteral administration: In vitro and in vivo , Pharm. Res. , 22 , 573 – 583 . 
192. Wang , X. P. , Yazawa , K. , Templeton , N. S. , Yang , J. , Liu , S. , Li , Z. , Li , M. , Yao , Q. , Chen , 
C. , and Brunicardi , F. C. ( 2005 ), Intravenous delivery of liposome - mediated nonviral 
DNA is less toxic than intraperitoneal delivery in mice , World J. Surg. , 29 , 339 – 343 . 
193. Kamps , J. A. A. M. , and Scherphof , G. L. ( 2004 ), Biodistribution and uptake of liposomes 
in vivo , Methods Enzymol. , 387 , 257 – 266 . 
194. Moghimi , S. M. , and Hunter , A. S. ( 2001 ), Recognition by macrophages and liver 
cells of opsonized phospholipid vesicles and phospholipid headgroups , Pharm. Res. , 18 , 
1 – 8 . 
195. Chiu , G. N. , Bally , M. B. , and Mayer , L. D. ( 2001 ), Selective protein interactions with 
phosphatidylserine containing liposomes alter the steric stabilization properties of 
poly(ethylene glycol) , Biochim. Biophys. Acta , 1510 , 56 – 69 . 
196. Harrington , K. J. , Syrigos , K. N. , and Vile , R. G. ( 2002 ), Liposomally targeted cytotoxic 
drugs for the treatment of cancer . J. Pharm. Pharmacol. , 54 , 1573 – 1600 . 
197. Forssen , E. A. , Coulter , D. M. , and Proffi tt , R. T. ( 1992 ), Selective in vivo localization of 
daunorubicin small unilamellar vesicles in solid tumours , Cancer Res. , 52 , 3255 – 3261 . 
198. Forssen , E. A. , Male - Brune , R. , Adler - Moore , J. P. , Lee , M. J. , Schmidt , P. G. , Krasieva , T. 
B. , Shimizu , S. , and Tromberg , B. J. ( 1996 ), Fluorescence imaging studies for the disposition 
of daunorubicin liposomes (DaunoXome) within tumour tissue , Cancer Res. , 56 , 
2066 – 2075 . 
199. Ogihara - Umeda , I. , Sasaki , T. , Kojima , S. , and Nishigori , H. ( 1996 ), Optimal radiolabelled 
liposomes for tumour imaging , J. Nucl. Med. , 37 , 326 – 332 . 
200. Sadzuka , Y. , and Hirota , S. ( 1998 ), Does the amount of an antitumor agent entrapped 
in liposomes infl uence its tissue distribution and cell uptake? Cancer Lett. , 131 , 
163 – 170 . 
201. Turner , A. F. , Presant , C. A. , Proffi tt , R. T. , Williams , L. E. , Winsor , D. W. , and Werner , 
J. L. ( 1988 ), In - 111 - labelled liposomes: Dosimetry and tumour depiction , Rdiology , 166 , 
761 – 765 . 

518 LIPOSOMES AND DRUG DELIVERY 
202. Presant , C. A. , Proffi tt , R. T. , Turner , A. F. , Williams , L. E. , Winsor , D. , Werner , J. L. , 
Kennedy , P. , Wiseman , C. , Gala , K. , and McKenna , R. J. ( 1988 ), Successful imaging of 
human cancer with In - 111 - labelled phospholipid vesicles , Cancer , 62 , 905 – 911 . 
203. Presant , C. A. , Blayney , D. , Proffi tt , R. D. , Turner , A. F. , Williams , L. E. , Nadel , H. I. , 
Kennedy , P. , Wiseman , C. , Gala , K. , and Crossley , R. J. ( 1990 ), Preliminary report: Imaging 
of Kaposi sarcoma and lymphoma in AIDS with indium - 111 - labelled liposomes , Lancet , 
335 , 1307 – 1309 . 
204. Kubo , A. , Nakamura , K. , Sammiya , T. , Katayama , M. , Hashimoto , T. , Hashimoto , S. , 
Kobayashi , H. , and Teramoto , T. ( 1993 ), Indium - 111 - labelled liposomes: Dosimetry and 
tumour depiction in patients with cancer , Eur. J. Nucl. Med. , 20 , 107 – 113 . 
205. Khalifa , A. , Dodds , D. , Rampling , R. , Paterson , J. , and Murray , T. , ( 1997 ), Liposomal 
distribution in malignant glioma: Possibilities for therapy , Nucl. Med. Commun. , 18 , 
17 – 23 . 
206. Zucchetti , M. , Boiardi , A. , Silvani , A. , Parisi , I. , Piccolrovazzi , S. , and D ’ Incalci , M. ( 1999 ), 
Distribution of daunorubicin and daunorubicinol in human glioma tumors after administration 
of liposomal daunorubicin , Cancer Chemother. Pharmacol. , 44 , 173 – 176 . 
207. British National Formulary (BNF) Edition 51, 2006 , Pharmaceutical Press , London . 
208. http://www.gilead.com/pdf/daxpius.pdf#search= ‘ DaunoXome ’ . Gilead Sciences, Inc. , 
2007 . 
209. Shimizu , K. , Takada , M. , Asai , T. , Kuromi , K. , Baba , K. , and Oku , N. ( 2002 ), Cancer chemotherapy 
by liposomal 6 - [[2 - (dimethylamino)ethyl]amino] - 3 - hydroxy - 7H - indeno[2,1 - 
c ]quinolin - 7 - one dihydrochloride (TAS - 103), a novel anticancer agent , Biol. Pharm. 
Bull. , 25 , 1385 – 1387 . 
210. Wu , P. C. , Tsai , Y. H. , Liao , C. C. , Chang , J. S. , and Huang , Y. B. ( 2004 ), Tha characterization 
and biodistribution of cefoxitin - loaded liposomes , Int. J. Pharm. , 271 , 31 – 39 . 
211. Alberts , D. S. , Markman , M. , Armstrong , D. , Rothenberg , M. L. , Muggia , F. , and Howell , 
S. B. ( 2002 ), Intraperitoneal therapy for stage III ovarian cancer: A therapy whose time 
has come! J. Clin. Oncol. , 20 , 3944 . 
212. Koga , S. , Hamazoe , R. , Maeta , M. , Shimizu , N. , Murakami , A. , and Wakatsuki , T. ( 1988 ), 
Prophylactic therapy for peritoneal recurrence of gastric cancer by continuous hyperthermic 
peritoneal perfusion with mitomycin C , Cancer , 61 , 232 . 
213. Speyer , J. L. ( 1985 ), The rationale behind intraperitoneal chemotherapy in gastrointestinal 
malignancies , Semin. Oncol. , 12 , 23 . 
214. Sadzuka , Y. , Hirota , S. , and Sonobe , T. ( 2000 ), Intraperitoneal administration of doxorubicin 
encapsulating liposomes against peritoneal dissemination , Toxicol. Lett. , 116 , 51 – 
59 . 
215. Sadzuka , Y. , Nakai , S. , Miyagishima , A. , Nozawa , Y. , and Hirota , S. ( 1997 ), Effects of 
administered route on tissue distribution and antitumour activity of polyethyleneglycol - 
coated liposomes containing adriamycin , Cancer Lett. , 111 , 77 – 86 . 
216. Marchettini , P. , Stuart , A. , Mohamed , F. , Yoo , D. , and Sugarbaker , P. H. ( 2002 ), Docetaxel: 
Pharmacokinetics and tissue levels after intraperitoneal and intravenous administration 
in a rat model , Cancer Chemother. Pharmacol. , 49 , 499 – 503 . 
217. Sadzuka , Y. , Hirama , R. , and Sonobe , T. ( 2002 ), Effects of intraperitoneal administration 
of liposomes and methods of preparing liposomes for local therapy , Toxicol. Lett. , 126 , 
83 – 90 . 
218. Di Stefano , A. , Carafa , M. , Sozio , P. , Pinnen , F. , Braghiroli , D. , Orlando , G. , Cannazza , 
G. , Ricciutelli , M. , Marianecci , C. , and Santucci , E. ( 2004 ), Evaluation of rat striatal 
l - dopa and DA concentration after intraperitoneal administration of l - dopa prodrugs 
in liposomal formulations , J. Controlled Release , 99 , 293 – 300 . 
219. Oussoren , C. , and Storm , G. ( 2001 ), Liposomes to target the lymphatics by subcutaneous 
administration , Adv. Drug Deliv. Rev. , 50 , 143 – 156 . 

REFERENCES 519 
220. Gregoriadis , G. , Bacon , A. , Caparros - Wanderley , W. , and McCormack , B. ( 2002 ), A role 
for liposomes in genetic vaccination , Vaccine 20 , B1 – B9 . 
221. Wu , M. S. , Robbins , J. C. , Bugianesi , R. L. , Ponpipom , M. M. , and Shen , T. Y. ( 1981 ), 
Modifi ed in vivo behaviour of liposomes containing synthetic glycolipids , Biochim. 
Biophys. Acta , 674 , 19 – 29 . 
222. Fujimoto , Y. , Okuhata , Y. , Tyngi , S. , Namba , Y. , and Oku , N. ( 2000 ), Magnetic resonance 
lymphography of profunded lymphnodes with liposomal gadolinium - diethylenetriamine 
penta - acetic acid , Biol. Pharm. Bull. , 23 , 97 – 100 . 
223. Misselwitz , B. , and Sachse , A. ( 1997 ), Interstitial MR lymphographyusing GD - carrying 
liposomes , Acta Radiol. Suppl. , 412 , 51 – 55 . 
224. Moll , K. P. , Stober , R. , Hermann , W. , Borchert , H. H. , and Utsumi , H. ( 2004 ), In vivo 
ESR studies on subcutaneously injected multilamellar liposomes in living mice , Pharm. 
Res. , 21 , 2017 – 2024 . 
225. Bacon , A. , Caparr o s - Wanderley , W. , Zadi , B. , and Gregoriadis , G. ( 2002 ), Induction of a 
cytotoxic T lymphocyte (CTL) response to plasmid DNA delivered by Lipodine ™ , 
J. Liposome Res. , 12 , 173 – 183 . 
226. Perrie , Y. , Barralet , J. E. , McNeil , S. , and Vangala , A. ( 2004 ), Surfactant vesicle - 
mediated delivery of DNA vaccines via the subcutaneous route , Int. J. Pharm. , 284 , 
31 – 41 . 
227. Klibanov , A. L. , Maruyama , K. , Torchilin , V. P. , and Huang , L. ( 1990 ), Amphipathic polyethyleneglycols 
effectively prolong the circulation time of liposomes , FEBS Lett. , 268 , 
235 – 237 . 
228. Gabizon , A. , and Papahadjopoulos , D. ( 1992 ), The role of surface charge and hydrophilic 
groups on liposome clearance in vivo , Biochim. Biophys. Acta , 1103 , 94 – 100 . 
229. Ishida , O. , Maruyama , K. , Sasaki , K. , and Iwatsuru , M. ( 1999 ), Size - dependent extravasation 
and interstitial localization of polyethyleneglycol liposomes in solid tumour - bearing 
mice , Int. J. Pharm. , 190 , 49 – 56 . 
230. Levchenko , T. S. , Rammohan , R. , Lukyanov , A. N. , Whiteman , K. R. , and Torchilin , V. P. 
( 2002 ), Liposome clearance in mice: The effect of a separate and combined presence of 
surface charge and polymer coating , Int. J. Pharm. , 240 , 95 – 102 . 
231. Lee , C. M. , Choi , Y. , Huh , E. J. , Lee , K. Y. , Song , H. C. , Sun , M. J. , Jeong , H. J. , Cho , C. S. , 
and Bom , H. S. ( 2005 ), Polyethylene glycol (PEG) modifi ed 99m Tc - HMPAO - liposome for 
improving blood circulation and biodistribution: The effect of the extent of PEG ylation , 
Cancer Biother. Radiopharm. , 20 , 620 – 628 . 
232. Bradley , A. J. , Devine , D. V. , and Ansell , S. M. ( 1998 ), Inhibition of liposome - induced 
complement activation by incorporated poly(ethylene glycol) - lipids , Arch. Biochem. 
Biophys. , 357 , 185 – 192 . 
233. Lu, W. L. , Qi, X. R. , Zhang , Q. , Li, R. Y. , Zhang , R. J. , and Wei, S. L. (2004 ), A PEGylated 
liposomal platform: pharmacokinetics, pharmacodynamics and toxicity in mice using 
doxorubicin as a model drug , J. Pharmacol. Sci. , 95 , 381 – 389 . 
234. Ortho Biotech Products, L. P. , http://www.doxil.com/common/prescribing_information/ 
DOXIL/PDF/DOXIL_PI_Booklet.pdf#search= ‘ DOXIL ’ , 2003 – 2007 . 
235. Blume , G. , Cevc , G. , Crommelin , M. D. , Bakker - Woudenberg , I. A. , Kluft , C. , and Storm , 
G. (1993), Specifi c targeting with poly(ethylene glycol) - modifi ed liposomes: Coupling of 
homing devices to the ends of the polymeric chains combines effective target binding 
with long circulation times , Biochim. Biophys. Acta , 1149 , 180 – 184 . 
236. Gabizon , A. , Horowitz , A. T. , Goren , D. , Tzemach , D. , Shmeeda , H. , and Zalipsky , S. 
( 2003 ), In vivo fate of folate - targeted polyethylene - glycol liposomes in tumor - bearing 
mice , Clin. Cancer Res. , 9 , 6551 – 6559 . 

520 LIPOSOMES AND DRUG DELIVERY 
237. Maeda , N. , Takeuchi , Y. , Takada , M. , Sadzuka , Y. , Namba , Y. , and Oku , N. ( 2004 ), Anti - 
neovascular therapy by use of tumour neovasculature - targeted long - circulating liposome 
, J. Controlled Release , 100 , 41 – 52 . 
238. Syrigos , K. N. , Vile , R. G. , Peters , M. , and Harrington , K. J. ( 2003 ), Biodistribution and 
pharmacokinetics of 111In - DTPA - labelled pegylated liposomes after intraperitoneal 
injection , Acta Oncol. , 42 , 147 – 153 . 
239. Cabanes , A. , Even - Chen , S. , Zimberoff , J. , Barenholz , Y. , Kedar , E. , and Gabizon , A. 
( 1999 ), Enhancement of antitumor activity of polyethylene glycol - coated liposomal 
doxorubicin with soluble and liposomal interleukin 2 , Clini. Cancer Res. , 5 , 687 – 693 . 
240. Oussoren , C. , Eling , W. M. , Crommelin , D. J. , Storm , G. , and Zuidema , J. ( 1998 ), The 
infl uence of the route of administration and liposome composition on the potential of 
liposomes to protect tissue against local toxicity of two antitumour drugs , Biochim. 
Biophys. Acta , 1369 , 159 – 172 . 
241. Patton , J. S. , Fishburn , C. S. , and Weers , J. G. ( 2004 ), The lungs as a portal of entry for 
systemic drug delivery , Proc. Am. Thorac. Soc. , 1 , 338 – 344 . 
242. Niven , R. W. , and Scheier , H. ( 1990 ), Nebulization of liposomes. I. Effects of lipid composition 
, Pharm. Res. , 7 , 1127 – 1133 . 
243. Niven , R. W. , Speer , M. , and Scheier , H. ( 1991 ), Nebulization of liposomes. II. The effects 
of size and modeling of solute release profi les , Pharm. Res. , 8 , 217 – 221 . 
244. Niven , R. W. , Carvajal , M. T. , and Scheier , H. ( 1992 ), Nebulization of liposomes. III. 
Effect of operating conditions , Pharm. Res. , 9 , 515 – 520 . 
245. Taylor , K. M. G. , Taylor , G. , Kellaway , I. W. , and Stevens , J. (1990), The stability of liposomes 
to nebulization , Inter. J. Pharm. , 58 , 57 – 61 . 
246. Schwarz , L. A. , Johnson, J. L. , Black, M. , Cheng , S. H. , Hogan, M. E. , and Waldrep , J. C. 
( 1996 ), Delivery of DNA - cationic liposome complexes by small particle aerosol , Hum. 
Gene Ther. , 24 , 35 – 36 . 
247. Eastman , S. J. , Tousignant , J. D. , Lukason , M. J. , Murray , H. , Siegel , C. S. , Constantino , P. , 
Harris , D. J. , Cheng , S. H. , and Scheule , R. K. ( 1997 ), Optimization of formulations and 
conditions for the aerosol delivery of functional cationic lipid: DNA complexes , Hum. 
Gen. Ther. , 8 , 313 – 322 . 
248. Gonzalez - Rothi , R. J. , and Schreier , H. ( 1993 ), Pulmonary delivery of liposomes , 
J. Controlled Release , 5 , 149 – 161 . 
249. Waldrep , J. C. , Gilbert , B. E. , Black , M. , and Knight , V. ( 1997 ), Operating characteristics 
of 18 different continuous - fl ow jet nebulizers with beclomethasone dipropionate liposome 
aerosol , Chest , 111 , 316 – 323 . 
250. Hochhaus , G. , Gonzalez - Rothi , R. J. , Lukyanov , A. , Derendorf , H. , and Dallas Costa , T. 
( 1995 ), Assessment of glycocorticoid lung targeting by ex vivo receptor binding studies 
in rats , Pharm. Res. , 12 , 134 – 137 . 
251. Gonzalez - Rothi , R. J. , Suarez , S. , Hochhaus , G. , Schreier , H. , Lukyanov , A. , Derendorf , 
H. , and Dallas Costa , T. ( 1996 ), Pulmonary targeting of liposomal triamcinolone acetonide 
, Pharm. Res. , 13 , 1699 – 1703 . 
252. Schreier , H. , McNicol , K. J. , Ausborn , M. , and Soucy , D. W. , Derendorf , H. , Stecenko , 
A. A. , and Gonzalez - Rothi , R. J. ( 1992 ), Pulmonary delivery of amikacin liposomes and 
acute liposomes toxicity in the sheep , Int. J. Pharm. , 87 , 183 – 193 . 
253. Omri , A. , Bealuc , C. , Bouhajib , M. , Montplaisir , S. , Sharkawi , M. , and Lagace , J. ( 1994 ), 
Pulmonary retention of free and liposome - encapsulated tobramycin after intratracheal 
administration in uninfected rats and rats infected with Pseudomonas aeruginosa , 
Antimicr. Agents Chemother. , 38 , 1090 – 1095 . 

REFERENCES 521 
254. Shek , P. N. , Suntres , Z. E. , and Brooks , J. I. ( 1994 ), Liposomes in pulmonary applications: 
Physicochemical considerations, pulmonary distribution and antioxidant delivery , 
J. Drug Targe. , 2 , 431 – 442 . 
255. Suntres , Z. E. , and Shek , P. N. ( 1994 ), Incorporation of . - tocopherol in liposomes promotes 
the retention of liposome - encapsulated glutathione in the rat lung , J. Pharm. 
Pharmacol. , 46 , 23 – 28 . 
256. Suntres , Z. E. , and Shek , P. N. ( 1995 ), Intratracheally administered liposomal alpha - 
tocopherol protects the lung against long - term toxic effects of paraquat , Biomed. 
Environ. Sci. , 8 , 289 – 300 . 
257. Walther , F. J. , David - Cu , R. , and Lopez , S. L. ( 1995 ), Antioxidant - surfactant 
liposomes mitigate hyperoxic lung injury in premature rabbits , Am. J. Physiol. , 269 , 
L613 – L617 . 
258. Vyas , S. P. , Kannan , M. P. , Jain , S. , Mishra , V. , and Singh , P. ( 2004 ), Design of liposomal 
aerosols for improved delivery of rifampicin to alveolar macrophages , Int. J. Pharm. , 269 , 
37 – 49 . 
259. Vyas , S. P. , Quraishi , S. , Gupta , S. , and Jaganathan , K. S. ( 2005 ), Aerosolized 
liposome - based delivery of amphotericin B to alveolar macrophages , Int. J. Pharm. , 296 , 
12 – 25 . 
260. Doddoli , C. , Ghez , O. , Barlesi , F. , D ’ Journo , B. , Robitail , S. , Thomas , P. , and Clerc , T. 
( 2005 ), In vitro and in vivo methotrexate disposition in alveolar macrophages: Comparison 
of pharmacokinetic parameters of two formulations , Int. J. Pharm. , 297 , 180 – 189 . 
261. Fidler , I. J. , Raz , A. , Fogler , W. E. , Kirsh , R. , Bugleski , P. , and Poste , G. ( 1980 ), Design of 
liposomes to improve delivery of macrophage - augmenting agents to alveolar macrophages 
, Cancer Res. , 40 , 4460 – 4466 . 
262. Juliano , R. L. , and McCullough , H. N. ( 1980 ), Controlled delivery of an antitumor drug: 
Localized action of liposome encapsulated cytosine arabinoside administered via the 
respiratory system , Am. Soc. Pharm. Exp. Ther. , 214 , 381 – 387 . 
263. McCullough , H. N. , and Juliano , R. L. ( 1979 ), Organ - selective action of an antitumor 
drug: Pharmacologic studies of liposome - encapsulated beta - cytosine arabinoside administered 
via the respiratory system of the rat , J. Natl. Cancer Inst. , 63 , 727 – 731 . 
264. Knight , V. , Kleinerman , E. S. , Waldrep , J. C. , Giovanella , B. C. , Gilbert , B. E. , and 
Koshkina , N. V. ( 2000 ), 9 - Nitrocamptothecin liposome aerosol treatment of human 
cancer subcutaneous xenografts and pulmonary cancer metastases in mice , Ann. N.Y. 
Acad. Sci. , 922 , 151 – 163 . 
265. Canonico , A. E. , Conary , J. T , Meyrick , B. O. , and Brigham , K. L. ( 1994 ), Aerosol and 
intravenous transfection of human a1 - antitrypsin gene to lungs of rabbits , Am. J. Respir. 
Cell Mol. Biol. , 10 , 24 – 29 . 
266. Stribling , R. , Brunette , E. , Liggitt , D. , Gaensler , K. , and Debs , R. ( 1992 ), Aerosol gene 
delivery in vivo , Proc. Natl. Acad. Sci. USA , 89 , 11277 – 11281 . 
267. Rowland , R. N. , and Woodley , J. F. ( 1980 ), The stability of liposomes in vitro to pH, bile 
salts and pancreatic lipase , Biochim. Biophys. Acta , 620 , 400 – 409 . 
268. Nagata , M. , Yotsuyanagi , T. , and Ikeda , M. ( 1988 ), A two step model of disintegration 
kinetics of liposomes in bile salts , Chem. Pharm. Bull. , 36 , 1508 – 1513 . 
269. Quadachi , S. , Paternostre , M. , andre , C. , Genin , I. , Thao , T. X. , Puisieux , F. , Devissaguet , 
J. , and Barratt , G. ( 1998 ), Liposomal formulations for oral immunotherapy: In - vitro stability 
in synthetic intestinal media and in - vivo effi cacy in the mouse , J. Drug Targe. , 5 , 
365 – 378 . 
270. Freund , O. ( 2001 ), Biodistribution and gastrointestinal drug delivery of new lipidic 
multilamellar vesicles , Drug Deliv. , 8 , 239 – 244 . 

522 LIPOSOMES AND DRUG DELIVERY 
271. Mezei , M. , and Gulasekharam , V. ( 1981 ), Liposomes: A selective drug delivery system 
for topical route of administration - gel dosage form , J. Pharm. Pharmacol. , 34 , 473 – 474 . 
272. Mezei , M. , and Gulasekharam , V. ( 1980 ), Liposomes: A selective drug delivery system 
for topical route of administration. I - lotion dosage form , Life Sci. , 26 , 1473 – 14777 . 
273. Honeywell-Nguyen, P. L. , and Bouwstra, J. A. (2005), Vesicles as a tool for transdermal 
and dermal delivery , Drug Discovery Today: Technol. , 2 , 67 – 74 . 
274. Knepp , V. M. , Hinz , R. S. , Szoka , F. C. , and Guy , R. H. ( 1988 ), Controlled drug release 
from a novel liposomal delivery system. I. Investigation of transdermal potential , 
J. Controlled Release , 5 , 211 – 221 . 
275. Yu , H. Y. , and Liao , H. M. ( 1996 ), Triamcinolone permeation from different liposome 
formulations through rat skin in vitro , Int. J. Pharm. , 127 , 1 – 7 . 
276. Fresta , M. , and Puglisi , G. ( 1997 ), Corticosteroid dermal delivery with skin lipid liposomes 
, J. Controlled Release , 44 , 141 – 151 . 
277. Liu , H. , Pan , W. S. , Tang , R. , and Luo , S. D. ( 2004 ), Topical delivery of acyclovir palmitate 
liposome formulations through rat skin in vitro , Pharmazie , 59 , 203 – 206 . 
278. Egbaria , K. , Ramanchandran , C. , and Weiner , N. ( 1991 ), Topical application of liposomally 
entrapped cyclosporin evaluated by in vitro diffusion studies with human skin , 
Skin Pharmacol. , 4 , 21 – 28 . 
279. Manosroi , A. , Konganeramit , L. , and Manosroi , J. ( 2004 ), Stability and transdermal 
absorption of topical amphotericin B liposome formulations , Int. J. Pharm. , 270 , 279 – 
286 . 
280. Aguillela , V. , Kontturi , K. , Murtomaki , L. , and Ramirez , P. ( 1994 ), Estimation of the 
pore size and charge density in human cadaver skin , J. Controlled Release , 32 , 249 – 
257 . 
281. Cevc , G. , Schatzlein , A. , and Richardsen , H. ( 2002 ), Ultradeformable lipid vesicles can 
penetrate the skin and other semi - permeable barriers unfragmented. Evidence from 
double label CLSM experiments and direct size measurements , Biochim. Biophys. Acta , 
564 , 21 – 30 . 
282. Cevc , G. ( 1996 ), Transferosomes, liposomes and other lipid suspensions on the skin: 
Permeation enhancement, vesicle penetration and transdermal drug delivery , Crit. Rev. 
Ther. Drug Carrier Syst. , 13 , 257 – 388 . 
283. de Plessis , J. , Ramachandran , C. , Weiner , N. , and Muller , D. G. ( 1994 ), The infl uence of 
particle size of liposomes on the deposition of the drug into skin , Int. J. Pharm. , 103 , 
277 – 282 . 
284. Verma , D. D. , Verma , S. , Blume , G. , and Fahr , A. ( 2003 ), Particle size of liposomes infl uences 
dermal delivery of substances into skin , Int. J. Pharm. , 258 , 141 – 151 . 
285. Miyachi , Y. , Imamura , S. , and Niwas , Y. ( 1987 ), Decreased skin superoxide dismutase 
activity by a single exposure of ultraviolet radiation is reduced by liposomal superoxide 
dismutase pretreatment , J. Invest. Dermatal. , 89 , 111 – 112 . 
286. Short , S. M. , Rubas , W. , Paasch , B. D. , Mrsny , R. ( 1995 ), Transport of biologically active 
interferon - gamma across human skin in vitro , Pharm. Res. , 12 , 1140 – 1145 . 
287. Cotsarelis , G. ( 2002 ), The hair follicle as a target for gene therapy , Ann. Dermatal Venereol. 
, 129 , 841 – 844 . 
288. Raghavachari , N. , and Fahl , W. E. ( 2002 ), Targeted gene delivery to skin cells in vivo: A 
comparative study of liposomes and polymers as delivery vehicles , J. Pharm. Sci. , 91 , 
615 – 622 . 
289. Balsari , A. L. , Morelli , D. , Menard , S. , Veronesi , U. , and Colnaghi , M. I. ( 1994 ), Protection 
against doxorubicin - induced alopecia in rats by liposome - entrapped monoclonal antibodies 
, Res. Commun. , 8 , 226 – 230 . 

REFERENCES 523 
290. Weiner , N. , Williams , N. , Birch , G. , Ramachandran , R. , Shipman , C. , Jr. , and Flynn , G. 
( 1989 ), Topical delivery of liposomally encapsulated interferon evaluated in a cutaneous 
herpes guinea pig model , Antimicrob. Agents Chemoter. , 33 , 1217 – 1221 . 
291. Godin , B. , and Touitou , E. ( 2003 ), Ethosomes: New prospects in transdermal delivery , 
Crit. Rev. Ther. Drug Carrier Syst. , 20 , 63 – 102 . 
292. Kalia , Y. N. , Naik , A. , Garrison , J. , Guy , R. H. ( 2004 ), Iontophoretic drug delivery , Adv. 
Drug Deliv. Rev. , 56 , 619 – 658 . 
293. Vulta , N. B. , Betageri , G. V. , and Banga , A. K. ( 1996 ), Transdermal iontophoretic delivery 
of enkephalin formulated in liposomes , J. Pharm. Sci. , 85 , 5 – 8 . 
294. Fang , J. Y. , Sung , K. C. , Lin , H. H. , and Fang , C. L. ( 1999 ), Transdermal iontophoretic 
delivery of enoxacin from various liposome - encapsulated formulations , J. Controlled 
Release , 60 , 1 – 10 . 
295. Essa , E. A. , Bonner , M. C. , and Barry , B. W. ( 2002 ), Iontophoretic estradiol skin 
delivery and tritium exchange in ultradeformable liposomes , Int. J. Pharm. , 240 , 55 – 
66 . 
296. Essa , E. A. , Bonner , M. C. , and Barry , B. W. ( 2004 ), Electrically assisted skin delivery of 
liposomal estradiol; phospholipid as damage retardant , J. Controlled Release , 95 , 
535 – 546 . 
297. Conjeevaram , R. , Chaturvedala , A. , Betageri , G. V. , Sunkara , G. , and Banga , A. K. ( 2003 ), 
Iontophoretic in vivo transdermal delivery of beta - blockers in hairless rats and reduced 
skin irritation by liposomal formulation , Pharm. Res. , 9 , 1496 – 1501 . 
298. Han , I. , Kim , M. , and Kim , J. ( 2004 ), Enhanced transfollicular delivery of adriamycin 
with a liposome and iontophoresis , Exp. Dermatol. , 13 , 86 – 92 . 
299. Wells , J. M. , Li , L. H. , Sen , A. , Jahreis , G. P. , and Hui , S. W. ( 2000 ), Electroporation - 
enhanced gene delivery in mammary tumors , Gene Ther. , 7 , 541 – 547 . 
300. Essa , E. A. , Bonner , M. C. , and Barry , B. W. ( 2003 ), Electroporation and ultradeformable 
liposomes; human skin barrier repair by phospholipids , J. Controlled Release , 92 , 
163 – 172 . 
301. Sen , A. , Zhao , Y. L. , and Hui , S. W. ( 2002 ), Saturated anionic phospholipids enhance 
transdermal transport by electroporation , Biophys. J. , 83 , 2064 – 2073 . 
302. Vyas , S. P. , Singh , R. , Asati , R. K. ( 1995 ), Liposomally encapsulated diclofenac for sonophoresis 
induced systemic delivery , J. Microencapsul. , 12 , 149 – 154 . 
303. Ahmed , I. , and Patton , T. F. ( 1987 ), Disposition of timolol and inulin in the rabbit eye 
following corneal versus noncorneal absorption , Int. J. Pharm. , 38 , 9 – 21 . 
304. Mishima , S. , Gasset , A. Klyce , S. D. , and Baum , J. L. ( 1966 ), Determination of tear volume 
and tear fl ow , Invest. Ophthalmol. , 5 , 264 – 276 . 
305. Ebrahim , S. , Peyman , G. , and Lee , P. J. ( 2005 ), Applications of liposomes in ophtalmology , 
Sur. Opthalmol. , 50 , 167 – 182 . 
306. Kaur , I. P. , Garg , A. , Singla , A. K. , and Aggarwal , D. ( 2004 ), Vesicular systems in ocular 
drug delivery: An overview , Int. J. Pharm. , 269 , 1 – 14 . 
307. Zimmer , A. K. , and Kreuter , J. ( 1991 ), Studies on the transport pathway of PBCA 
nanoparticles in ocular tissues , J. Microencapsul. , 8 , 497 – 504 . 
308. Marchal - Heussler , Sirbat D. , and Hoffman , M. ( 1993 ), Poly - E - caprolactone nanocapsules 
in carteolol ophthalmic delivery , Pharm. Res. , 10 , 386 – 390 . 
309. Calvo , P. , Thomas , C. , and Alonso , M. J. ( 1994 ), Study of the mechanism of interaction 
of poly - . - caprolactone nanocapsules with the cornea by confocal laser scanning microscopy 
, Int. J. Pharm. , 103 , 283 – 291 . 

524 LIPOSOMES AND DRUG DELIVERY 
310. Schaeffer , H. E. , Brietfelter , J. M. , and Krohn , D. L. ( 1982 ), Lectin - mediated attachment 
of liposomes to cornea: Infl uence on transcorneal drug fl ux , Invest. Ophthalmol. Vis. Sci. , 
23 , 530 – 533 . 
311. Dharma , S. K. , Fishman , P. H. , and Peyman , G. A. ( 1986 ), A preliminary study of corneal 
penetration of 125I - labelled iodoxuridine liposome , Acta Ophthalmol. (Copenh.) , 64 , 
298 – 301 . 
312. Smolin , G. , Okumoto , M. , Feiler , S. , and Condon , D. ( 1981 ), Iodoxuridine - liposome 
therapy for herpes simplex keratitis , Am. J. Ophthalmol. , 91 , 220 – 226 . 
313. Schaeffer , H. E. , and Krohn , D. L. ( 1982 ), Liposomes in topical drug delivery , Invest. 
Ophthalmol. Vis. Sci. , 22 , 220 – 227 . 
314. Stratford , R. E. J. , Yang , D. C. , Redell , M. A. , and Lee , V. H. L. ( 1983 ), Effects of topically 
applied liposomes and disposition of epinephrine and inulin in albino rabbit eye , Int. J. 
Pharm. , 13 , 263 – 272 . 
315. Stratford , R. E. J. , Yang , D. C. , Redell , M. A. , and Lee , V. H. L. ( 1983 ), Ocular distribution 
of liposome encapsulated epinephrine and inulin in the albino rabbit , Curr. Eye Res. , 2 , 
377 – 386 . 
316. Lee , V. H. L. , Takemoto , K. A. , and Iimoto , D. S. ( 1984 ), Precorneal factors infl uencing 
the ocular distribution of topically applied liposomal inulin , Curr. Eye Res. , 3 , 585 – 
591 . 
317. Angelucci , D. ( 2001 , Nov.), New solutions for dry eye, EyeWorld, available: http://eyeworld.
org.pastissue.php . 
318. Lin , H. H. , Ko , S. M. , Hsu , L. R. , and Tsai , Y. H. ( 1996 ), The preparation of norfl oxacin - 
loaded liposomes and their in - vitro evaluation in pig ’ s eye , J. Pharm. Pharmacol. , 48 , 
801 – 805 . 
319. Frucht - Perry , J. , Assil , K. K. , Ziegler , E. , et al . ( 1992 ), Fibrin - enmeshed tobramycin 
liposomes: Single application topical therapy of Pseudomonas keratitis , Cornea , 11 , 
393 – 397 . 
320. Assil , K. K. , Frucht - Perry , J. , Ziegler , E. , et al . ( 1991 ), Tobramycin liposomes. Single 
subconjunctival therapy of pseudomonal keratitis , Invest. Ophthalmol. Vis. Sci. , 32 , 
3216 – 3220 . 
321. Barza , M. , Doft , B. , and Lynch , E. ( 1993 ), Ocular penetration of ceftriaxone, ceftazidime, 
and vancomycin after subconjunctival injection in humans , Arch. Ophthalmol. , 111 , 
492 – 494 . 
322. Pleyer , U. , Grammar , J. , Pleyer , J. H. , et al . ( 1995 ), Amphotericin B — bioavailability in 
the cornea. Studies with local administration of liposome incorporated amphotericin B , 
Ophthalmologe , 92 , 469 – 475 . 
323. Law , S. L. , Huang , K. J. , and Chiang , C. H. ( 2000 ), Acyclovir - containing liposomes for 
potential ocular delivery. Corneal penetration and absorption , J. Controlled Release , 
63 ( 1 – 2 ), 135 – 140 . 
324. Fresta , M. , Panico , A. M. , Bucolo , C. , et al . ( 1999 ), Characterization and in - vivo ocular 
absorption of liposome - encapsulated acyclovir , J. Pharm. Pharmacol. , 51 , 565 – 576 . 
325. Al - Muhammed , J. , Ozer , A. Y. , Ercan , M. T. , and Hincal , A. A. ( 1996 ), In - vivo studies on 
dexamethasone sodium phosphate liposomes , J. Microencapsul. , 13 , 293 – 306 . 
326. Pleyer , U. , Elkins , B. , Ruckert , D. , et al . ( 1994 ), Ocular absorption of cyclosporine A from 
liposomes incorporated into collagen shields , Curr. Eye Res. , 13 , 177 – 181 . 
327. Pleyer , U. , Lutz , S. , Jusko , W. J. , et al . ( 1993 ), Ocular absorption of topically applied 
FK506 from liposomal and oil formulations in the rabbit eye , Invest. Ophthalmol. Vis. 
Sci. , 34 , 2737 – 2742 [erratum appears Invest. Ophthalmol. Vis. Sci ., 34, 3481, 1993]. 

REFERENCES 525 
328. Torres , P. F. , Slegers , T. P. , Peek , R. , et al . ( 1999 ), Changes in cytokine mRNA levels in 
experimental corneal allografts after local clodronate - liposome treatment , Invest. Ophthalmol. 
Vis. Sci. , 40 , 3194 – 3201 . 
329. Slegers , T. P. , van Rooijen , N. , van Rij , G. , and van der Gaag , R. ( 2000 ), Delayed graft 
rejection in pre - vascularised corneas after subconjunctival injection of clodronate liposomes 
, Curr. Eye Res. , 20 , 322 – 324 . 
330. Van der Veen , G. , Broersma , L. , Dijkstra , C. D. , et al . ( 1994 ), Prevention of corneal 
allograft rejection in rats treated with subconjunctival injections of liposomes containing 
dichloromethylene diphosphonate , Invest. Ophthalmol. Vis. Sci. , 35 , 3505 – 3515 . 
331. Van der Veen , G. , Broersma , L. , Van Rooijen , N. , et al . ( 1998 ), Cytotoxic T lymphocytes 
and antibodies after orthotropic penetrating keratoplasty in rats treated with 
dichloromethylene diphosphonate encapsulated liposomes , Curr. Eye Res. , 17 , 1018 – 
1026 . 
332. Lee , V. H. , and Carson , L. W. ( 1986 ), Ocular disposition of inulin from single and multiple 
doses of positively charged multilamellar liposomes: Evidence for alterations in tear 
dynamics and ocular surface characteristics , J. Ocul. Pharmacol. , 2 , 353 – 364 . 
333. Elorza, B. , Elorza, M. A. , Sainz, M. C. , and Chantres , J. R. (1993), Comparison of particle 
size and encapsulation parameters of three liposomal preparations , J. Microencapsul. , 
10 , 237 – 248 . 
334. Felt , O. , Furrer , P. , Mayer , J. M. , Plazonnet , B. , Buri , P. , and Gurny , R. ( 1999 ), Topical use 
of chitosan in ophthalmology: Tolerance assessment and evaluation of precorneal retention 
, Int. J. Pharm. , 180 , 185 – 193 . 
335. Felt , O. , Baeyens , V. , Zignani , M. , Buri , P. , and Gurny , R. , ( 1999 ), Mucosal drug delivery , 
in Mathiowitz , E. , Ed., The Encyclopedia of Controlled Drug Delivery , Wiley , New York , 
pp. 605 – 626 . 
336. McCalden , T. A. , and Levy , M. ( 1990 ), Retention of topical liposomal formulations on 
the cornea , Experientia , 46 , 713 – 715 . 
337. Grass , G. M. , and Robinson , J. R. ( 1988 ), Mechanisms of corneal drug penetration. I: 
In vivo and in vitro kinetics , J. Pharm. Sci. , 77 , 3 – 14 . 
338. Burrow , J. , Tsibouklis , J. , and Smart , J. D. , ( 2002 ), Drug delivery to the eye , The Drug 
Delivery Company Report , p. 4 . 
339. Taniguchi , K. , Yamamoto , Y. , Itakura , K. , Miichi , H. , and Hayashi , S. ( 1988 ), Assessment 
of ocular irritability of liposome preparations , J. Pharmacobiodyn. , 11 , 607 – 611 . 
340. Barber , R. F. , and Shek , P. N. ( 1990 ), Tear induced release of liposome entrapped agents , 
Int. J. Pharm. , 60 , 219 – 227 . 
341. Kaufman , H. E. , Steinemann , T. L. , Lehman , E. , Thompson , H. W. , Varnell , E. D. , Jacob - 
LaBarre , J. T. , and Gebhardt , B. M. ( 1994 ), Collagen - based drug delivery and artifi cial 
tears , J. Ocul. Pharmacol. , 10 , 17 – 27 . 
342. Yerushalmi , N. , and Margalit , R. ( 1994 ), Bioadhesive, collagen - modifi ed liposomes: 
Molecular and cellular level studies on the kinetics of drug release and on binding to 
cell monolayers , Biochim. Biophys. Acta , 1189 , 13 – 20 . 
343. Bochot , A. , Fattal , E. , Gulek , A. , and Aonarraze , G. ( 1998 ), Liposome dispersed within 
thermosensitive gel. A new dosage form for ocular delivery of oligonucleotide , Pharm. 
Res. , 15 , 1364 – 1369 . 
344. Bochot , A. , Couvreur , P. , and Fattal , E. ( 2000 ), Intravitreal administration of 
antisense oligonucleotides: Potential of liposomal delivery , Prog. Retin. Eye Res. , 19 , 
131 – 147 . 
345. Bochot , A. , Fattal , E. , Boutet , V. , Deverre , J. R. , Jeamy , J. C. , Chacun , H. , and Couvreur , 
P. ( 2002 ), Intravitreal delivery of oligonucleotides by sterically stabilized liposomes , 
Invest. Opthalmol. Vis. Sci. , 43 , 253 – 259 . 

526 LIPOSOMES AND DRUG DELIVERY 
346. Schmidt - Erfurth , U. , Flotte , T. J. , Gragoudas , E. S. , et al . ( 1996 ), Benzoporphyrin - 
lipoprotein - mediated photodestruction of intraocular tumors , Exp. Eye Res. , 62 , 1 – 10 . 
347. Kawakami , S. , Yamamura , K. , Mukai , T. , Nishida , K. , Nakamura , J. , Sakaeda , T. , 
Nakashima , M. , and Sasaki , H. ( 2001 ), Sustained ocular delivery of tilisolol to rabbits 
after topical administration or intravitreal injection of lipophilic prodrug incorporated 
in liposomes , J. Pharm. Pharmacol. , 53 , 157 – 161 . 
348. Harris , A. S. , Svensson , E. , Wagner , Z. G. , Lethagen , S. , and Nilsson , I. M. ( 1988 ), Effect 
of viscosity on particle - size, deposition, and clearance of nasal delivery systems containing 
desmopressin , J. Pharm. Sci. , 77 , 405 – 408 . 
349. Vyas , S. P. , Goswami , S. K. , and Singh , R. ( 1995 ), Liposomes based nasal delivery system 
of nifedipine — development ad characterization , Int. J. Pharm. , 118 , 23 – 30 . 
350. Shahiwala A , and Misra , A. ( 2004 ), Nasal delivery of levonorgestrel for contraception: 
An experimental study in rats , Fertil. Steril. 81 ( Suppl. 1 ), 893 – 898 . 
351. Kuper , H. , Adami , H. O. , and Trichopoulos , D. ( 2000 ), Infection as a major preventable 
cause of human cancer , J. Intern. Med. , 248 , 171 – 183 . 
352. Alpar , H. O. , Somavarapu , S. , Atuah , K. N. , and Bramwell , V. W. ( 2005 ), Biodegradable 
mucoadhesive particulates for nasal and pulmonary antigen and DNA delivery , Adv. 
Drug Deliv. Rev. , 57 , 411 – 430 . 
353. De Haan, A. , Renegar , K. B. , Small, P. A. , Wilschut, J. , (1995), Vaccine , 13 , 613 – 616 . 
354. Wang , D. , Christopher , M. E. , Nagata , L. P. , Zabielski , M. A. , Li , H. , Wong , J. P. , and 
Samuel , J. ( 2004 ), Intranasal immunization with liposome - encapsulated plasmid DNA 
encoding infl uenza virus hemagglutinin elicits mucosal, cellular and humoral immune 
responses , J. Clin. Virol. , 31 ( Suppl. 1 ), 99 – 106 . 
355. Tafaghodi , M. , Jaafari , M. - R. , and Abolghasem , S. , and Tabassi , S. ( 2006 ), Nasal immunization 
studies using liposomes loaded with tetanus toxoid and CpG - ODN , Eur. J. 
Pharm. Biopharm. , 64 , 138 – 145 . 
356. de Jonge , M. I. , Hamstra , H. J. , Jiskoot , W. , Roholl , P. , Williams , N. A. , Dankert , J. , 
van Alphen , L. , and Van der Ley , P. ( 2004 ), Intranasal immunisation of mice with liposomes 
containing recombinant meningococcal OpaB and OpaJ proteins , Vaccine , 22 , 
4021 – 4028 . 
357. Sloat , B. R. , and Zhengrong , C. ( 2006 ), Strong mucosal and systemic immunities induced 
by nasal immunization with anthrax protective antigen protein incorporated in liposome 
- protamine - DNA particles , Pharm. Res. , 23 , 262 – 269 . 
358. Paveli , Z. , S kalko - Basnet , N. , and Schubert , R. ( 2001 ), Liposomal gels for vaginal drug 
delivery , Int. J. Pharm. , 219 , 139 – 149 . 
359. Paveli , Z. , S kalko - Basnet , N. , and Jal s enjak , I. ( 2005 ), Characterisation and in vitro 
evaluation of bioadhesive liposome gels for local therapy of vaginitis , Int. J. Pharm. , 301 , 
140 – 148 . 
360. Paveli , Z. , S kalko - Basnet , N. , Filipovi - Gr i , J. , Martinac , A. , and Jal s enjak , I. ( 2005 ), 
Development and in vitro evaluation of a liposomal vaginal delivery system for acyclovir 
, J. Controlled Release , 106 , 34 – 43 . 
361. Zhang , Y. , Cristofaro , P. , Silbermann , R. , Pusch , O. , Boden , D. , Konkin , T. , Hovanesian , 
V. , Monfi ls , P. R. , Resnick , M. , Moss S. F. , and Ramratnam , B. ( 2006 ), Engineering 
mucosal RNA interference in vivo , Mol. Ther. , 14 , 336 – 342 . 
362. Hartikka , J. , Bozoukova , V. , Ferrari , M. , Sukhu , L. , Enas , J. , Sawdey , M. , et al . ( 2001 ), 
Vaxfectin enhances the humoral immune response to plasmid DNA - encoded antigens , 
Vaccine , 19 , 1911 – 1923 . 
363. Lochera , C. P. , Witta , S. A. , Ashlocka , B. M. , Polacinob , P. , Hub , S. L. , Shiboskic , S. , 
Schmidtb , A. M. , Agyb , M. B. , anderson , D. M. , Stapransd , S. I. , Megedee , J. , and Levy , J. 

REFERENCES 527 
A. ( 2004 ), Human immunodefi ciency virus type 2 DNA vaccine provides partial protection 
from acute baboon infection , Vaccine , 22 , 2261 – 2272 . 
364. Davidson , R. N. , Croft , S. L. , Scott , A. , Maini , M. , Moody , A. H. , and Bryceson , A. D. 
( 1991 ), Liposomal amphotericin B in drug - resistant visceral leishmaniasis , Lancet , 337 , 
1061 – 1062 . 
365. Guaglianone , P. , Chan , K. , DelaFlor - Weiss , E. , Hanisch , R. , Jeffers , S. , Sharma , D. , et al . 
( 1994 ), Phase I and pharmacologic study of liposomal daunorubicin (DaunoXome) , 
Invest. New Drugs , 12 , 103 – 110 . 
366. Gabizon , A. , Peretz , T. , Sulkes , A. , Amselem , S. , Ben - Yosef , R. , Ben - Baruch , N. , et al . 
( 1989 ), Systemic administration of doxorubicin - containing liposomes in cancer patients: 
A phase I study , Eur. J. Cancer Clin. Oncol. , 25 , 1795 – 1803 . 
367. Donald , P. R. , Sirgel , F. A. , Venter , A. , Smit , E. , Parkin , D. P. , VandeWal , B. W. , et al . 
( 2001 ), The early bactericidal activity of a low - clearance liposomal amikacin in pulmonary 
tuberculosis , J. Antimicrob. Chemother. , 48 , 877 – 880 . 
368. Kadry , A. A. , Al - Suwayeh , S. A. , Abd - Allah , A. R. , and Bayomi , M. A. ( 2004 ), Treatment 
of experimental osteomyelitis by liposomal antibiotics , J. Antimicrob. Chemother. , 54 , 
1103 – 1108 . 
369. Nishiyama , N. , and Kataoka , K. ( 2006 ), Current state, achievements, and future prospects 
of polymeric micelles as nanocarriers for drug and gene delivery, Pharmacol. Therap. , 
112 , 630 – 648 . 
370. Zamboni , W. C. , Gervais , A. C. , Egorin , M. J. , Schellens , J. H. , Zuhowski , E. G. , Pluim , 
D. , et al . ( 2004 ), Systemic and tumor disposition of platinum after administration 
of cisplatin or STEALTH liposomal - cisplatin formulations (SPI - 077 and SPI - 077 B103) 
in a preclinical tumor model of melanoma , Cancer Chemother. Pharmacol. , 53 , 329 – 
336 . 
371. Proulx , M. E. , Desormeaux , A. , Marquis , J. F. , Olivier , M. , and Bergeron , M. G. ( 2001 ), 
Treatment of visceral leishmaniasis with sterically stabilized liposomes containing camptothecin 
, Antimicrob. Agents Chemother. , 45 , 2623 – 2627 . 
372. Giles , F. J. , Tallman , M. S. , Garcia - Manero , G. , Cortes , J. E. , Thomas , D. A. , Wierda , W. G. , 
et al . ( 2004 ), Phase I and pharmacokinetic study of a low - clearance, unilamellar liposomal 
formulation of lurtotecan, a topoisomerase 1 inhibitor, in patients with advanced 
leukemia , Cancer , 100 , 1449 – 1458 . 
373. Verschraegen , C. F. , Gilbert , B. E. , Loyer , E. , Huaringa , A. , Walsh , G. , Newman , R. A. , 
et al . ( 2004 ), Clinical evaluation of the delivery and safety of aerosolized liposomal 9 - 
nitro - 20(s) - camptothecin in patients with advanced pulmonary malignancies , Clin. 
Cancer Res. , 10 , 2319 – 2326 . 
374. Rudin , C. M. , Marshall , J. L. , Huang , C. H. , Kindler , H. L. , Zhang , C. , Kumar , D. , et al . 
( 2004 ), Delivery of a liposomal c - raf - 1 antisense oligonucleotide by weekly bolus dosing 
in patients with advanced solid tumors: A phase I study , Clin. Cancer Res. , 10 , 
7244 – 7251 . 
375. Chien , P. Y. , Wang , J. , Carbonaro , D. , Lei , S. , Miller , B. , Sheikh , S. , et al . ( 2004 ), Novel 
cationic cardiolipin analogue - based liposome for effi cient DNA and small interfering 
RNA delivery in vitro and in vivo , Cancer Gene Ther. , 12 , 321 – 328 . 
376. Asano , T. , and Kleinerman , E. S. ( 1993 ), Liposome - encapsulated MTP - PE: A novel biologic 
agent for cancer therapy , J. Immunother. , 14 , 286 – 292 . 
377. Dagar , S. , Krishnadas , A. , Rubinstein , I. , Blend , M. J. , and Onyuksel , H. ( 2003 ), VIP 
grafted sterically stabilized liposomes for targeted imaging of breast cancer: In vivo 
studies , J. Controlled Release , 91 , 123 – 133 . 

528 LIPOSOMES AND DRUG DELIVERY 
378. Park , J. W. , Hong , K. , Kirpotin , D. B. , Colbern , G. , Shalaby , R. , Baselga , J. , et al . ( 2002 ), 
Anti - HER2 immunoliposomes: Enhanced effi cacy attributable to targeted delivery , 
Clin. Cancer Res. , 8 , 1172 – 1181 . 
379. Pakunlu , R. I. , Wang , Y. , Tsao , W. , Pozharov , V. , Cook , T. J. , and Minko , T. ( 2004 ), Enhancement 
of the effi cacy of chemotherapy for lung cancer by simultaneous suppression of 
multidrug resistance and antiapoptotic cellular defense: Novel multicomponent delivery 
system , Cancer Res. , 64 , 6214 – 6224 . 
380. Pardridge , W. M. ( 1996 ), Vector - mediated drug delivery to the brain , Adv. Drug Deliv. , 
36 , 299 – 321 . 
381. Huwyler , J. , Wu , D. , and Pardridge , W. M. ( 1996 ), Brain drug delivery of small molecules 
using immunoliposomes , Proc. Natl. Acad. Sci. USA , 93 , 14164 – 14169 . 
382. Huwyler , J. , Yang , J. , and Pardridge , W. M. ( 1997 ), Targeted delivery of daunomycin using 
immunoliposomes: Pharmacokinetics and tissue distribution in the rat , J. Pharmacol. 
Exp. Ther. , 282 , 1541 – 1546 . 
383. Schnyder , A. , Krahenbuhl , S. , Drewe , J. , and Huwyler , J. ( 2005 ), Targeting of daunomycin 
using biotinylated immunoliposomes: Pharmacokinetics, tissue distribution and in vitro 
pharmacological effects , J. Drug Targeting. , 13 , 325 – 335 . 
384. Xie , Y. , Ye , L. , Zhang , X. , Cui , W. , Lou , J. , Nagai , T. , and Hou , X. ( 2005 ), Transport of 
nerve growth factor encapsulated into liposomes across the blood - brain barrier: In vitro 
and in vivo studies , J. Controlled Release , 105 , 106 – 119 . 
385. Brignole , C. , Marimpietri , D. , Pagnan , G. , Di Paolo , D. , Zancolli , M. , Pistoia , V. , Ponzoni , 
M. , and Pastorino , F. ( 2005 ), Neuroblastoma targeting by c - myb - selective antisense oligonucleotides 
entrapped in anti - GD2 immunoliposome: Immune cell - mediated anti - 
tumor activities , Cancer Lett. , 228 , 181 – 186 . 
386. Siegal , T. , Horowitz , A. , and Gabizon , A. ( 1995 ), Doxorubicin encapsulated in sterically 
stabilized liposomes for the treatment of a brain tumor model: Biodistribution and 
therapeutic effi cacy , J. Neurosurg. , 83 , 1029 – 1037 . 
387. Sharma , U. S. , Sharma , A. , Chau , R. I. , and Straubinger , R. M. ( 1997 ), Liposome - mediated 
therapy of intracranial brain tumors in a rat model , Pharm. Res. , 14 , 992 – 998 . 
388. Aoki , H. , Kakinuma , K. , Morita , K. , Kato , M. , Uzuka , T. , Igor , G. , Takahashi , H. , and 
Tanaka , R. ( 2004 ), Therapeutic effi cacy of targeting chemotherapy using local hyperthermia 
and thermosensitive liposome: Evaluation of drug distribution in a rat glioma 
model , Int. J. Hypertherm. , 20 , 595 – 605 . 
389. Koukourakis , M. I. , Koukouraki , S. , Fezoulidis , I. , Kelekis , N. , Kyrias , G. , Archimandritis , 
S. , and Karkavitsas , N. ( 2000 ), High intratumoural accumulation of stealth liposomal 
doxorubicin (Caelyx) in glioblastomas and in metastatic brain tumors , Br. J. Cancer , 83 , 
1281 – 1286 . 
390. Arnold , R. D. , Mager , D. E. , Slack , J. E. , and Straubinger , R. M. ( 2005 ), Effect of repetitive 
administration of Doxorubicin - containing liposomes on plasma pharmacokinetics 
and drug biodistribution in a rat brain tumor model , Clin. Cancer Res. , 11 , 8856 – 8865 . 
391. Marina , N. M. , Cochrane , D. , Harney , E. , Zomorodi , K. , Blaney , S. , Winick , N. , Bernstein , 
M. , and Link , M. P. ( 2002 ), Dose escalation and pharmacokinetics of pegylated liposomal 
doxorubicin (Doxil) in children with solid tumors: A pediatric oncology group study , 
Clin. Cancer Res. , 8 , 413 – 418 . 
392. Caraglia , M. , Addeo , R. , Costanzo , R. , Montella , L. , Faiola , V. , Marra , M. , Abruzzesse , 
A. , Palmieri , G. , Budillon , A. , Grillone , F. , Venuta , S. , Tagliaferri , P. , and Del Prete , S. 
( 2006 ), Phase II study of temozolomide plus pegylated liposomal doxorubicin in the 
treatment of brain metastases from solid tumors , Cancer Chemother. Pharmacol. , 57 , 
34 – 39 . 

REFERENCES 529 
393. Fiorillo , A. , Maggi , G. , Greco , N. , Migliorati , R. , D ’ Amico , A. , Del Basso De Caro , M. , 
Sabbatino , M. S. , and Buffardi , F. ( 2004 ), Second - line chemotherapy with the association 
of liposomal daunorubicin, carboplatin and etoposide in children with recurrent malignant 
brain tumors , J. Neuro - Oncol. , 66 , 179 – 185 . 
394. Barth , R. F. , Coderre , J. A. , Graca , M. , Vicente , H. , and Blue , T. E. ( 2005 ), Boron neutron 
capture therapy of cancer: Current status and future prospects , Clin. Cancer Res. , 11 , 
3987 – 4002 . 
395. Hawthorne , M. F. , Feakes , D. A. , and Shelly , K. ( 1996 ), Recent results with liposomes as 
boron delivery vehicles from boron neutron capture therapy , in Mishima , Y. , Ed., Cancer 
Neutron Capture Therapy , Plenum , New York , pp 27 – 36 . 
396. Feakes , D. A. , Waller , R. C. , Hathaway , D. K. , and Morton , V. S. ( 1999 ), Synthesis and in 
vivo murine evaluation of Na4[1 - (1 . - B10H9) - 6 - SHB10H8] as a potential agent for boron 
neutron capture therapy , Proc. Natl. Acad. Sci. USA , 96 , 6406 – 6410 . 
397. Carlsson , J. , Kullberg , E. B. , Capala , J. , Sjoberg , S. , Edwards , K. , and Gedda , L. ( 2003 ), 
Ligand liposomes and boron neutron capture theory , J. Neuro - Oncol. , 62 , 47 – 59 . 
398. Barth , R. F. , Yang , W. , Adams , D. M. , Rotaru , J. H. , Shukla , S. , Sekido , M. , Tjarks , W. , 
Fenstermaker , R. A. , Ciesielski , M. , Nawrocky , M. M. , and Coderre , J. A. ( 2002 ), Molecular 
targeting of the epidermal growth factor receptor for neutron capture therapy of 
gliomas , Cancer Res. , 62 , 3159 – 3166 . 
399. Wikstrand , C. J. , Cokgor , I. , Sampson , J. H. , and Bigner , D. D. ( 1999 ), Monoclonal antibody 
therapy of human gliomas: Current status and future approaches , Cancer Metastasis 
Rev. , 18 , 451 – 464 . 
400. Hermanson , M. , Funa , K. , Koopmann , J. , Maintz , D. , Waha , A. , Westermark , B. , Heldin , 
C. H. , Wiestler , O. D. , Louis , D. N. , von Deimling , A. , and Nister , M. ( 1996 ), Association 
of loss of heterozygosity on chromosome 17p with high platelet derived growth factor 
alpha receptor expression in human malignant gliomas , Cancer Res. , 56 , 164 – 171 . 
401. Akabani , G. , Cokgor , I. , Coleman , R. E. , Gonzalez Trotter , D. , Wong , T. Z. , Friedman , 
H. S. , Friedman , A. H. , Garcia - Turner , A. , Herndon , J. E. , DeLong , D. , McLendon , R. E. , 
Zhao , X. G. , Pegram , C. N. , Provenzale , J. M. , Bigner , D. D. , and Zalutsky , M. R. ( 2000 ), 
Dosimetry and dose – response relationships in newly diagnosed patients with malignant 
gliomas treated with iodine - 131 - labeled anti - tenascin monoclonal antibody 81C6 
therapy , Int. J. Radiat. Oncol. Biol. Phys. , 46 , 947 – 995 . 
402. Yoshida , J. , and Mizuno , M. ( 2003 ), Clinical gene therapy for brain tumors. Liposomal 
delivery of anticancer molecule to glioma , J. Neuro - Oncol. , 65 , 261 – 267 . 
403. Felgner , P. L. , Gadek , T. R. , Holm , M. , Roman , R. , Chan , H. W. , Wenz , M. , Northrop , 
J. R. , Ringold , G. M. , and Danielson , M. ( 1987 ), Lipofection: A highly effi cient, lipid - 
mediated DNA - transfection procedure , Proc. Natl. Acad. Sci. USA , 84 , 7413 – 7414 . 
404. Zhang , Y. , Jeong, Lee , H. , Boado , R. J. , and Pardridge , W. M. ( 2002 ), Receptro - mediated 
delivery of an antisense gene to human brain cancer cells , J. Gene Med. , 4 , 183 – 194 . 
405. Zhang , Y. , Zhu , C. , and Pardridge , W. M. ( 2002 ), Antisense gene therapy of brain cancer 
with an artifi cial virus gene delivery system , Mol. Ther. , 6 , 67 – 72 . 
406. Zhang , Y. , Zhang , Y. F. , Bryant , J. , Charles , A. , Boado , R. J. , and Pardridge , W. M. ( 2004 ), 
Intravenous RNA interference gene therapy targeting the human epidermal growth 
factor receptor prolongs survival in intracranial brain cancer , Clin. Cancer Res. , 10 , 
3667 – 3677 . 
407. Zerrouqi , A. , Rixe , O. , Ghoumari , A. M. , Yarovoi , S. V. , Mouawad , R. , Khayat , D. , and 
Soubrane , C. ( 1996 ), Liposomal delivery of the herpes simplex virus thymidine kinase 
gene in glioma: Improvement of cell sensitization to ganciclovir , Cancer Gene Ther. , 3 , 
385 – 392 . 

530 LIPOSOMES AND DRUG DELIVERY 
408. Voges , J. , Weber , F. , Reszka , R. , Sturm , V. , Jacobs , A. , Heiss , W. D. , Wiestler , O. , and Kapp , 
J. F. ( 2002 ), Clinical protocol; Liposomal gene therapy with the herpes simplex thymidine 
kinase gene/ganciclovir system for the treatment of glioblastoma multiforme , Hum. 
Gene Ther. , 13 , 675 – 685 . 
409. Mizuno , M. , and Yoshida , J. ( 1998 ), Improvement of transduction effi ciency of recombinant 
adeno - associated virus vector by entrapment in multilamellar liposomes , Jpn. J. 
Cancer Res. , 89 , 352 – 354 . 
410. Huynh , G. H. , Deen , D. F. , and Szoka , F. C. , Jr. ( 2006 ), Barriers to carrier mediated drug 
and gene delivery to brain tumours , J. Controlled Release , 110 , 236 – 259 . 
411. Bomgaars , L. , Geyer , J. R. , Franklin , J. , Dahl , G. , Park , J. , Winick , N. J. , Klenke , R. , Berg , 
S. L. , and Blaney , S. M. ( 2004 ), Phase I trial of intrathecal liposomal cytarabine in children 
with neoplastic meningitis , J. Clin. Oncol. , 22 , 3916 – 3921 . 
412. Glantz , M. , LaFollette , S. , Jaeckle , K. , Shapiro , W. , Swinnen , L. , Rozental , J. , Phuphanich , 
S. , Rogers , L. , Gutheil , J. , Batchelor , T. , Lyter , D. , Chamberlain , M. , Maria , B. , Schiffer , 
C. , Bashir , R. , Thomas , D. , Cowens , W. , and Howell , S. B. ( 1999 ), A randomized trial 
of a slow - release versus a standard formulation of cytarabine for the intrathecal treatment 
of lymphomatous meningitis , J. Clin. Oncol. 17 , 3110 – 3116 . 
413. MakCay , A. J. , Deen , D. F. , and Szoka , F. C. , Jr. ( 2005 ), Distribution in brain of liposomes 
after convection enhanced delivery; modulation by particle charge, particle diameter, 
and presence of steric coating , Brain Res. , 1035 , 139 – 153 . 
414. Polfl iet , M. M. , Goede , P. H. , van Kesteren - Hendrikx , E. M. , van Rooijen , N. , Dijkstra , 
C. D. , and van De Berg , T. K. ( 2001 ), Amethod for the selective depletion of perivascular 
and meningeal macrophages in the central nervous system , J. Neuroimmunol. , 116 , 
188 – 195 . 
415. Mamot , C. , Nguyen , J. B. , Pourdehnad , M. , Hadaczek , P. , Saito , R. , Bringas , J. R. , Drummond 
, D. C. , Park , J. W. , and Bankiewicz , K. S. ( 2004 ), Extensive distribution of liposomes 
in rodent brains and brain tumors following convection - enhanced delivery , J. Neuro - 
Oncol. , 68 , 1 – 9 . 
416. Saito , R. , Bringas , J. R. , McKnight , T. R. , Wendland , M. F. , Mamot , C. , Drummond , D. C. , 
Kiprotin , D. B. , Park , J. W. , Berger , M. S. , and Bankiewicz , K. S. ( 2004 ), Distribution of 
liposomes into brain and rat brain tumor models by convection - enhanced delivery 
monitored with magnetic resonance imaging , Cancer Res. , 64 , 2572 – 2579 . 
417. Saito , R. , Krauze , M. T. , Bringas , J. R. , Noble , C. , McKnight , T. R. , Jackson , P. , Wendland , 
M. F. , Mamot , C. , Drummond , D. C. , Kiprotin , D. B. , Hong , K. , Berger , M. S. , Park , J. W. , 
and Bankiewicz , K. S. ( 2005 ), Gadolinium - loaded liposomes allow for real - time magnetic 
resonance imaging of convection - enhaced delivery in the primate brain , Exp. Neurol. , 
196 , 381 – 389 . 
418. Groothuis , D. R. ( 2000 ), The blood - brain and blood - tumor barriers: A review of strategies 
for increasing drug delivery , Neuro - Oncol. , 2 , 45 – 59 . 
419. Imaginis Corp. (The Women ’ s Health Resource) , http://www.imaginis.com/breasthealth/ 
treatment.asp , 1997 – 2007 . 
420. Hofheinz , R. D. , Gnad - Vogt , S. U. , Beyer , U. , and Hochhaus , A. ( 2005 ), Liposomal encapsulated 
anti - cancer drugs , Anti - Cancer Drugs , 16 , 691 – 707 . 
421. Gradishar , W. J. ( 2005 ), The future of breast cancer: The role of prognostic factors , Breast 
Cancer Res. Treat. , 89 , S17 – S26 . 
422. Batist , G. , Ramakrishnan , G. , and Rao , C. S. ( 2001 ), Reduced cardiotoxicity and preserved 
antitumor effi cacy of liposome - encapsulated doxorubicin and cyclophosphamide 
compared with conventional doxorubicin and cyclophosphamide in a randomized, multicenter 
trial of metastatic breast cancer , J. Clin. Oncol. , 19 , 1444 – 1454 . 

REFERENCES 531 
423. Paridaens , R. , Van Aaelst , F. , and Georgoulias , V. ( 2003 ), A randomized phase II study 
of alternating and sequential regimens of docetaxel and doxorubicin as fi rst line chemotherapy 
for metastatic breast cancer , Ann. Oncol. , 14 , 433 – 440 . 
424. Chan , S. , Davidson , N. , Juozaityte , E. , Erdkamp , F. , Pluzanska , A. , Azarnia , N. , and Lee , 
L. W. ( 2004 ), Phase III trial of liposomal doxorubicin and cyclophosphamide compared 
with epirubicin and cyclophosphamide as fi rst line therapy for metastatic breast cancer , 
Ann. Oncol. , 15 , 1527 – 1534 . 
425. Mrozek , E. , Rhoades , C. A. , Allen , J. , Hade , E. M. , and Shapiro , C. L. ( 2005 ), Phase I trial 
of liposomal encapsulated doxorubicin (Myocet ™ ; D - 99) and weekly docetaxel in 
advanced breast cancer patients , Ann. Oncol. , 16 , 1087 – 1093 . 
426. Schmid , P. , Krocker , J. , Jehn , C. , Michniewicz , K. , Lehenbauer - Dehm , S. , Eggemann , H. , 
Heilmann , V. , Kummel , S. , Schulz , C. O. , Dieing , A. , Wischnewsky , M. B. , Hauptmann , S. , 
Elling , D. , Possinger , K. , and Flath , B. ( 2005 ), Primary chemotherapy with gemcitabine 
as prolonged infusion, non - pegylated liposomal doxorubicin and docetaxel in 
patients with early breast cancer: Final results of a phase II trial , Ann. Oncol. , 16 , 
1624 – 1631 . 
427. Keller , A. M. , Mennel , R. G. , Georgoulias , V. A. , Nabholtz , J. M. , Erazo , A. , Lluch , A. , 
Vogel , C. L. , Kaufmann , M. , Minckwitz , G. , Henderson , G. , Mellars , L. , Alland , L. , and 
Tendler , G. ( 2004 ), Randomized phase III trial of pegylated liposomal doxorubicin 
versus vinorelbine or mitomycin C plus vinblastine in women with taxane - refractory 
advanced breast cancer . J. Clin. Oncol. , 22 , 3893 – 3901 . 
428. Vorobiof , D. A. , Rapoport , B. L. , Chasen , M. R. , Slabber , C. , McMichael , G. , Eek , R. , and 
Mohammed , C. ( 2004 ), First in line therapy with paclitaxel (Taxol ® ) and pegylated liposomal 
doxorubicin (Caelyx ® ) in patients with metstatic breast cancer: A multicentre 
phase II study , Breast , 13 , 219 – 226 . 
429. Overmoyer , B. , Silvermann , P. Holder , L. W. , Tripathy , D. , and Henderson , I. C. ( 2005 ), 
Pegylated liposomal doxorubicin and cyclophosphamide as fi rst - line therapy for patients 
with metastatic or recurrent breast cancer , Clin. Breast Cancer , 6 , 150 – 157 . 
430. Coleman , R. E. , Biganzoli , L. , Canney , P. , Dirix , L. , Mauriac , L. , Chollet , P. , Batter , V. , 
Ngalula - Kabanga , E. , Dittrich , C. , and Piccart , M. ( 2006 ), A randomized phase II study 
of two different schedules of pegylated liposomal doxorubicin in metastatic breast 
cancer (EORTC - 10993) , Eur. J. Cancer , 42 , 882 – 887 . 
431. Stover , T. C. , Sharma , A. , Robertson , G. P. , and Kester , M. ( 2005 ), Systemic delivery of 
liposomal short - chain ceramide limits solid tumor growth in murine models of breast 
adenocarcinoma , Clin. Cancer Ther. , 11 , 3465 – 3474 . 
432. Swenson , S. , Costa , F. , Minea , R. , Sherwin , R. P. , Ernst , W. , Fujii , G. , Yang , D. , and 
Markland , F. S. , Jr. ( 2004 ), Intravenous liposomal delivery of the snake venom disintegrin 
contortrostatin limits breast cancer progression , Mol. Cancer Ther. , 3 , 499 – 511 . 
433. Nagami , H. , Matsumoto , Y. , and Ueoka , R. ( 2006 ), Induction of apoptosis by hybrid 
liposomes for human breast tumor calls along with activation caspases , Biol. Pharm. 
Bull. , 29 , 380 – 381 . 
434. Frenkel , V. , Etherington , A. , Greene , M. , Quijano , J. , Xie , J. , Hunter , F. , Dromi , S. , and 
Li , K. C. P. ( 2006 ), Delivery of liposomal doxorubicin (Doxil) in a breast cancer tumor 
model: Investigation of potential enhancement by pulsed - high intensity focused ultrasound 
exposure , Acad. Radiol. , 13 , 467 – 479 . 
435. Mukherjee , A. , Prassad , T. K. , Rao , N. M. , and Banerjee , R. ( 2005 ), Haloperidol - 
associated stealth liposomes , J. Biol. Chem. , 280 , 16(22) , 1561 – 1562 . 
436. Seki , M. , Iwakawa , J. , Cheng , H. , and Cheng , P. W. ( 2002 ), p53 and PTEN/MMAC1/TEP1 
gene therapy of human prostate PC - 3 carcinoma xenograft, using transferrin - facilitated 
lipofection gene delivery strategy , Hum. Gene Ther. , 13 , 761 – 773 . 

532 LIPOSOMES AND DRUG DELIVERY 
437. Basma , H. , El - Refaey , H. , Sgagias , M. K. , Cowan , K. H. , Luo , X. , and Cheng , P. W. ( 2005 ), 
Bcl - 2 antisense and cisplatin combination treatment of MCF - 7 breast cancer cells with 
or without functional p53 , J. Biom. Sci. , 12 , 999 – 1011 . 
438. Meidan , V. M. , Glezer , J. , Salomon , S. , Sidi , Y. , Barenholz , Y. , Cohen , J. S. , and Lilling G. 
( 2006 ), Specifi c lipoplex - mediated antisense against bcl - 2 in breast cancer cells: A comparison 
between different formulations , J. Liposome Res. , 16 , 27 – 43 . 
439. Gradishar , W. J. ( 2005 ), The future of breast cancer: The role of prognostic factors , Breast 
Cancer Res. Treat. , 89 , S17 – S26 . 
440. NetDoctor.co.uk , http://www.netdoctor.co.uk/diseases/facts/lungcancer.htm , 2007 . 
441. http://www.merck.de/servlet/PB/menu/1508710/index.html . 
442. Leighl , N. , Burkes , R. L. , Dancey , J. E. , Lopez , P. G. , Higgins , B. P. , Walde , P. L. D. , 
Rudinskas , L. C. , Rahim , Y. H. , Rodgers , A. , Pond , G. R. , and Shepherd , F. A. ( 2003 ), 
A phase I study of pegylated liposomal doxorubicin (Caelyx ™ ) in combination with 
cyclophosphamide and vincristine as second - line treatment of patients with small - cell 
lung cancer , Clin. Lung Cancer , 5 , 107 – 112 . 
443. Peer , D. , and Margalit , R. ( 2004 ), Tumor - targeted hyaluronan nanoliposomes increase 
the antitumor activity of liposomal doxorubicin in syngeneic and human xenograft 
mouse tumor models , Neoplasia , 6 , 343 – 353 . 
444. Ito , I. , Ji , L. , Tanaka , F. , Saito , Y. , Gopalan , B. , Branch , C. D. , Xu , K. , Atkinson , E. N. , 
Bekele , B. N. , Stephens , L. C. , Minna , J. D. , Roth , J. A. , and Ramesh , R. ( 2004 ), Liposomal 
vector mediated delivery of the 3p FUS1 gene demonstrate potent antitumor activity 
against human lung cancer in vivo , Cancer Gene Ther. , 11 , 733 – 739 . 
445. Li , W. , Ishida , T. , Okada , N. , and Kiwada , H. ( 2005 ), Increased gene expression by cationic 
liposomes (TFL - 3) in lung metastases following intravenous injection , Biol. Pharm. 
Bull. , 28 , 701 – 706 . 
446. Kawakami , A. , Suzuki , S. , Yamashita , F. , and Hashida , M. ( 2006 ), Induction of apoptosis 
in A549 human lung cancer cells by all - trans retinoic acid incorporated in DOTAP/ 
cholesterol liposomes , J. Controlled Release , 110 , 514 – 521 . 
447. Remaut , K. , Lucas , B. , Braeckmans , K. , Sanders , N. N. , Demeester , J. , and De Smedt , S. 
C. ( 2006 ), Delivery of phosphodiester oligonucleotides: Can DOTAP/DOPE liposomes 
do the trick? Biochemistry , 45 , 1755 – 1764 . 
448. American Cancer Society, Inc. , http://www.cancer.org , 2007 . 
449. Johnston , S. ( 2004 ), Ovarian cancer: Review of the national institute for clinical excellence 
(NICE) guidance recommendations , Cancer Inv. , 22 , 730 – 742 . 
450. Rose , P. ( 2005 ), Pegylated liposomal doxorubicin: Optimizing the dosing schedule in 
ovarian cancer , Oncologist , 10 , 205 – 214 . 
451. Tambaro , R. , Greggi , S. , Iaffaioli , R. V. , Rossi , A. , Pisano , C. , Manzione , L. , Ferrari , E. , 
Di Maio , M. , Iodice , F. , Casella , G. , Laurelli , G. , and Pignata , S. ( 2003 ), An escalating 
dose fi nding study of liposomal doxorubicin and vinorelbine for the treatment of refractory 
or resistant epithelial ovarian cancer , Ann. Oncol. , 14 , 1406 – 1411 . 
452. Katsaros , D. , Oletti , M. V. , Rigault de la Longrais , I. A. , Ferrero , A. , Celano , A. , Fracchioli , 
S. , Donadio , M. , Passera , R. , Cattel , L. , and Bumma , C. ( 2005 ), Clinical and pharmacokinetic 
phase II study of pegylated liposomal doxorubicin and vinorelbine in heavily 
pretreated recurrent ovarian carcinoma , Ann Oncol. , 16 , 300 – 306 . 
453. Gordon , A. N. , Fleagle , J. T. , Guthrie , D. , et al . ( 2001 ), Recurrent epithelial ovarian carcinoma: 
A randomized phase III study of pegylated liposomal doxorubicin versus topotecan 
, J. Clin. Oncol. , 19 , 3312 – 3322 . 
454. Verhaar - Langereis , M. , Karakus , A. , van Eijkeren , M. , Voest , E. , and Witteveen , E. 
( 2006 ), Phase II study of the combination of pegylated liposomal doxorubicin and topotecan 
in platinum - resistant ovarian cancer , Int. J. Gynecol. Cancer , 16 , 65 – 70 . 

REFERENCES 533 
455. Seiden , M. V. , Muggia , F. , Astrow , A. , Matulonis , U. , Campos , S. , Roche , M. , Sivret , J. , 
Rusk , J. , and Barrett , E. ( 2004 ), A phase II study of liposomal lurtotecan (OSI - 211) in 
patients with topotecan resistant ovarian cancer , Gynecol. Oncol. , 93 , 229 – 232 . 
456. Calvert , A. H. , Grimshaw , R. , Poole , C. , Dark , G. , Swnerton , K. , Gore , M. , et al . ( 2002 ), 
Randomized phase II trial of two intravenous schedules of the liposomal topoisomerase 
I inhibitor, NX211, in women with relapsed epithelial ovarian cancer (OVCA): An NCIC 
CTG study , Proc. Am. Soc. Clin. Oncol. , 21 , 208a . 
457. National Cancer Institute, U.S. National Institutes of Health , http://www.cancer.gov/ 
search/ResultsClinicalTrials , 2007 . 
458. Gallo , D. , Fruscella , E. , Ferlini , C. , Apollonio , P. , Mancuso , S. , and Scambia , G. ( 2006 ), 
Preclinical in vivo activity of a combination gemcitabine/liposomal doxorubicin against 
cisplatin - resistant human ovarian cancer (A2780/CDDP) , Int. J. Gynecol. Cancer , 16 , 
222 – 230 . 
459. Anderson , K. , Lawson , K. A. , Simmons - Menchaca , M. , Sun , L. , SAnders , B. G. , and Kline , 
K. ( 2004 ), . - TEA plus cisplatin reduces human cisplatin - resistant ovarian cancer cell 
tumor burden and metastasis , Exp. Biol. Med. , 229 , 1169 – 1176 . 
460. Turk , M. J. , Waters , D. J. , and Low , P. S. ( 2004 ), Folate - conjugated liposomes preferentially 
target macrophages associated with ovarian carcinoma , Cancer Lett. , 213 , 165 – 172 . 
461. Kim , C. K. , Choi , E. J. , Choi , S. H. , Park , J. S. , Haider , K. H. , and Ahn , W. S. ( 2003 ), 
Enhanced p53 gene transfer to human ovarian cancer cells using the cationic nonviral 
vector, DDC , Gynecol. Oncol. , 90 , 265 – 272 . 
462. Landen , C. N. , Jr. , Chavez - Reyes , A. , Bucana , C. , SchmAndt , R. , Deavers , M. T. , Lopez - 
Berestein , G. , and Sood , A. K. ( 2005 ), Therapeutic EphA2 gene targeting in vivo using 
neutral liposomal small interfering RNA delivery , Cancer Res. , 65 , 6910 – 6918 . 


535 
5.4 
BIODEGRADABLE NANOPARTICLES 
Sudhir S. Chakravarthi and Dennis H. Robinson 
University of Nebraska Medical Center, College of Pharmacy, Omaha, Nebraska 
Contents 
5.4.1 Introduction 
5.4.1.1 Classifi cation of Nanoparticles 
5.4.2 Natural Biodegradable Polymeric Nanoparticles 
5.4.2.1 Physical Properties of Natural Polymers and Methods Used to Prepare 
Nanoparticles 
5.4.2.2 Drug Delivery Applications and Biological Fate of Natural Polymeric 
Nanoparticles 
5.4.3 Synthetic Biodegradable Polymeric Nanoparticles 
5.4.3.1 Synthetic Polymers: Physical Properties and Methods of Preparation of 
Nanoparticles 
5.4.3.2 Drug Delivery Applications and Biological Fate of Synthetic Biodegradable 
Polymers 
5.4.4 Thermosensitive and pH - Sensitive Nanoparticles 
5.4.4.1 Physical Properties and Methods of Preparation 
5.4.4.2 Drug Delivery Applications and Biological Fate of Thermosensitive and 
pH – Sensitive Nanoparticles 
5.4.5 Applications of Biodegradable Nanoparticles Other Than Drug Delivery 
5.4.6 Physicochemical Characterization of Polymeric Nanoparticles 
5.4.6.1 Molecular Weight 
5.4.6.2 Hydrophobicity 
5.4.6.3 Glass Transition Temperature 
5.4.6.4 Particle Size and Particle Size Distribution 
5.4.6.5 Surface Charge and Zeta Potential 
5.4.6.6 Surface Hydrophilicity 
5.4.6.7 Drug Loading and Encapsulation Effi ciency 
5.4.6.8 Drug Release 
5.4.6.9 Physical Stability of Polymeric Nanoparticles 
5.4.7 Targeting Nanoparticles by Surface Conjugation with Ligands 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

536 BIODEGRADABLE NANOPARTICLES 
5.4.8 Cellular Traffi cking of Biodegradable Nanoparticles 
5.4.9 Conclusions 
References 
5.4.1 INTRODUCTION 
Interest in nanotechnology has increased exponentially in many scientifi c areas, 
including drug delivery, nanoimaging, and other medical - related applications. 
Nanoparticles can be fabricated in many different shapes and sizes using a wide 
range of organic and inorganic materials. However, by defi nition, these particles 
must be within the size range of 1 – 1000 nm. Because the use of nanoparticles in drug 
delivery and nanomedicine invariably requires parenteral administration, there has 
been, and continues to be, a major need for the use of polymeric carriers that are 
both biocompatible and biodegradable. This review will focus on the application of 
nanotechnology to deliver therapeutic or diagnostic agents using biodegradable 
polymeric nanoparticles for systemic, localized, or targeted delivery. 
5.4.1.1 Classifi cation of Nanoparticles 
Depending on the method of preparation and resulting structure, nanoparticles are 
broadly classifi ed as either matrix - type or encapsulated particles. Hence, a drug is 
either homogenously dispersed in the polymeric matrix or encapsulated within the 
core of the particle. In drug delivery applications, biodegradable polymers used to 
prepare nanoparticles are either natural or synthetic in origin (Table 1 ). Natural 
polymers, or biopolymers, include alginates, chitosan, cellulose, gelatin, gliadin, and 
pullulan. A recent review describes the applications of these polymers in gene delivery 
and tissue engineering [1] . Natural polymers may vary widely in their composition 
and therefore physicochemical properties. Such variability in properties may 
result in poor reproducibility in delivery characteristics, such as drug loading and 
release kinetics. Further, their purifi cation from natural sources may be diffi cult. In 
contrast, synthetic polymers can be prepared with relatively precise properties such 
as molecular weight, solubility, and permeability characteristics. Examples of synthetic 
polymers used to make biodegradable nanoparticles include polylactide, 
poly(lactide -co - glycolide) (PLGA), polyanhydride, polycyanoacrylate, poly( . - 
caprolactone), and polyphosphoester. As liposomes are more commonly prepared 
in the micrometer - size range, they were considered out of the scope of this review. 
Because there are numerous reviews of these polymers in the literature, their physicochemical 
properties, methods of synthesis, applications, and biological fate of each 
of these polymers are only briefl y described in this chapter. 
5.4.2 NATURAL BIODEGRADABLE POLYMERIC NANOPARTICLES 
Natural polymers extracted and purifi ed from plant and animal sources often vary 
signifi cantly in their purity. For example, alginate is available in over 200 different 

TABLE 1 Classifi cation of Natural and Synthetic Polymers and Their Methods of 
Purifi cation or Synthesis 
Polymer Source 
Method of 
Purifi cation or 
Synthesis Solubility 
Sodium alginate Natural 
(seaweed) 
Alkali - based 
extraction 
Water soluble (pH > 
3), insoluble in 
organic solvents 
Chitosan Natural 
(crab 
shells) 
Deacetylation of 
chitin 
Soluble in aqueous 
solutions (low pH), 
insoluble in organic 
solvents 
Gelatin Natural 
(collagen) 
Hydrolysis Soluble in hot water 
(> 34 ° C), acetic acid, 
forms insoluble gel 
with water at room 
temperature, 
insoluble in organic 
solvents 
Polysaccharides Natural Enzymatic reactions Pullulans (soluble in 
water), dextrans 
(soluble in water) 
Albumin Natural 
(plants, 
animals) 
Separation 
techniques 
(chromatography) 
Soluble in water 
Gliadin Natural 
(wheat) 
Alcohol extraction Insoluble in water, 
soluble in ethanol 
Poly(lactide) and 
Poly(lactide- co -glycolide) 
Synthetic Ring - opening 
polymerization 
Insoluble in water, 
soluble in organic 
solvents 
Poly( . - caprolactone) Synthetic Anionic, cationic, 
free - radical, ring - 
opening 
polymerization 
Soluble in select 
organic solvents 
such as chloroform, 
dichloromethane 
Polyanhydrides Synthetic Melt condensation, 
ring - opening 
polymerization 
Most polyanhydrides 
soluble in organic 
solvents, insoluble in 
water 
Poly - alkylcyanoacrylates Synthetic Emulsion and 
interfacial 
polymerization 
Soluble in organic 
solvents 
Polyphosphoesters Synthetic Polyaddition, ring - 
opening 
polymerization 
Available as water - 
soluble and water - 
insoluble types 
grades and is extracted from various sources that differ in molecular weight and the 
percentage and arrangement of guluronic and mannuronic acid blocks. Further, 
chitosan, poly - . (1 - 4 - d - glucosamine), is available in grades varying in molecular 
weight, degree of deacetylation (from parent compound, chitin), and viscosity [2] . 
NATURAL BIODEGRADABLE POLYMERIC NANOPARTICLES 537

538 BIODEGRADABLE NANOPARTICLES 
Some natural polymers may be chemically modifi ed to tailor solubility properties. 
An example is the reaction of the free amino groups of chitosan to form the more 
water soluble derivative, methoxy - polyethylene glycol (PEG) chitosan [3] . Collagen 
is marketed as six different types, I – VI, depending on its source and physiological 
applicability. Similarly, the properties of gelatin are dependent on the method of 
preparation using acid - or base - catalyzed hydrolysis from collagen. 
5.4.2.1 Physical Properties of Natural Polymers and Methods Used to 
Prepare Nanoparticles 
Sodium Alginate Alginates are primarily derived from the algae Macrocystis pyrifera 
and Laminaria hyperborea . These are linear, unbranched polymers containing 
. - (1 – 4) - linked mannuronic acid and . - (1 – 4) - linked guluronic acid residues that are 
either arranged in blocks, commonly called G blocks and M blocks, or alternate with 
each other. Alginates are hydrophilic, anionic polymers that vary in molecular 
weight, depending primarily on the G and M blocks. They are characterized by the 
ratio of guluronic and mannuronic acids, which can be quantifi ed by ultraviolet 
(UV) spectrophotometry, gas chromatography, and high - performance liquid chromatography 
(HPLC) [4] . For example, the polymer obtained from M. pyrifera has 
an M/G ratio of 1.6. Alginate obtained from seaweed must be purifi ed by one of 
several applicable alkali and acid treatment protocols [5, 6] . 
Alginate nanoparticles can be prepared using ionotropic gelation, emulsifi cation/
internal gelation, and emulsifi cation/solidifi cation methods. Ionotropic gelation 
results when the anionic alginate reacts with cationic ions or molecules such as 
calcium or poly - l - lysine. Gelation occurs when cations chelate the guluronic and 
mannuronic acid groups to produce an “ egg - box ” structure that encapsulates the 
drug. The size of the alginate particles is determined by the molar concentration of 
calcium or poly - l - lysine and the method of addition of these counterions to alginate 
[7] . In the emulsifi cation/internal gelation method, the sodium alginate and an 
insoluble calcium salt are dispersed in a vegetable oil and the calcium ions are liberated 
to form an alginate gel when the pH of the dispersion is lowered [8, 9] . An 
advantage of the use of alginate polymers to deliver drugs is that nanoparticles are 
prepared in aqueous media and may be more suitable to formulate compounds that 
are unstable in organic solvents such as proteins and peptides. However, since the 
chelation to form the gel is reversible, a disadvantage of unmodifi ed, alginate - based 
delivery systems is rapid drug release due to the collapse of the egg - box structure 
when exposed to monovalent ions in physiological media. 
Chitosan Chitosan is a nontoxic, biodegradable polymer obtained by hydrolysis 
of chitin, a natural polysaccharide that is a chief component of the crustacean exoskeleton. 
Unmodifi ed chitosan is soluble in acidic media and has signifi cant mucoadhesive 
properties. 
Chitosan nanoparticles may be prepared using various methods, including emulsion 
cross - linking, coacervation – precipitation, spray drying, emulsion droplet 
coalescence, ionic gelation, reverse - micellar method, and sieving. A relatively recent 
review describes the methods and applications of chitosan nanoparticles in drug 
delivery [10] . Chitosan nanoparticles have also been prepared using water - soluble 
cross - linking agents such as carbodiimide with the size being controlled by changing 

pH [11] . More monodisperse nanoparticles may be prepared using fractionated and 
deacetylated chitosan [12] . In general, the size of nanoparticles will depend on the 
molecular weight of chitosan, its concentration, and its surface charge [13] . The 
physicochemical properties of chitosan are determined by the solution pH and ionic 
strength [14] . 
Gelatin Gelatin is obtained by either alkaline or acidic hydrolysis of collagen. It 
has a triple helical structure with a high content of glycine, proline, and hydroxyproline 
residues. Gelatin that is formed from alkaline treatment of collagen has 
more carboxyl groups and a lower isoelectric point than that derived from acidic 
hydrolysis [15] . The physicochemical properties of gelatin depend on the method 
of extraction and the extent of thermal denaturation that occurs during the 
purifi cation. 
Gelatin nanoparticles can be prepared by various methods, including chemical 
cross - linking, water - in - oil (w/o) emulsifi cation, and desolvation. Gelatin is cross - 
linked with agents such as glutaraldehyde. Effi cient cross - linking usually results in 
decreased rate of drug release. The w/o emulsifi cation involves extruding a preheated, 
aqueous solution of gelatin into vegetable oils, such as corn or olive oils 
[15] . The two - step desolvation method involves the dropwise addition of a water - 
miscible nonsolvent such as acetone and ethanol [16] . While the use of collagen, 
the parent compound of gelatin, in drug delivery is rare, collagen nanoparticles 
have been used to deliver genes by exploiting the electrostatic interaction between 
the positively charged polymer and negatively charged deoxyribonucleic acid 
(DNA) [17] . 
Polysaccharides The macromolecular polysaccharides that include pullulan, 
mannan, and dextran are the main constituents of the cellular glycocalyx and play 
an important role in cell – cell adhesion and the cell – cell recognition process [18] . 
Pullulan is a nonimmunogenic, nontoxic, water - soluble, linear, nonionic polysaccharide 
with . (1 – 4) and . (1 – 6) linkages with free hydroxyl groups for drug conjugation 
[19] . Pullulans are intracellularly synthesized and secreted by a fungus, 
Aureobasidium pullulans [20] . On the other hand, dextrans are anionic glucose 
polymers derived from sucrose with . (1 – 6) glucosidic linkage. A class of enzymes, 
glucansucrases, produced by two genera of lactic acid bacteria, namely, Leuconostoc 
and Streptococcus, catalyze the synthesis of dextrans from sucrose. The extraction 
of dextrans as well as their physical properties and drug delivery aspects has been 
reviewed [21, 22] . When coated with mannans, the biological response of both 
natural and synthetic polymeric nanoparticles may be changed [23] . 
To make pullulan nanoparticles, the polymer must fi rst be made hydrophobic, 
typically by conjugating alkyl groups, cholesterol groups, or succinyl groups. Hydrophobic 
pullulans self - assemble to form stable hydrogel nanoparticles [24] . Alternately, 
pullulan nanoparticles can be formed by cross - linking reverse micelles of the 
polymer with glutaraldehyde [25] . Nanoparticles form when a solution of pullulan 
acetate in N,N - dimethyl acetamide is used and dialyzed with borate buffer [26] . 
Dextran is commonly conjugated to other polymers, such as PLGA, PEG, 
polystyrene, and poly(methyl methacrylate) for preparation of nanoparticles. 
Complex coacervation has been used to prepare dextran nanoparticles using 
oppositely charged polymers such as polyethyleneimine [27] . Although nanoparticle 
NATURAL BIODEGRADABLE POLYMERIC NANOPARTICLES 539

540 BIODEGRADABLE NANOPARTICLES 
formulation using mannans has not been reported, mannan - coated nanoparticles 
increased cell binding and macrophage uptake [28] . 
Albumin Human serum albumin, the most abundant plasma protein, is a positively 
charged, multifunctional protein and is involved in transport, ligand binding, and 
enzymatic activities. Albumin is a globular protein containing approximately 585 
amino acids in an . - helical tertiary structure. Several exogenous and endogenous 
compounds can covalently or reversibly bind to albumin, and because of the 
excellent adsorptive properties of human serum albumin, this polypeptide can be 
adsorbed onto the surface of polymeric nanoparticles [29] . In addition, as it is 
amphoteric, albumin can be used as a surfactant during the preparation of 
nanoparticles where it is irreversibly adsorbed onto the surface of biodegradable 
polymers such as poly(lactic acid) (PLA) and PLGA [30] . These protein – particle 
interactions are mainly driven by electrostatic forces further stabilized by hydrophobic 
forces [31] . 
Albumin nanoparticles can be prepared by controlled desolvation, pH - induced 
coacervation, and/or chemical cross - linking with glutaraldehyde. Briefl y, the pH of 
an aqueous solution of albumin is raised to about 9.0 and nanoparticles are precipitated 
by adding a miscible cosolvent such as acetone [32] . A study reports attempts 
to optimize the desolvation method to prepare albumin nanoparticles of a more 
controlled particle size and narrower particle size distribution [33] . 
Gliadin Gliadin, a glycoprotein derived from gluten, is extracted from wheat and 
separated by capillary electrophoresis [34, 35] . Gliadin is classifi ed as . - 5, . - 1,2, . , 
and . - type based on its structure and electrophoretic mobility [36] . The glycoprotein 
is water insoluble due to the presence of interpolypeptide disulfi de bonds and 
hydrophobic interactions. A limitation on the use of gliadins is that patient sensitivity 
causes an autoimmune disorder called celiac disease. 
Gliadin nanoparticles are prepared by the desolvation method by fi rst pouring 
an organic solution of polymer into an aqueous phase such as physiological saline 
containing a surfactant stabilizer (e.g., Pluronic). The nanoparticles are formed by 
evaporating the organic solvent. 
5.4.2.2 Drug Delivery Applications and Biological Fate of Natural 
Polymeric Nanoparticles 
Alginate Nanoparticles Alginate nanoparticles have been used to formulate a 
wide range of drugs. Because they are prepared in an aqueous environment under 
mild conditions, alginate nanoparticles are particularly suitable for formulating 
proteins, peptides, and oligonucleotides [37] . Further, in addition to being biodegradable, 
alginates are nonimmunogenic. To decrease the rate of exchange of cations 
such as Ca 2+ with monovalent ions in the dissolution medium, the anionic alginates 
are often treated with cationic molecules such as chitosan, poly - l - lysine, or tripolyphosphate. 
Some examples of the wide range of applications of alginate - based 
nanoparticles are described. Alginate nanoparticles prepared with tripolyphosphate 
were used in oral delivery [38] . A study of physical properties demonstrated that 
alginate – chitosan nanoparticles are suitable for the delivery of DNA [39] . Alginate - 
coated chitosan nanoparticles increased stability and decreased the burst release of 

ovalbumin [40] . A study reported that chitosan - stabilized alginate nanoparticles 
increased bioavailability and sustained release of antifungal drugs compared to 
PLGA nanoparticles [41] . Although predominantly used for oral administration, 
inhaled alginate nanoparticles improved the bioavailability of antitubercular drugs 
[42] . In vivo, alginate nanoparticles accumulate in the Kupffer cells, parenchymal 
cells of liver, and phagocytes of spleen and lungs [43, 44] . Alginate nanoparticles 
have also been reported to be absorbed into Peyer ’ s patches, suggesting that they 
may enhance targeting to the intestinal mucosa [45] . In the body, the alginates 
degrade by acidic hydrolysis of the guluronic and mannuronic segments [46] . 
Chitosan Nanoparticles In addition to low - molecular - weight drugs and nutraceuticals, 
chitosan nanoparticles are widely used in the delivery of macromolecules such 
as DNA and small interfering ribonucleic acid (siRNA) [47] . Apart from sustaining 
the release of macromolecules, chitosan nanoparticles protect them from nucleases. 
Placebo chitosan nanoparticles exhibited antibacterial activity against several 
microbes, including Escherichia coli [48] . The surface of chitosan nanoparticles was 
hydrophobically modifi ed with linoleic acid for delivery of trypsin [49] . Other applications 
of chitosan nanoparticles include lung [50] and ocular delivery [51] . The 
primary amine group at the 2 position can be modifi ed to tailor chitosan for specifi c 
applications. For example, chemical conjugation of these amine groups to methoxy - 
PEG groups increased water solubility [52] . Thiolation of chitosan enhanced the 
mucosal permeation of the nanoparticles [53] . Hydrophobically modifi ed glycol 
chitosans that self - assemble into nanoparticles have been used to deliver doxorubicin 
[54] . Targeting chitosan nanoparticles to folate receptors on the surface of cells 
enhanced the transfection effi ciency of DNA [55] . N - Succinyl chitosan nanoparticles 
containing 5 - fl uorouracil demonstrated excellent activity against sarcoma tumors 
[56] . Self - assembled N - acetyl histidine – conjugated glycol chitosan nanoparticles 
were effi ciently internalized into cells by adsorptive endocytosis [57] . No toxic 
effects have been observed with chitosan nanoparticles. Upon intravenous (i.v.) 
administration, chitosan nanoparticles accumulated in the liver with minimal concentrations 
in the heart and lung [54] . 
Gelatin Nanoparticles Gelatin nanoparticles have been used as a delivery system 
for several drugs, including pilocarpine, hydrocortisone [58] , methotrexate [59] , 
paclitaxel [60] , and chloroquine [61] . High protein loading and sustained release 
were achieved using composite gelatin and PLGA nanoparticles [62] . Surprisingly, 
placebo gelatin nanoparticles exhibited antimelanoma activity in vivo [63] . Primary 
amine groups of gelatin molecule can be chemically conjugated or cross - linked using 
bifunctional cross - linkers. This is demonstrated in the delivery of biotinylated 
peptide nucleic acid using avidin - cross - linked gelatin nanoparticles [64] . PEGylation 
of gelatin nanoparticles containing hydrophilic drugs prolonged circulation time in 
the body [65] . Thiolated gelatin nanoparticles produced effective transfection of 
plasmid DNA encoding the green fl uorescent protein [66] . Following endocytic 
uptake by the cells, gelatin nanoparticles concentrate in the perinuclear region [65] . 
In vitro, the gelatin nanoparticles are effi ciently internalized into macrophages and 
monocytes [67] . In tumor - bearing mice, PEGylated gelatin nanoparticles predominantly 
accumulated in the liver and tumor [68] while in dendritic cells they are primarily 
localized in the lysosomes [69] . In vivo, gelatin is degraded by proteases to 
NATURAL BIODEGRADABLE POLYMERIC NANOPARTICLES 541

542 BIODEGRADABLE NANOPARTICLES 
amino acids. Although cardiotoxicity and mild immunogenicity were reported with 
gelatin nanoparticles covalently coupled to doxorubicin, this was attributed to the 
coupling reagent glutaraldehyde [70] . 
Pullulan and Dextran Nanoparticles Pullulan nanoparticles successfully delivered 
HER2 oncoprotein to induce humoral and cellular immune responses against 
HER2 - expressing murine sarcomas [71] . Hydrophobic polysaccharides such as pullulan 
and mannan enable soluble proteins to induce cellular immunity and therefore 
may be a potential delivery vehicle for vaccines [72] . pH - sensitive pullunan nanoparticles 
prepared by conjugating sulfonamides are stable at physiological pH but 
aggregate and release the encapsulated drug when exposed to the lower tumor pH 
[73] . Coating of magnetic nanoparticles with pullulan enhanced their cellular uptake 
by endocytosis [74] . Amphotericin - loaded, dextran – polyethyleneimine nanoparticles 
were active against Candida albicans [27] . Similarly, insulin - containing dextran – 
polyethyleneimine nanoparticles prolonged the hypoglycemic effect in diabetic 
rats [75] . Hydrogels prepared from blends of polyvinyl alcohol and dextrans have 
been used as matrices to entrap PLGA nanoparticles [76] . In vitro, immuno- 
fl uorescent staining illustrated that pullulan nanoparticles are internalized by active 
endocytosis [25] . However, other studies suggest that both absorption and 
internalization of pullulan nanoparticles are inhibited by coating them with dextran 
[77] . 
Albumin Nanoparticles A signifi cant development in the drug delivery of albumin 
nanoparticles has been the recent marketing of the commercial product Abraxane ® 
for chemotherapy of breast cancer. This delivery system is prepared by nab TM 
technology, which involves noncovalent complexation of albumin with paclitaxel. 
Albumin nanoparticles have been used to deliver antisense oligonucleotides, interferon 
. , and anticytomegaloviral drugs [78, 79] . Intravitreal injection of gancicyclovir 
- loaded albumin nanoparticles was attempted to prolong residence time in the 
eye [79] . PLA nanoparticles coated with albumin degrade more rapidly in the gastrointestinal 
(GI) region, resulting in effi cient delivery of water - soluble drugs across 
the GI tract [80] . In addition, intra - arterial chemotherapy with paclitaxel - containing 
albumin nanoparticles effectively treated squamous cell carcinoma [81] . Human 
serum albumin – polyethyleneimine nanoparticles optimized transfection of the 
luciferase gene in human, embryonic, and epithelial kidney cells [82] . Conjugation 
of the cellular targeting agent, folic acid, resulted in increased cellular uptake of 
albumin nanoparticles compared to unmodifi ed particles [83, 84] . Transferrin - conjugated 
PEGylated albumin nanoparticles demonstrated enhanced uptake into the 
brain tissues [85] . Glycyrrhizin was conjugated to the amine groups of albumin 
nanoparticles targeting hepatocytes [86] . After i.v. administration, albumin - coated 
PLA nanoparticles were distributed in the liver, bone marrow, lymph nodes, spleen, 
and peritoneal macrophages [87] . In the body, albumin nanoparticles are actively 
taken up by macrophages. 
Gliadin Nanoparticles The hydrophobic nature of gliadin makes this polymer 
ideal for delivery of hydrophobic compounds such as all - trans retinoic acid and 
vitamin E. Gliadin nanoparticles adhered to the stomach mucosa and signifi cantly 
increased the bioavailability of carbazole [88] . Typically, encapsulation effi ciency 

and drug loading of gliadin nanoparticles are higher for hydrophobic drugs [89] . 
Gliadin nanoparticles were targeted to Helicobacter pylori by chemical conjugation 
of lectin glycoproteins to their surface, resulting in a twofold increase in inhibition 
of bacterial activity compared to unmodifi ed nanoparticles [90] . Although gliadins 
adhere to the mucosa, internalization of gliadin nanoparticles into cells has not been 
reported. More defi nitive studies are required to fully understand the biological fate 
of these particles. 
5.4.3 SYNTHETIC BIODEGRADABLE POLYMERIC NANOPARTICLES 
A detailed description of the methods of polymerization and variables employed in 
polymer synthesis are beyond the scope of this review and can be found in many 
texts and review articles. The focus of this section is to provide an overview of the 
properties and methods used to prepare nanoparticles from each class of synthetic 
polymer. 
5.4.3.1 Synthetic Polymers: Physical Properties and Methods of Preparation 
of Nanoparticles 
Poly(lactic acid) and Poly(lactide -co-glycolide) These poly - hydroxy acids are 
approved for human use by the Food and Drug Administration (FDA) and have 
been widely used to prepare biodegradable nanoparticles. PLA exists in optically 
active and inactive forms and is a semicrystalline, hydrophobic molecule that 
degrades in the body over a period of months. Conversely, poly(glycolic acid) is 
amorphous and hydrophilic and degrades more rapidly than PLA. In aqueous 
media, these polymers degrade by random hydrolysis of ester bonds that is autocatalyzed 
in acidic media to form lactic and glycolic acids [91] . The factors that affect 
the rate of hydrolytic degradation include type and composition of the polymer 
backbone, nature of pendent groups, molecular weight, pH, enzymes, and geometry 
of the delivery device. 
The preparation and characterization of PLA and PLGA nanoparticles have 
been extensively reviewed elsewhere [92, 93] . Various techniques may be used to 
prepare PLA and PLGA nanoparticles, including simple and multiple emulsions, 
nanoprecipitation, gas antisolvent method, supercritical fl uid technology, coacervation/
phase separation, and spray drying [91] . Briefl y, in the single - emulsion method, 
an organic solution of the polymer and drug is emulsifi ed with an aqueous solution 
of surfactant such as polyvinyl alcohol (PVA). While PLA and PLGA nanoparticles 
containing hydrophobic drugs are prepared by the two - phase emulsion method, a 
w/o/w multiple - emulsion method is needed to encapsulate hydrophilic drugs. In the 
phase separation method, the addition of a nonsolvent precipitates or coacervates 
the polymer from solution to encapsulate the drug. The experimental variables for 
each protocol can be altered to infl uence the physicochemical properties, such as 
particle size, particle size distribution, morphology, and zeta potential [93] . The 
release of encapsulated drug from PLA and PLGA nanoparticles may occur by a 
combination of diffusion and polymer degradation at a rate that is infl uenced by 
properties of the polymer and nanoparticles and the environment. The surface of 
SYNTHETIC BIODEGRADABLE POLYMERIC NANOPARTICLES 543

544 BIODEGRADABLE NANOPARTICLES 
both PLGA and PLA nanoparticles can be modifi ed to target cells and organs by 
conjugation with ligands such as folates, transferrin, HIV - TAT, aptamers, heparin, 
and lectins. The negative zeta potential of PLA and PLGA nanoparticles can be 
altered by coating with cationic polymers such as chitosan and polyethyleneimine, 
which promote nanoparticle – cell interaction. As with liposomes, PEGylation of 
PLGA nanoparticles prolongs circulation times in the body. 
Poly( e-caprolactone) Poly( . - caprolactone) is a semicrystalline polymer synthesized 
by anionic, cationic, free - radical, or ring - opening polymerization [94] . It is available 
in a range of molecular weights and degrades by bulk hydrolysis autocatalyzed 
by the carboxylic acid end groups. The presence of enzymes such as protease, amylase, 
and pancreatic lipase accelerates polymer degradation [95] . The various methods of 
preparation of poly( . - caprolactone) nanoparticles include emulsion polymerization, 
interfacial deposition, emulsion – solvent evaporation, desolvation, and dialysis. These 
methods and various applications are extensively reviewed [94] . 
Polyanhydrides Polyanhydrides have a hydrophobic backbone with a hydrolytically 
labile anhydride linkage. These polymers widely vary in chemical composition 
and include aliphatic, aromatic, and fatty acid – based polyanhydrides. The rate of 
degradation depends on the chemical composition of the polymer. In general, aliphatic 
polyanhydrides degrade more rapidly than the aromatic polymer. Hence, 
copolymer blends with varying ratios of aliphatic - to - aromatic polyanhydrides can 
be synthesized to suit specifi c applications. 
The synthesis and physical properties of polyanhydrides have been reviewed [96, 
97] . Polyanhydride nanospheres are commonly prepared by the emulsion – solvent 
evaporation method using PVA as a stabilizer. However, as polyanhydrides are 
hydrolabile, they need to be fl ash frozen in liquid nitrogen and lyophilized immediately 
[98] . An example of their use to deliver drugs is entrapment of bovine zinc 
insulin by phase inversion nanoencapsulation [99] . Although not used to formulate 
nanoparticles, polyanhydride microspheres have been prepared using alternate 
techniques involving nonaqueous solvents, such as solid/oil/oil double emulsion and 
cryogenic atomization techniques [100] . The surface of polymeric nanoparticles can 
be modifi ed for targeted delivery by reaction of the anhydride with an amino group 
to form an amide linkage with a ligand [101] . However, application of the ligand - 
conjugated nanoparticles for drug delivery is yet to be explored extensively. 
Poly(alkyl -cyanoacrylates) As poly(alkyl - cyanoacrylates) form strong bonds 
with polar substrates including the skin and living tissues, they exhibit bioadhesive 
properties. These polymers are synthesized by free - radical, anionic, or zwitterionic 
polymerization. As detailed in a recent review, poly(alkyl - cyanoacrylate) nanoparticles 
are prepared by emulsion polymerization, interfacial polymerization, nanoprecipitation, 
and emulsion – solvent evaporation methods [102] . 
Solid–lipid Nanoparticles Solid – lipid nanoparticles (SLNs) are obtained by high - 
pressure homogenization of molten lipids in the presence of surfactant. The major 
advantage of using SLN is the ability to have high drug loading and prolonged stability 
with lipophilic compounds. In addition to drug delivery, SLNs have been used 
in dermatological and cosmetic preparations. The most common carriers used to 

prepare SLNs are triglycerides, glycerides, fatty acids, steroids, and waxes and may 
contain a wide range of emulsifi ers. Methods of preparation of SLNs include high 
speed hot and cold homogenization, ultrasound, emulsion – solvent evaporation, and 
microemulsion. The various parameters involved in the preparation of SLNs have 
been optimized and thoroughly reviewed and their physicochemical properties 
elucidated [103, 104] . 
Other Synthetic Biodegradable Polymers Although well investigated for drug 
delivery, polyorthoesters, polyurethanes, and polyamides have found limited application 
as nanoparticles. A report documents the synthesis and characterization of 
polyorthoester nanoparticles [105] . 
5.4.3.2 Drug Delivery Applications and Biological Fate of Synthetic 
Biodegradable Polymers 
PLA / PLGA Nanoparticles A wide range of hydrophilic and hydrophobic drugs, 
including low - and high - molecular - weight compounds, have been encapsulated into 
PLGA/PLA nanoparticles for a wide range of therapeutic applications and routes 
of administration, including oral, intravenous, intra - arterial, nasal, and inhalation 
delivery [92, 106] . Extensive reviews describing the application of PLGA nanoparticles 
in drug therapy are available [92, 107, 108] . 
After i.v. administration, the PLGA nanoparticles are removed from systemic 
circulation by the mononuclear phagocytic system in the liver [109] . PLGA nanoparticles 
enter cells by absorptive endocytosis and may escape the lysosomes to accumulate 
in cytoplasm [110, 111] . In the body, PLA and PLGA degrade into the 
monomers lactic and glycolic acids, which enter the citric acid cycle, where they are 
metabolized and eliminated as CO 2 and H 2 O. Glycolic acid may also be excreted 
through the kidney [91] . Humoral response to these results in mild, acute, and 
chronic infl ammation [112] . 
Poly( e-caprolactone) Nanoparticles As important applications of poly( . - 
caprolactone) nanoparticles have been reviewed previously, only representative 
examples will be given [94] . Decreased cardiovascular adverse effects of cartelol 
was observed upon ophthalmic administration of poly( . - caprolactone) nanocapsules 
[113] . Poly( . - caprolactone) nanoparticles, nanocapsules, or nanoemulsions 
increased the ocular uptake of indomethacin [114] . The cytotoxicity of retinoic acid 
was enhanced when delivered in core - shell - type nanoparticles formed from poly( . - 
caprolactone – polyethylene glycol) blends [115] . Alternately, these nanoparticles 
were also chemically modifi ed with folic acid to target the folate receptors for 
enhanced cellular uptake [116] . Coating poly( . - caprolactone) nanoparticles with 
polysaccharides such as galactose resulted in lectin - dependent aggregation, demonstrating 
the potential as a targeted delivery system to hepatocytes [117] . Stable 
complexes were formed between anionic DNA and chitosan - modifi ed poly( . - 
caprolactone) nanoparticles, demonstrating high transfection effi ciency [118] . After 
i.v. administration, these particles are eliminated by macrophages of the reticuloendothelial 
system and biodegradation occurs by bulk scission of polymer chains 
[94] . However, dextran - coated poly( . - caprolactone) nanoparticles lowered their 
uptake into macrophages [119] . 
SYNTHETIC BIODEGRADABLE POLYMERIC NANOPARTICLES 545

546 BIODEGRADABLE NANOPARTICLES 
Polyanhydride Nanoparticles Polyanhydrides have been more commonly used 
to prepare microparticles than nanoparticles. However, the technology is adaptable 
for nanoparticles. The transfection effi ciency of fi refl y luciferase DNA was 
enhanced when delivered in nanoparticles prepared from polyanhydride – lactic 
acid blends, demonstrating the potential application in gene delivery [120] . The 
degradation and elimination of polyanhydrides have been reviewed [97] . In vivo, 
the anhydride bond degrades to form diacid monomers that are eliminated from 
the body. 
Poly(alkyl -cyanoacrylate) Nanoparticles The applications of poly(alkyl - 
cyanoacrylate) nanoparticles have been reviewed elsewhere and therefore only 
representative examples are described [102] . Because of their adhesive properties, 
nanoparticles have the potential to prophylactically treat candidiasis of the oral 
cavity [121] . Not surprisingly, poly(alkyl - cyanoacrylate) nanoparticles have been 
used to deliver drugs to tumors [122] . Enhanced absorption and prolonged 
hypoglycemic effect were observed when insulin was delivered in poly(alkyl - 
cyanoacrylate) nanoparticles [121] . Nuclear accumulation of antisense oligonucleotides 
into vascular smooth muscle cells was increased when delivered using 
poly(alkyl - cyanoacrylate) nanoparticles [123] . Dextran - coated poly(alkyl - 
cyanoacrylate) nanoparticles lowered protein adsorption in the blood [124] . 
Poly(alkyl - cyanoacrylates) degrade by hydrolysis of the ester bond of the alkyl 
side chains to form water - soluble alkyl alcohol and poly(cyanoacrylic acid). After 
in vivo administration, poly(alkyl - cyanoacrylate) nanoparticles are predominantly 
distributed in the liver, spleen, and bone marrow where they are endocytosed into 
the cells to become localized in the lysosomes. However, the mechanisms of lysosomal 
escape have not been identifi ed [102] . 
Solid–Lipid Nanoparticles SLNs have been used to deliver small molecules and 
macromolecules such as DNA and peptides. The in vitro and in vivo applications of 
SLNs are reviewed elsewhere [125, 126] . The stability and oral bioavailability of 
insulin were enhanced when administered in wheat germ agglutinin – conjugated 
nanoparticles [127] . A polyoxyethylene stearate coat on the SLN confers stealth 
properties [128] . 
5.4.4 THERMOSENSITIVE AND p H - SENSITIVE NANOPARTICLES 
5.4.4.1 Physical Properties and Methods of Preparation 
Thermosensitive Polymeric Nanoparticles Thermoresponsive “ smart ” polymers 
that change their physical characteristics, such as shape, surface properties, or solubility, 
in response to changes in temperature have been developed to target drugs 
[129] . The encapsulated drug is released when the nanoparticles are exposed to 
changes in temperature, such as body temperature or external heat source. For 
example, poly( N - isopropyl acrylamide), or poly(NIPAAm), is water soluble at room 
temperature but aggregates and is insoluble above its lower critical solution temperature 
(LCST), which typically ranges between 37 and 42 ° C [129] . The methods 
used to prepare thermosensitive nanoparticles have been thoroughly reviewed [127, 

129 – 131] . Interestingly, when heated above the LCST, poly(NIPAAm) – PEG block 
copolymers spontaneously self - assemble into nanoparticles whose size is controlled 
by the rate of heating [132] . 
pH-Sensitive Polymeric Nanoparticles Enteric - coated polymers have long been 
used to protect drugs from the acidic pH in the stomach. Chitosan is a pH - sensitive 
polymer that is soluble only in acidic media. Similarly, the solubility of sulfonamide - 
modifi ed pullulans is dependent on pH [133] . Chitosan – insulin nanoparticles are 
stable at low pH but dissociate at physiological pH, releasing insulin [134] . The pH 
of tumor interstitium is lower than the normal tissue. The pH - sensitive polyethylene 
oxide – poly( . - amino ester) microparticles containing paclitaxel signifi cantly reduced 
tumor burden [135, 136] . Polyketals, a new generation of acid - sensitive polymers, 
degrade by acidic hydrolysis [137] . Low pH inside the endosomes facilitated the 
escape of pH - responsive plasmid – lipid nanoparticles, resulting in enhanced transfection 
effi ciency [138] . 
5.4.4.2 Drug Delivery Applications and Biological Fate of Thermosensitive and 
p H - Sensitive Nanoparticles 
Thermosensitive block copolymer nanoparticles containing doxorubicin increased 
cytotoxicity against Lewis lung carcinoma cells when activated by heating above 
the LCST [139] . Chitosan was chemically conjugated to NIPAAm/vinyl laurate 
copolymer to enhance gene transfection in mouse myoblast cells [140] . Upon i.v. 
administration, poly(NIPAAm) nanoparticles are taken up by the reticuloendothelial 
cells of the liver and mild infl ammatory and fi brotic responses are observed 
[141] . 
After internalization into SKOV - 3 (ovarian adenocarcinoma) cells, polyethylene 
oxide – modifi ed poly( . - amino ester) nanoparticles rapidly disintegrated and released 
the drug in the low pH of the endosomes [142] . Intravenous administration of polyethylene 
oxide – modifi ed poly( . - amino ester) nanoparticles containing paclitaxel 
signifi cantly reduced tumor burden in mice with ovarian cancer [142] . N - Acetyl 
histidine – conjugated glycol chitosan nanoparticles were used to deliver drugs into 
the cytoplasm. These pH - responsive nanoparticles are endocytosed where their 
structural integrity is lost due to protonation of imidazoles, resulting in their endo - 
lysosomal escape [57] . As most of the pH - sensitive biodegradable polymers are 
blends of natural and synthetic polymers, they are degraded by mechanisms specifi c 
to individual polymers. 
5.4.5 APPLICATIONS OF BIODEGRADABLE NANOPARTICLES 
OTHER THAN DRUG DELIVERY 
Diagnosis and imaging are important applications of nanoparticles that are briefl y 
described. The ability to encapsulate or conjugate fl uorescent compounds into or 
onto biodegradable nanoparticles has been used extensively in imaging. Compounds 
that have been encapsulated into nanoparticles for imaging include gadolinium, 
fl uorescein isothiocyanate (FITC) – dextrans, Bodipy, and the autofl uorescent anticancer 
drug doxorubicin. Nanoparticles encapsulating radioactive ligands, such as 
APPLICATIONS OF BIODEGRADABLE NANOPARTICLES 547

548 BIODEGRADABLE NANOPARTICLES 
99m Tc - labeled colloids and 111 In, have been used in scintigraphic imaging [143] . In 
addition, fl uorescent or radioactive moieties can be targeted by noncovalently or 
covalently tagging the nanoparticles through avidin – biotin conjugation and thiol 
formation [144] . In vitro imaging enables the dynamics of cellular internalization and 
localization of nanoparticles to be studied. For example, Bodipy — loaded PLGA 
nanoparticles have been used to study their cellular disposition in vitro [145] as well 
as the effect of storage temperature on their physical properties [146] . The biotinylated 
antibody, specifi c to the CD3 antigen on lymphocytes, was chemically conjugated 
to nanoparticles and their binding to leukemic and primary T lymphocytes investigated 
[143] . The instrumentation that facilitates imaging of nanoparticles includes 
confocal laser scanning microscopy, liquid scintigraphy, and fl ow cytometry. 
5.4.6 PHYSICOCHEMICAL CHARACTERIZATION OF 
POLYMERIC NANOPARTICLES 
The selection of polymer is critical to the performance, properties, and application 
of nanoparticles. Further, the physicochemical properties of the polymer will determine 
the surface properties of nanoparticles with polymer molecular weight, hydrophobicity, 
and glass transition temperature being particularly important. The surface 
properties that infl uence their biodistribution and cellular response include particle 
size, zeta potential, and surface hydrophilicity. 
5.4.6.1 Molecular Weight 
Many reviews and textbooks document the experimental methods available to 
determine number - average, weight - average, viscosity - average, and z - average molecular 
weights. The molecular weight of the polymer will infl uence many parameters 
during the preparation of nanoparticles as well as their properties such as drug 
loading and rate and extent of drug release. 
5.4.6.2 Hydrophobicity 
The experimental methods of determining hydrophobicity include interaction chromatography 
and two - phase partition using fl uorescent or radiolabeled hydrophobic 
probes [147] . Generally, a more hydrophobic polymer degrades more slowly and 
releases drugs at a decreased rate. Hence, a blend of hydrophobic and hydrophilic 
polymers can be used to tailor drug release kinetics. Depending on the conditions 
of polymerization, mixtures of two or more monomers of different types yield block, 
alternate, or random cocopolymers, each of which will possess different hydrophobicities 
and consequently drug release kinetics. Additionally, the degree of hydrophobicity 
can change the mechanism of degradation. For example, by inserting 
hydrophilic ethylene glycol moieties within the hydrophobic backbone, polyanhydrides 
have been tailored to specifi cally degrade by bulk or surface erosion [148] . 
Incorporation of aromatic side chains generally increases hydrophobicity. A difference 
in hydrophobicity of individual polymers results in microphase separation of 
copolymers followed by a thermodynamic partition and altered release profi le of 
encapsulated drugs [149] . 

5.4.6.3 Glass Transition Temperature 
The morphology and physical properties of nanoparticles are affected by the glass 
transition temperature and physical state of the polymer or polymer blends. The 
glass transition temperature ( Tg ) is the temperature at which polymers undergo 
a change in heat capacity and transform their physical arrangement. The crystallinity 
or amorphous nature of the polymers can be altered by synthesizing polymer 
blends of varying ratios. The Tg of a polymer is experimentally determined by 
differential scanning calorimetry (DSC). The Tg is mathematically calculated using 
multidimensional lattice representations and statistical methods [150] . The various 
factors that infl uence the Tg of polymers include molecular weight, composition 
and stereochemistry of the polymer backbone, type and length of pendent 
groups, additives such as copolymers, and plasticizers. Additives and copolymers 
may also be used to alter the Tg . For example, incorporation of mPEG signifi - 
cantly lowered the Tg of PLA, resulting in rapid release of drug from the nanoparticles 
[151] . 
5.4.6.4 Particle Size and Particle Size Distribution Instrumentation 
Particle size is a critical characteristic of nanoparticles and by defi nition differentiates 
them from microparticles. Particle size and particle size distribution play an 
important role in the biological performance of the nanoparticles. Important techniques 
for measuring particle size are photon correlation spectroscopy (PCS), electron 
microscopy, and atomic force microscopy, which have been comprehensively 
described [147] . Particle size and particle size distribution are determined by the 
method of preparation and experimental variables during manufacture. For example, 
in the emulsion – solvent evaporation method, the particle size is determined by 
controlling the energy of emulsifi cation and the resulting droplet size of the internal 
phase. Control of particle size is also possible by altering experimental variables 
such as the volume and phase ratio of the internal and external phases and the 
concentration, type, and viscosity of the emulsifying agent. As an example, the 
experimental parameters that can infl uence the particle size and size distribution of 
PLGA nanoparticles include the method used, polymer concentration, surfactant 
concentration, stirring speed, ratio of aqueous and organic phases, and concentration 
of the emulsifi er [152] . Methods of separation such as fi ltration and centrifugation 
can also infl uence particle size distribution. When chitosan nanoparticles are 
prepared using ionic gelation, the critical parameters for a narrow particle size distribution 
are molecular weight, degree of deacetylation, concentration and molar 
ratio of chitosan, and the presence and concentrations of counterions (e.g., tripolyphosphate) 
[12] . 
5.4.6.5 Surface Charge and Zeta Potential 
The surface charge of nanoparticles is important because it determines the nature 
and extent of aggregation of colloids and their interaction with cells and other biological 
components within the body. The zeta potential is the potential at the solid – 
liquid interface and is commonly determined using light scattering [153] . Decreasing 
the zeta potential of nanoparticles below a critical value increases the rate and 
PHYSICOCHEMICAL CHARACTERIZATION OF POLYMERIC NANOPARTICLES 549

550 BIODEGRADABLE NANOPARTICLES 
extent of their aggregation, resulting in conglomerates with different physical properties. 
The surface charge can also be altered by chemical conjugation of ligands to 
nanoparticles. As the following examples demonstrate, surface charge may be 
modifi ed to facilitate targeting. The degree of positive charge on chitosan – tripolyphosphate 
nanoparticles was increased by controlling the processing parameters, 
resulting in a stronger electrostatic interaction with negatively charged cell surfaces 
[13] . An increase in surface charge increased the adsorption of nanoparticles to 
plasma proteins [154] . Positively charged ligands such as chitosan as well as peptides 
neutralize the surface charge of negatively charged particles made using polymers 
such as PLGA [155] . Positively charged tripalmitin nanoparticles increased circulation 
time and higher blood concentrations of etoposide compared to negatively 
charged particles [156] . On the other hand, neutrally charged particles may be protected 
from opsonization [157] . 
5.4.6.6 Surface Hydrophilicity 
The surface hydrophilicity of nanoparticles also infl uences the nature and extent 
of their interaction with cells and their behavior in the biological environment. The 
techniques for determining surface hydrophilicity have been described previously 
[147] . As hydrophobic nanoparticles are opsonized and eliminated by the mononuclear 
phagocytic system, surface hydrophilicity is an important parameter to 
ensure longer circulation times. The hydrophilicity to the nanoparticles may be 
modifi ed using several methods, including adsorption of nonionic surfactants 
such as Poloxamer as well as adsorption or conjugation of hydrophilic polymers 
such as polysaccharides, polyacrylamides, PVA, poly( N - vinyl - 2 - pyrrolidone), PEG, 
polyoxamines, and polysorbates [157] . Stealth nanoparticles can be prepared by 
adsorbing or conjugating PEG to the particle surface, which protects them from 
opsonization [157] . The biological properties of stealth nanoparticles have been 
reviewed elsewhere [158] . 
5.4.6.7 Drug Loading and Encapsulation Effi ciency 
Drug loading or the weight of the drug encapsulated in the polymeric carrier is 
expressed as a percentage (w/w) of the delivery system. Encapsulation effi ciency is 
the difference between the amount of the drug encapsulated into nanoparticles 
compared to the total amount added during preparation. The encapsulation effi - 
ciency is dependent on the properties of the polymer and excipients and the method 
of preparation of nanoparticles. For example, increased miscibility between the 
nanoparticle and water at lower temperatures and high rate of solvent evaporation 
at high temperatures result in the formation of an outer sphere wall, increasing 
encapsulation effi ciency [159] . Effi cient encapsulation depends on (1) physicochemical 
properties of the drug, such as solubility, hydrophilicity, and crystallinity; 
(2) physicochemical properties of the polymer, including molecular weight, hydrophobicity, 
drug – polymer interactions, and solubility parameter; and (3) variables 
involved in the preparation of the nanoparticles, such as drug – polymer weight ratio, 
solvents, method of preparation of particles, solvent evaporation rate, type as well 
as volume, and concentration of the surface - active agent used. 

5.4.6.8 Drug Release 
Many cumulative and differential dissolution methods are used to monitor the rate 
and extent of drug release. The rate of drug release from nanoparticles depends on 
the chemical properties of the polymer, properties of particles such as hydrophobicity 
and surface area, and environmental factors such as pH. Drug release may occur 
by one or a combination of the following: diffusion, dissolution, degradation, or 
swelling. Hence, drug release normally follows fi rst - rather than zero - order kinetics 
[160 – 162] . A disadvantage of nanoparticles is that they may have a signifi cant burst 
release ( . 40%) due to their high surface area – mass ratio. Surface treatment of 
nanoparticles can reduce the burst release of encapsulated drug [163, 164] . 
5.4.6.9 Physical Stability of Polymeric Nanoparticles 
The main indication of physical instability of nanoparticles is irreversible aggregation. 
The principal factors that affect the extent of aggregation are type of polymer, 
zeta potential, duration and temperature of storage, and presence of electrolytes. 
Generally, homopolymers such as PLA and poly( . - caprolactone) are more stable 
than copolymers. However, the stability of PLGA copolymers can be prolonged 
by appropriate storage [165] . The duration of storage can also be important. For 
example, poly( . - caprolactone) nanoparticles aggregated after four months [166] . 
Another study compared the physical stability of PLGA nano - and microspheres 
after incubation at different temperatures and demonstrated that, while microspheres 
did not aggregate, the extent of aggregation of nanospheres increased as 
storage temperature increased [146] . Ideally, PLGA nanoparticles should be stored 
desiccated at 4 ° C. Aqueous dispersions of SLN are stable for three years [167] . It 
is important to note that electrolytes can accelerate the aggregation and instability 
of nanoparticles. For example, multivalent ions caused solid – lipid nanoparticles to 
gel [168] . Physical stability can be improved by coating the surface of nanoparticles 
with hydrophilic polymers or surfactants [169] . As discussed previously, a reduction 
in the zeta potential below critical values causes fl occulation of colloidal systems. 
For example, neutralization of the surface charge of colloidal systems by addition 
of cationic oligonucleotides resulted in aggregation [170] . 
5.4.7 TARGETING NANOPARTICLES BY SURFACE CONJUGATION 
WITH LIGANDS 
Nanoparticles can be targeted to cells using specifi c or nonspecifi c ligands. Specifi c 
ligands can be covalently conjugated to the nanoparticles to target them to a 
selected site of action. Some examples of targeting ligands are folic acid [171] , 
transferrin [172] , lectin [173] , and epidermal growth factor [174] . Nonspecifi c targeting 
is achieved by attaching ligands that alter the biodistribution of the nanoparticles. 
Examples of nonspecifi c targeting include the use of PEG for imparting stealth 
properties or hydrolytically cleavable linkers that protect the drug from degradation. 
Common methods of conjugation of ligands include carbodiimide coupling, 
glutaraldehyde conjugation, peptide bond formation, disulfi de, and thiol linkages 
[175 – 178] . 
TARGETING NANOPARTICLES BY SURFACE CONJUGATION WITH LIGANDS 551

552 BIODEGRADABLE NANOPARTICLES 
5.4.8 CELLULAR TRAFFICKING OF 
BIODEGRADABLE NANOPARTICLES 
Biodegradable nanoparticles are internalized by one or more of the following 
mechanisms: phagocytosis, macropinocytosis, and clathrin - and caveolin - mediated 
endocytosis (Figure 1 ). While phagocytosis by macrophages eliminates nanoparticles 
from the body, effi cient cellular uptake occurs when high - affi nity receptors 
capture the nanoparticles through receptor - mediated endocytosis [179] . On the cell 
surface, nanoparticles activate caveolin, a dimeric protein, resulting in their internalization 
through caveolae. Clathrin - mediated endocytosis occurs when nanoparticles 
accumulate on the plasma membrane and clathrin - coated pits are formed to 
transport the nanoparticles into the cell, resulting in the formation of endosomes. 
Macropinocytosis is restricted to larger particles, such as nanoparticles typically 
greater than 800 nm. The mechanism by which particles enter cells depends on the 
composition of the nanoparticles, type of the cell, and particle size. For example, the 
uptake of chitosan nanoparticles into lung epithelial and Caco - 2 cells was mediated, 
in part, by the clathrin - mediated pathway [180, 181] . However, PLGA nanoparticles 
of size 100 nm are internalized by the clathrin - and caveolin - independent pathway 
[110] . There are comprehensive reviews of the mechanisms responsible for the 
uptake of nanoparticles [182 – 184] . 
Nanoparticles that are internalized into cells by these mechanisms fi rst enter the 
primary endosomes of the cell and are then transported into sorting endosomes. 
While some nanoparticles in the sorting endosomes are transported out of the cell 
by recycling endosomes, the remaining nanoparticles are transported into secondary 
endosomes that fuse with the lysosomes [107] . The surface charge of PLGA nanoparticles 
is reversed in the acidic lysosome, resulting in their escape into the cytoplasm 
[111] . A high external concentration of nanoparticles outside the cell prolongs their 
intracellular concentration within the cytoplasm [107] . 
Ligand - mediated endocytosis targets specifi c cell surface receptors and these 
nanoparticles are internalized by a receptor - mediated endocytic pathway. Examples 
of targeting transferrin, folate, lectins, and epidermal growth factor receptors are 
contained in the literature [185 – 187] . 
FIGURE 1 Mechanisms of cellular internalization of biodegradable nanoparticles. 
Mechanisms of 
internalization 
of 
nanoparticles 
Phagocytosis 
Pinocytosis/ 
fluid-phase 
endocytosis 
Receptormediated 
endocytosis 
Caveolinmediated 
endocytosis 
Clathrin- & 
caveolinindependent 
endocytosis 
Macropinocytosis 
Clathrinmediated 
endocytosis 
Transferrin, 
folate, 
asialoglycoprotein, 
Epidermal growth 
factor, etc.

5.4.9 CONCLUSIONS 
Biodegradable nanoparticles are a very active area of research in drug delivery, 
imaging, and diagnostics. This review has primarily focused on drug delivery applications. 
Natural and synthetic polymers used to prepare nanoparticles were discussed 
as well as their physicochemical properties that infl uence the biological performance 
of particles. Methods used to prepare and characterize the properties of 
nanoparticles have also been reviewed. Specifi c and nonspecifi c methods used to 
target nanoparticles to cells were mentioned as well as the mechanism for cellular 
and intracellular transport. As research into the various uses of biodegradable 
nanoparticles increases, so will our knowledge to further optimize their preparation 
and formulation and hence improve drug therapy and diagnosis. 
REFERENCES 
1. Dang , J. M. , and Leong , K. W. ( 2006 ), Natural polymers for gene delivery and tissue 
engineering , Adv. Drug Deliv. Rev. , 58 ( 4 ), 487 – 499 . 
2. Singla , A. K. , and Chawla , M. ( 2001 ), Chitosan: Some pharmaceutical and biological 
aspects — an update , J. Pharm. Pharmacol. , 53 ( 8 ), 1047 – 1067 . 
3. Kulkarni , A. R. , Lin , Y. H. , Liang , H. F. , Chang , W. C. , Hsiao , W. W. , and Sung , H. W. 
( 2006 ), A novel method for the preparation of nanoaggregates of methoxy polyethyleneglycol 
linked chitosan , J. Nanosci. Nanotechnol. , 6 ( 9 – 10 ), 2867 – 2873 . 
4. Sanchez - Machado , D. I. , Lopez - Cervantes , J. , Lopez - Hernandez , J. , Paseiro - Losada , P. , 
and Simal - Lozano , J. ( 2004 ), Determination of the uronic acid composition of seaweed 
dietary fi bre by HPLC , Biomed. Chromatogr. , 18 ( 2 ), 90 – 97 . 
5. Dusseault , J. , Tam , S. K. , Menard , M. , Polizu , S. , Jourdan , G. , Yahia , L. , and Halle , J. P. 
( 2006 ), Evaluation of alginate purifi cation methods: Effect on polyphenol, endotoxin, 
and protein contamination , J. Biomed. Mater. Res. A. , 76 ( 2 ), 243 – 251 . 
6. Tonnesen , H. H. , and Karlsen , J. ( 2002 ), Alginate in drug delivery systems , Drug Dev. 
Ind. Pharm. , 28 ( 6 ), 621 – 630 . 
7. Rajaonarivony , M. , Vauthier , C. , Couarraze , G. , Puisieux , F. , and Couvreur , P. ( 1993 ), 
Development of a new drug carrier made from alginate , J. Pharm. Sci. , 82 ( 9 ), 912 – 917 . 
8. Poncelet , D. , Lencki , R. , Beaulieu , C. , Halle , J. P. , Neufeld , R. J. , and Fournier , A. ( 1992 ), 
Production of alginate beads by emulsifi cation/internal gelation. I. Methodology , Appl. 
Microbiol. Biotechnol. , 38 ( 1 ), 39 – 45 . 
9. Reis , C. P. , Neufeld , R. J. , Vilela , S. , Ribeiro , A. J. , and Veiga , F. ( 2006 ), Review and current 
status of emulsion/dispersion technology using an internal gelation process for the 
design of alginate particles , J. Microencapsul. , 23 ( 3 ), 245 – 257 . 
10. Agnihotri , S. A. , Mallikarjuna , N. N. , and Aminabhavi , T. M. ( 2004 ), Recent advances on 
chitosan - based micro - and nanoparticles in drug delivery , J. Controlled Release , 100 ( 1 ), 
5 – 28 . 
11. Bodnar , M. , Hartmann , J. F. , and Borbely , J. ( 2005 ), Preparation and characterization of 
chitosan - based nanoparticles , Biomacromolecules , 6 ( 5 ), 2521 – 2527 . 
12. Zhang , H. , Oh , M. , Allen , C. , and Kumacheva , E. ( 2004 ), Monodisperse chitosan nanoparticles 
for mucosal drug delivery , Biomacromolecules , 5 ( 6 ), 2461 – 2468 . 
13. Gan , Q. , Wang , T. , Cochrane , C. , and McCarron , P. ( 2005 ), Modulation of surface charge, 
particle size and morphological properties of chitosan - TPP nanoparticles intended for 
gene delivery , Colloids Surf. B. Biointerfaces , 44 ( 2 – 3 ), 65 – 73 . 
REFERENCES 553

554 BIODEGRADABLE NANOPARTICLES 
14. Lopez - Leon , T. , Carvalho , E. L. , Seijo , B. , Ortega - Vinuesa , J. L. , and Bastos - Gonzalez , 
D. ( 2005 ), Physicochemical characterization of chitosan nanoparticles: Electrokinetic 
and stability behavior , J. Colloid Interface Sci. , 283 ( 2 ), 344 – 351 . 
15. Young , S. , Wong , M. , Tabata , Y. , and Mikos , A. G. ( 2005 ), Gelatin as a delivery vehicle 
for the controlled release of bioactive molecules , J. Controlled Release , 109 ( 1 – 3 ), 
256 – 274 . 
16. Azarmi , S. , Huang , Y. , Chen , H. , McQuarrie , S. , Abrams , D. , Roa , W. , Finlay , W. H. , Miller , 
G. G. , and Lobenberg , R. ( 2006 ), Optimization of a two - step desolvation method for 
preparing gelatin nanoparticles and cell uptake studies in 143B osteosarcoma cancer 
cells , J. Pharm. Pharmacal. Sci. , 9 ( 1 ), 124 – 132 . 
17. Lee , C. H. , Singla , A. , and Lee , Y. ( 2001 ), Biomedical applications of collagen , Int. J. 
Pharm. , 221 ( 1 – 2 ), 1 – 22 . 
18. Sihorkar , V. , and Vyas , S. P. ( 2001 ), Potential of polysaccharide anchored liposomes in 
drug delivery, targeting and immunization , J. Pharm. Pharmacal. Sci. , 4 ( 2 ), 138 – 158 . 
19. Jeong , Y. I. , Na , H. S. , Oh , J. S. , Choi , K. C. , Song , C. E. , and Lee , H. C. ( 2006 ), Adriamycin 
release from self - assembling nanospheres of poly( dl - lactide - co - glycolide) - grafted pullulan 
, Int. J. Pharm. , 322 ( 1 – 2 ), 154 – 160 . 
20. Leathers , T. D. ( 2005 ), in Steinbuchel , A. R. , (Ed.), Polysaccharides and Polyamides in 
the Food Industry. Properties, Production, and Patents , Vol. 1, Wiley - VCH , Weinheim , pp. 
387 – 421 . 
21. Garach , V. ( 1975 ), The synthesis of dextrans and levans — a review of the literature , 
Diastema , 4 ( 3 ), 25 – 28 . 
22. Mehvar , R. ( 2000 ), Dextrans for targeted and sustained delivery of therapeutic and 
imaging agents , J. Controlled Release , 69 ( 1 ), 1 – 25 . 
23. Tizard , I. R. , Carpenter , R. H. , McAnalley , B. H. , and Kemp , M. C. ( 1989 ), The 
biological activities of mannans and related complex carbohydrates , Mol. Biother. , 1 ( 6 ), 
290 – 296 . 
24. Hasegawa , U. , Nomura , S. M. , Kaul , S. C. , Hirano , T. , and Akiyoshi , K. ( 2005 ), Nanogel - 
quantum dot hybrid nanoparticles for live cell imaging , Biochem. Biophys. Res. Commun. , 
331 ( 4 ), 917 – 921 . 
25. Gupta , M. , and Gupta , A. K. ( 2004 ), Hydrogel pullulan nanoparticles encapsulating 
pBUDLacZ plasmid as an effi cient gene delivery carrier , J. Controlled Release , 99 ( 1 ), 
157 – 166 . 
26. Na , K. , Lee , E. S. , and Bae , Y. H. ( 2003 ), Adriamycin loaded pullulan acetate/sulfonamide 
conjugate nanoparticles responding to tumor pH: pH - dependent cell interaction, internalization 
and cytotoxicity in vitro , J. Controlled Release , 87 ( 1 – 3 ), 3 – 13 . 
27. Tiyaboonchai , W. , Woiszwillo , J. , and Middaugh , C. R. ( 2001 ), Formulation and characterization 
of amphotericin B - polyethylenimine - dextran sulfate nanoparticles , J. Pharm. 
Sci. , 90 ( 7 ), 902 – 914 . 
28. Cui , Z. , Hsu , C. H. , and Mumper , R. J. ( 2003 ), Physical characterization and macrophage 
cell uptake of mannan - coated nanoparticles , Drug Dev. Ind. Pharm. , 29 ( 6 ), 689 – 700 . 
29. Quinlan , G. J. , Martin , G. S. , and Evans , T. W. ( 2005 ), Albumin: Biochemical properties 
and therapeutic potential , Hepatology , 41 ( 6 ), 1211 – 1219 . 
30. Verrecchia , T. , Huve , P. , Bazile , D. , Veillard , M. , Spenlehauer , G. , and Couvreur , P. ( 1993 ), 
Adsorption/desorption of human serum albumin at the surface of poly(lactic acid) 
nanoparticles prepared by a solvent evaporation process , J. Biomed. Mater. Res. , 27 ( 8 ), 
1019 – 1028 . 
31. Bousquet , Y. , Swart , P. J. , Schmitt - Colin , N. , Velge - Roussel , F. , Kuipers , M. E. , Meijer , D. 
K. , Bru , N. , Hoebeke , J. , and Breton , P. ( 1999 ), Molecular mechanisms of the adsorption 

of a model protein (human serum albumin) on poly(methylidene malonate 2.1.2) 
nanoparticles , Pharm. Res. , 16 ( 1 ), 141 – 147 . 
32. Lin , W. , Garnett , M. C. , Davis , S. S. , Schacht , E. , Ferruti , P. , and Illum , L. ( 2001 ), Preparation 
and characterisation of rose Bengal - loaded surface - modifi ed albumin nanoparticles , 
J. Controlled Release , 71 ( 1 ), 117 – 126 . 
33. Langer , K. , Balthasar , S. , Vogel , V. , Dinauer , N. , von Briesen , H. , and Schubert , D. ( 2003 ), 
Optimization of the preparation process for human serum albumin (HSA) nanoparticles 
, Int. J. Pharm. , 257 ( 1 – 2 ), 169 – 180 . 
34. Mamone , G. , Addeo , F. , Chianese , L. , Di Luccia , A. , De Martino , A. , Nappo , A. , 
Formisano , A. , De Vivo , P. , and Ferranti , P. ( 2005 ), Characterization of wheat gliadin 
proteins by combined two - dimensional gel electrophoresis and tandem mass spectrometry 
, Proteomics , 5 ( 11 ), 2859 – 2865 . 
35. Ezpeleta , I. , Irache , J. M. , Stainmesse , S. , Chabenat , C. , Gueguen , J. , Popineau , Y. , and 
Orecchioni , A. ( 1996 ), Gliadin nanoparticles for the controlled release of all - trans - 
retinoic acid , Int. J. Pharm. , 131 ( 2 ), 191 – 200 . 
36. Wieser , H. (1996), Relation between gliadin structure and coeliac toxicity , Acta Paediatr. 
Suppl. , 412 , 3 – 9 . 
37. Lambert , G. , Fattal , E. , and Couvreur , P. ( 2001 ), Nanoparticulate systems for the delivery 
of antisense oligonucleotides , Adv. Drug Deliv. Rev. , 47 ( 1 ), 99 – 112 . 
38. Bodmeier , R. , Chen , H. G. , and Paeratakul , O. ( 1989 ), A novel approach to the oral 
delivery of micro - or nanoparticles , Pharm. Res. , 6 ( 5 ), 413 – 417 . 
39. Douglas , K. L. , and Tabrizian , M. ( 2005 ), Effect of experimental parameters on the formation 
of alginate - chitosan nanoparticles and evaluation of their potential application 
as DNA carrier , J. Biomater. Sci. Polym. Ed. , 16 ( 1 ), 43 – 56 . 
40. Borges , O. , Borchard, G. , Verhoef , J. C. , de Sousa, A. , and Junginger , H. E. (2005), Preparation 
of coated nanoparticles for a new mucosal vaccine delivery system , Int. J. Pharm. , 
299 ( 1 – 2 ), 155 – 166 . 
41. Pandey , R. , Ahmad , Z. , Sharma , S. , and Khuller , G. K. ( 2005 ), Nano - encapsulation of 
azole antifungals: Potential applications to improve oral drug delivery , Int. J. Pharm. , 
301 ( 1 – 2 ), 268 – 276 . 
42. Zahoor , A. , Sharma , S. , and Khuller , G. K. ( 2005 ), Inhalable alginate nanoparticles as 
antitubercular drug carriers against experimental tuberculosis , Int. J. Antimicrob. Agents , 
26 ( 4 ), 298 – 303 . 
43. Yi , Y. M. , Yang , T. Y. , and Pan , W. M. ( 1999 ), Preparation and distribution of 5 - 
fl uorouracil (125)I sodium alginate - bovine serum albumin nanoparticles , World J. Gastroenterol. 
5 ( 1 ), 57 – 60 . 
44. Ahmad , Z. , Pandey , R. , Sharma , S. , and Khuller , G. K. ( 2006 ), Pharmacokinetic and 
pharmacodynamic behaviour of antitubercular drugs encapsulated in alginate nanoparticles 
at two doses , Int. J. Antimicrob. Agents , 27 ( 5 ), 409 – 416 . 
45. Borges , O. , Cordeiro - da - Silva , A. , Romeijn , S. G. , Amidi , M. , de Sousa , A. , Borchard , G. , 
and Junginger , H. E. ( 2006 ), Uptake studies in rat Peyer ’ s patches, cytotoxicity and 
release studies of alginate coated chitosan nanoparticles for mucosal vaccination , 
J. Controlled Release , 114 ( 3 ), 348 – 358 . 
46. Holtan , S. , Zhang , Q. , Strand , W. I. , and Skjak - Braek , G. ( 2006 ), Characterization of the 
hydrolysis mechanism of polyalternating alginate in weak acid and assignment of the 
resulting MG - oligosaccharides by NMR spectroscopy and ESI - mass spectrometry , Biomacromolecules 
, 7 ( 7 ), 2108 – 2121 . 
47. Chen , L. , and Subirade , M. ( 2005 ), Chitosan/beta - lactoglobulin core - shell nanoparticles 
as nutraceutical carriers , Biomaterials , 26 ( 30 ), 6041 – 6053 . 
REFERENCES 555

556 BIODEGRADABLE NANOPARTICLES 
48. Qi , L. , Xu , Z. , Jiang , X. , Hu , C. , and Zou , X. ( 2004 ), Preparation and antibacterial activity 
of chitosan nanoparticles , Carbohydr. Res. , 339 ( 16 ), 2693 – 2700 . 
49. Liu , C. G. , Desai , K. G. , Chen , X. G. , and Park , H. J. ( 2005 ), Preparation and characterization 
of nanoparticles containing trypsin based on hydrophobically modifi ed chitosan , 
J. Agric. Food Chem. , 53 ( 5 ), 1728 – 1733 . 
50. Grenha , A. , Seijo , B. , and Remunan - Lopez , C. ( 2005 ), Microencapsulated chitosan 
nanoparticles for lung protein delivery , Eur. J. Pharm. Sci. , 25 ( 4 – 5 ), 427 – 437 . 
51. Enriquez de Salamanca , A. , Diebold , Y. , Calonge , M. , Garcia - Vazquez , C. , Callejo , S. , 
Vila , A. , and Alonso , M. J. ( 2006 ), Chitosan nanoparticles as a potential drug delivery 
system for the ocular surface: Toxicity, uptake mechanism and in vivo tolerance , Invest. 
Ophthalmol. Vis. Sci. , 47 ( 4 ), 1416 – 1425 . 
52. Saito , H. , Wu , X. , Harris , M. , and Hoffman , A. ( 2003 ), Graft copolymers of poly(ethylene 
glycol) (PEG) and chitosan , Macromol. Rapid Commun. , 18 ( 7 ), 547 – 550 . 
53. Bernkop - Schnurch , A. ( 2000 ), Chitosan and its derivatives: Potential excipients for 
peroral peptide delivery systems , Int. J. Pharm. , 194 ( 1 ), 1 – 13 . 
54. Hyung Park , J. , Kwon , S. , Lee , M. , Chung , H. , Kim , J. H. , Kim , Y. S. , Park , R. W. , Kim , I. 
S. , Bong Seo , S. , Kwon , I. C. , and Young Jeong , S. ( 2006 ), Self - assembled nanoparticles 
based on glycol chitosan bearing hydrophobic moieties as carriers for doxorubicin: In 
vivo biodistribution and anti - tumor activity , Biomaterials , 27 ( 1 ), 119 – 126 . 
55. Mansouri , S. , Cuie , Y. , Winnik , F. , Shi , Q. , Lavigne , P. , Benderdour , M. , Beaumont , E. , 
and Fernandes , J. C. ( 2006 ), Characterization of folate - chitosan - DNA nanoparticles for 
gene therapy , Biomaterials , 27 ( 9 ), 2060 – 2065 . 
56. Yan , C. , Chen , D. , Gu , J. , and Qin , J. ( 2006 ), Nanoparticles of 5 - fl uorouracil (5 - FU) 
loaded N - succinyl - chitosan (Suc - Chi) for cancer chemotherapy: Preparation, characterization 
— in - vitro drug release and anti - tumour activity , J. Pharm. Pharmacol. , 58 ( 9 ), 
1177 – 1181 . 
57. Park , J. S. , Han , T. H. , Lee , K. Y. , Han , S. S. , Hwang , J. J. , Moon , D. H. , Kim , S. Y. , and 
Cho , Y. W. ( 2006 ), N - acetyl histidine - conjugated glycol chitosan self - assembled nanoparticles 
for intracytoplasmic delivery of drugs: Endocytosis, exocytosis and drug release , 
J. Controlled Release. , 115 ( 1 ), 37 – 45 . 
58. Vandervoort , J. , and Ludwig , A. ( 2004 ), Preparation and evaluation of drug - loaded 
gelatin nanoparticles for topical ophthalmic use , Eur. J. Pharm. Biopharm. , 57 ( 2 ), 
251 – 261 . 
59. Cascone , M. G. , Lazzeri , L. , Carmignani , C. , and Zhu , Z. ( 2002 ), Gelatin nanoparticles 
produced by a simple W/O emulsion as delivery system for methotrexate , J. Mater. Sci. 
Mater. Med. , 13 ( 5 ), 523 – 526 . 
60. Lu , Z. , Yeh , T. K. , Tsai , M. , Au , J. L. , and Wientjes , M. G. ( 2004 ), Paclitaxel - loaded gelatin 
nanoparticles for intravesical bladder cancer therapy , Clin. Cancer Res. , 10 ( 22 ), 
7677 – 7684 . 
61. Bajpai , A. K. , and Choubey , J. ( 2006 ), Design of gelatin nanoparticles as swelling 
controlled delivery system for chloroquine phosphate , J. Mater. Sci. Mater. Med. , 17 ( 4 ), 
345 – 358 . 
62. Li , J. K. , Wang , N. , and Wu , X. S. ( 1997 ), A novel biodegradable system based on gelatin 
nanoparticles and poly(lactic -co - glycolic acid) microspheres for protein and peptide 
drug delivery , J. Pharm. Sci. , 86 ( 8 ), 891 – 895 . 
63. Farrugia , C. A. , and Groves , M. J. ( 1999 ), The activity of unloaded gelatin nanoparticles 
on murine melanoma B16 - F0 growth in vivo , Anticancer Res. , 19 ( 2A ), 1027 – 1031 . 
64. Coester , C. , Kreuter , J. , von Briesen , H. , and Langer , K. ( 2000 ), Preparation of avidin - 
labelled gelatin nanoparticles as carriers for biotinylated peptide nucleic acid (PNA) , 
Int. J. Pharm. , 196 ( 2 ), 147 – 149 . 

65. Kaul , G. , and Amiji , M. ( 2002 ), Long - circulating poly(ethylene glycol) - modifi ed gelatin 
nanoparticles for intracellular delivery , Pharm. Res. , 19 ( 7 ), 1061 – 1067 . 
66. Kommareddy , S. , and Amiji , M. ( 2005 ), Preparation and evaluation of thiol - modifi ed 
gelatin nanoparticles for intracellular DNA delivery in response to glutathione , Bioconjug. 
Chem. , 16 ( 6 ), 1423 – 1432 . 
67. Coester , C. J. , Langer , K. , van Briesen , H. , and Kreuter , J. ( 2000 ), Gelatin nanoparticles 
by two step desolvation — a new preparation method, surface modifi cations and cell 
uptake , J. Microencapsul. , 17 ( 2 ), 187 – 193 . 
68. Kaul , G. , and Amiji , M. ( 2004 ), Biodistribution and targeting potential of poly(ethylene 
glycol) - modifi ed gelatin nanoparticles in subcutaneous murine tumor model , J. Drug 
Target. , 12 ( 9 – 10 ), 585 – 591 . 
69. Coester , C. , Nayyar , P. , and Samuel , J. ( 2006 ), In vitro uptake of gelatin nanoparticles by 
murine dendritic cells and their intracellular localisation , Eur. J. Pharm. Biopharm. , 
62 ( 3 ), 306 – 314 . 
70. Leo , E. , Arletti , R. , Forni , F. , and Cameroni , R. ( 1997 ), General and cardiac toxicity of 
doxorubicin - loaded gelatin nanoparticles , Farmaco , 52 ( 6 – 7 ), 385 – 388 . 
71. Gu , X. G. , Schmitt , M. , Hiasa , A. , Nagata , Y. , Ikeda , H. , Sasaki , Y. , Akiyoshi , K. , 
Sunamoto , J. , Nakamura , H. , Kuribayashi , K. , and Shiku, H. (1998), A novel hydrophobized 
polysaccharide/oncoprotein complex vaccine induces in vitro and in vivo cellular 
and humoral immune responses against HER2 - expressing murine sarcomas , Cancer 
Res. , 58 ( 15 ), 3385 – 3390 . 
72. Shiku , H. , Wang , L. , Ikuta , Y. , Okugawa , T. , Schmitt , M. , Gu , X. , Akiyoshi , K. , Sunamoto , 
J. , and Nakamura , H. ( 2000 ), Development of a cancer vaccine: Peptides, proteins, and 
DNA , Cancer Chemother. Pharmacol. , 46 (Suppl), S77 – 82 . 
73. Na , K. , and Bae , Y. H. ( 2002 ), Self - assembled hydrogel nanoparticles responsive to tumor 
extracellular pH from pullulan derivative/sulfonamide conjugate: Characterization, 
aggregation, and adriamycin release in vitro , Pharm. Res. , 19 ( 5 ), 681 – 688 . 
74. Gupta , A. K. , and Gupta , M. ( 2005 ), Cytotoxicity suppression and cellular uptake 
enhancement of surface modifi ed magnetic nanoparticles , Biomaterials , 26 ( 13 ), 
1565 – 1573 . 
75. Tiyaboonchai , W. , Woiszwillo , J. , Sims , R. C. , and Middaugh , C. R. ( 2003 ), Insulin containing 
polyethylenimine - dextran sulfate nanoparticles , Int. J. Pharm. , 255 ( 1 – 2 ), 139 – 151 . 
76. Cascone , M. G. , Pot , P. M. , Lazzeri , L. , and Zhu , Z. ( 2002 ), Release of dexamethasone 
from PLGA nanoparticles entrapped into dextran/poly(vinyl alcohol) hydrogels , 
J. Mater. Sci. Mater. Med. , 13 ( 3 ), 265 – 269 . 
77. Jaulin , N. , Appel , M. , Passirani , C. , Barratt , G. , and Labarre , D. ( 2000 ), Reduction of the 
uptake by a macrophagic cell line of nanoparticles bearing heparin or dextran covalently 
bound to poly(methyl methacrylate) , J. Drug Target. , 8 ( 3 ), 165 – 172 . 
78. Segura , S. , Gamazo , C. , Irache , J. M. , and Espuelas , S. ( 2007 ), Interferon - . loaded onto 
albumin nanoparticles: In vitro and in vivo activity against brucella abortus, Antimicrob. 
Agents Chemother. , 51 ( 4 ), 1310 – 1314 . 
79. Irache , J. M. , Merodio , M. , Arnedo , A. , Camapanero , M. A. , Mirshahi , M. , and Espuelas , 
S. ( 2005 ), Albumin nanoparticles for the intravitreal delivery of anticytomegaloviral 
drugs , Mini. Rev. Med. Chem. , 5 ( 3 ), 293 – 305 . 
80. Landry , F. B. , Bazile , D. V. , Spenlehauer , G. , Veillard , M. , and Kreuter , J. ( 1998 ), Peroral 
administration of 14C - poly( d , l - lactic acid) nanoparticles coated with human serum 
albumin or polyvinyl alcohol to guinea pigs , J. Drug Target. , 6 ( 4 ), 293 – 307 . 
81. Damascelli , B. , Patelli , G. L. , Lanocita , R. , Di Tolla , G. , Frigerio , L. F. , Marchiano , A. , 
Garbagnati , F. , Spreafi co , C. , Ticha , V. , Gladin , C. R. , Palazzi , M. , Crippa , F. , Oldini , C. , 
REFERENCES 557

558 BIODEGRADABLE NANOPARTICLES 
Calo , S. , Bonaccorsi , A. , Mattavelli , F. , Costa , L. , Mariani , L. , and Cantu , G. ( 2003 ), A 
novel intraarterial chemotherapy using paclitaxel in albumin nanoparticles to treat 
advanced squamous cell carcinoma of the tongue: Preliminary fi ndings , Am. J. Roentgenol. 
, 181 ( 1 ), 253 – 260 . 
82. Rhaese , S. , von Briesen , H. , Rubsamen - Waigmann , H. , Kreuter , J. , and Langer , K. ( 2003 ), 
Human serum albumin - polyethylenimine nanoparticles for gene delivery , J. Controlled 
Release , 92 ( 1 – 2 ), 199 – 208 . 
83. Zhang , L. K. , Hou , S. X. , Mao , S. J. , Wei , D. P. , Song , X. R. , and Qiao , X. R. ( 2004 ), [ Study 
on the tumor cell targetability of folate - conjugated albumin nanoparticles ] Sichuan Da 
Xue Xue Bao Yi Xue Ban , 35 ( 2 ), 165 – 168 . 
84. Zhang , L. , Hou , S. , Mao , S. , Wei , D. , Song , X. , and Lu , Y. ( 2004 ), Uptake of folateconjugated 
albumin nanoparticles to the SKOV3 cells , Int. J. Pharm. , 287 ( 1 – 2 ), 
155 – 162 . 
85. Mishra , V. , Mahor , S. , Rawat , A. , Gupta , P. N. , Dubey , P. , Khatri , K. , and Vyas , S. P. ( 2006 ), 
Targeted brain delivery of AZT via transferrin anchored pegylated albumin nanoparticles 
, J. Drug Target. , 14 ( 1 ), 45 – 53 . 
86. Mao , S. J. , Hou , S. X. , Zhang , L. K. , Jin , H. , Bi , Y. Q. , and Jiang , B. ( 2003 ), [ Preparation 
of bovine serum albumin nanoparticles surface - modifi ed with glycyrrhizin ], Yao Xue 
Xue Bao , 38 ( 10 ), 787 – 790 . 
87. Bazile , D. V. , Ropert , C. , Huve , P. , Verrecchia , T. , Marlard , M. , Frydman , A. , Veillard , M. , 
and Spenlehauer , G. ( 1992 ), Body distribution of fully biodegradable [ 14 C] - poly(lactic 
acid) nanoparticles coated with albumin after parenteral administration to rats , Biomaterials 
, 13 ( 15 ), 1093 – 1102 . 
88. Arangoa , M. A. , Campanero , M. A. , Renedo , M. J. , Ponchel , G. , and Irache , J. M. ( 2001 ), 
Gliadin nanoparticles as carriers for the oral administration of lipophilic drugs. Relationships 
between bioadhesion and pharmacokinetics , Pharm. Res. , 18 ( 11 ), 1521 – 1527 . 
89. Duclairoir , C. , Orecchioni , A. M. , Depraetere , P. , Osterstock , F. , and Nakache , E. ( 2003 ), 
Evaluation of gliadins nanoparticles as drug delivery systems: A study of three different 
drugs , Int. J. Pharm. , 253 ( 1 – 2 ), 133 – 144 . 
90. Umamaheshwari , R. B. , and Jain , N. K. ( 2003 ), Receptor mediated targeting of lectin 
conjugated gliadin nanoparticles in the treatment of Helicobacter pylori , J. Drug Target. , 
11 ( 7 ), 415 – 423 ; discussion 423 – 424. 
91. Jain , R. A. ( 2000 ), The manufacturing techniques of various drug loaded biodegradable 
poly(lactide -co - glycolide) (PLGA) devices , Biomaterials , 21 ( 23 ), 2475 – 2490 . 
92. Bala , I. , Hariharan , S. , and Kumar , M. N. ( 2004 ), PLGA nanoparticles in drug delivery: 
The state of the art , Crit. Rev. Ther. Drug Carrier Syst. , 21 ( 5 ), 387 – 422 . 
93. Astete , C. E. , and Sabliov , C. M. ( 2006 ), Synthesis and characterization of PLGA 
nanoparticles , J. Biomater. Sci. Polym. Ed. , 17 ( 3 ), 247 – 289 . 
94. Sinha , V. R. , Bansal , K. , Kaushik , R. , Kumria , R. , and Trehan , A. ( 2004 ), Poly - epsilon - 
caprolactone microspheres and nanospheres: An overview , Int. J. Pharm. , 278 ( 1 ), 1 – 23 . 
95. Seppala , J. V. , Helminen , A. O. , and Korhonen , H. ( 2004 ), Degradable polyesters through 
chain linking for packaging and biomedical applications , Macromol. Biosci. , 4 ( 3 ), 
208 – 217 . 
96. Kumar , N. , Langer , R. S. , and Domb , A. J. ( 2002 ), Polyanhydrides: An overview , Adv. 
Drug Deliv. Rev. , 54 ( 7 ), 889 – 910 . 
97. Jain , J. P. , Modi , S. , Domb , A. J. , and Kumar , N. ( 2005 ), Role of polyanhydrides as localized 
drug carriers , J. Controlled Release. , 103 ( 3 ), 541 – 563 . 
98. Pfeifer , B. A. , Burdick , J. A. , and Langer , R. ( 2005 ), Formulation and surface modifi cation 
of poly(ester - anhydride) micro - and nanospheres , Biomaterials , 26 ( 2 ), 117 – 124 . 

99. Carino , G. P. , Jacob , J. S. , and Mathiowitz , E. ( 2000 ), Nanosphere based oral insulin 
delivery , J. Controlled Release , 65 ( 1 – 2 ), 261 – 269 . 
100. Torres , M. P. , Determan , A. S. , and Anderson , G. L. , Mallapragada , S. K. , and Narasimhan , 
B. ( 2007 ), Amphiphilic polyanhydrides for protein stabilization and release , Biomaterials 
, 28 ( 1 ), 108 – 116 . 
101. Gao , J. , Niklason , L. , Zhao , X. M. , and Langer , R. ( 1998 ), Surface modifi cation of polyanhydride 
microspheres , J. Pharm. Sci. , 87 ( 2 ), 246 – 248 . 
102. Vauthier , C. , Dubernet , C. , Fattal , E. , Pinto - Alphandary , H. , and Couvreur , P. ( 2003 ), Poly 
(alkylcyanoacrylates) as biodegradable materials for biomedical applications , Adv. Drug 
Deliv. Rev. , 55 ( 4 ), 519 – 548 . 
103. Mehnert , W. , and Mader , K. ( 2001 ), Solid lipid nanoparticles: Production, characterization 
and applications , Adv. Drug Deliv. Rev. , 47 ( 2 – 3 ), 165 – 196 . 
104. Uner , M. ( 2006 ), Preparation, characterization and physico - chemical properties of solid 
lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC): Their benefi ts as 
colloidal drug carrier systems , Pharmazie , 61 ( 5 ), 375 – 386 . 
105. Heller , J. , Barr , J. , Ng , S. Y. , Abdellauoi , K. S. , and Gurny , R. ( 2002 ), Poly(ortho esters): 
Synthesis, characterization, properties and uses , Adv. Drug Deliv. Rev. , 54 ( 7 ), 1015 – 
1039 . 
106. Barichello , J. M. , Morishita , M. , Takayama , K. , and Nagai , T. ( 1999 ), Encapsulation of 
hydrophilic and lipophilic drugs in PLGA nanoparticles by the nanoprecipitation 
method , Drug Dev. Ind. Pharm. , 25 ( 4 ), 471 – 476 . 
107. Panyam , J. , and Labhasetwar , V. ( 2003 ), Biodegradable nanoparticles for drug and gene 
delivery to cells and tissue , Adv. Drug Deliv. Rev. , 55 ( 3 ), 329 – 347 . 
108. Avgoustakis , K. ( 2004 ), Pegylated poly(lactide) and poly(lactide -co - glycolide) nanoparticles: 
Preparation, properties and possible applications in drug delivery , Curr. Drug 
Deliv. , 1 ( 4 ), 321 – 333 . 
109. Panagi , Z. , Beletsi , A. , Evangelatos , G. , Livaniou , E. , Ithakissios , D. S. , and Avgoustakis , 
K. ( 2001 ), Effect of dose on the biodistribution and pharmacokinetics of PLGA and 
PLGA - mPEG nanoparticles , Int. J. Pharm. , 221 ( 1 – 2 ), 143 – 152 . 
110. Qaddoumi , M. G. , Gukasyan , H. J. , Davda , J. , Labhasetwar , V. , Kim , K. J. , and Lee , V. H. 
( 2003 ), Clathrin and caveolin - 1 expression in primary pigmented rabbit conjunctival 
epithelial cells: Role in PLGA nanoparticle endocytosis , Mol. Vis. , 9 , 559 – 568 . 
111. Panyam , J. , Zhou , W. Z. , Prabha , S. , Sahoo , S. K. , and Labhasetwar , V. ( 2002 ), Rapid 
endo - lysosomal escape of poly( dl - lactide - co - glycolide) nanoparticles: Implications for 
drug and gene delivery , FASEB J. , 16 ( 10 ), 1217 – 1226 . 
112. Shive , M. S. , and Anderson , J. M. ( 1997 ), Biodegradation and biocompatibility of PLA 
and PLGA microspheres , Adv. Drug Deliv. Rev. , 28 ( 1 ), 5 – 24 . 
113. Marchal - Heussler , L. , Sirbat , D. , Hoffman , M. , and Maincent , P. ( 1993 ), Poly(epsilon - 
caprolactone) nanocapsules in carteolol ophthalmic delivery , Pharm. Res. , 10 ( 3 ), 
386 – 390 . 
114. Calvo , P. , Vila - Jato , J. L. , and Alonso , M. J. ( 1996 ), Comparative in vitro evaluation of 
several colloidal systems, nanoparticles, nanocapsules, and nanoemulsions, as ocular 
drug carriers , J. Pharm. Sci. , 85 ( 5 ), 530 – 536 . 
115. Jeong , Y. I. , Kang , M. K. , Sun , H. S. , Kang , S. S. , Kim , H. W. , Moon , K. S. , Lee , K. J. , Kim , 
S. H. , and Jung , S. ( 2004 ), All - trans - retinoic acid release from core - shell type nanoparticles 
of poly(epsilon - caprolactone)/poly(ethylene glycol) diblock copolymer , Int. J. 
Pharm. , 273 ( 1 – 2 ), 95 – 107 . 
116. Park , E. K. , Lee , S. B. , and Lee , Y. M. ( 2005 ), Preparation and characterization 
of methoxy poly(ethylene glycol)/poly(epsilon - caprolactone) amphiphilic block 
REFERENCES 559

560 BIODEGRADABLE NANOPARTICLES 
copolymeric nanospheres for tumor - specifi c folate - mediated targeting of anticancer 
drugs , Biomaterials , 26 ( 9 ), 1053 – 1061 . 
117. Cade , D. , Ramus , E. , Rinaudo , M. , Auzely - Velty , R. , Delair , T. , and Hamaide , T. ( 2004 ), 
Tailoring of bioresorbable polymers for elaboration of sugar - functionalized nanoparticles 
, Biomacromolecules , 5 ( 3 ), 922 – 927 . 
118. Haas , J. , Ravi Kumar , M. N. , Borchard , G. , Bakowsky , U. , and Lehr , C. M. ( 2005 ), 
Preparation and characterization of chitosan and trimethyl - chitosan - modifi ed poly - 
(epsilon - caprolactone) nanoparticles as DNA carriers , AAPS Pharm. Sci. Tech. , 6 ( 1 ), 
E22 – 30 . 
119. Lemarchand , C. , Gref , R. , Passirani , C. , Garcion , E. , Petri , B. , Muller , R. , Costantini , D. , 
and Couvreur , P. ( 2006 ), Infl uence of polysaccharide coating on the interactions of 
nanoparticles with biological systems , Biomaterials , 27 ( 1 ), 108 – 118 . 
120. Pfeifer , B. A. , Burdick , J. A. , Little , S. R. , and Langer , R. ( 2005 ), Poly(ester - anhydride): 
poly(beta - amino ester) micro - and nanospheres: DNA encapsulation and cellular transfection 
, Int. J. Pharm. , 304 ( 1 – 2 ), 210 – 219 . 
121. McCarron , P. A. , Donnelly , R. F. , Canning , P. E. , McGovern , J. G. , and Jones , D. S. ( 2004 ), 
Bioadhesive, non - drug - loaded nanoparticles as modulators of candidal adherence to 
buccal epithelial cells: A potentially novel prophylaxis for candidosis , Biomaterials , 
25 ( 12 ), 2399 – 2407 . 
122. Vauthier , C. , Dubernet , C. , Chauvierre , C. , Brigger , I. , and Couvreur , P. ( 2003 ), Drug 
delivery to resistant tumors: The potential of poly(alkyl cyanoacrylate) nanoparticles , 
J. Controlled Release , 93 ( 2 ), 151 – 160 . 
123. Toub , N. , Angiari , C. , Eboue , D. , Fattal , E. , Tenu , J. P. , Le Doan , T. , and Couvreur , P. 
( 2005 ), Cellular fate of oligonucleotides when delivered by nanocapsules of poly(isobu 
tylcyanoacrylate) , J. Controlled Release , 106 ( 1 – 2 ), 209 – 213 . 
124. Labarre , D. , Vauthier , C. , Chauvierre , C. , Petri , B. , Muller , R. , and Chehimi , M. M. ( 2005 ), 
Interactions of blood proteins with poly(isobutylcyanoacrylate) nanoparticles decorated 
with a polysaccharidic brush , Biomaterials , 26 ( 24 ), 5075 – 5084 . 
125. Wissing , S. A. , Kayser , O. , and Muller , R. H. ( 2004 ), Solid lipid nanoparticles for parenteral 
drug delivery , Adv. Drug Deliv. Rev. , 56 ( 9 ), 1257 – 1272 . 
126. Manjunath , K. , Reddy , J. S. , and Venkateswarlu , V. ( 2005 ), Solid lipid nanoparticles as 
drug delivery systems , Methods Find. Exp. Clin. Pharmacol. , 27 ( 2 ), 127 – 144 . 
127. Zhang , N. , Ping , Q. , Huang , G. , Xu , W. , Cheng , Y. , and Han , X. ( 2006 ), Lectin - modifi ed 
solid lipid nanoparticles as carriers for oral administration of insulin , Int. J. Pharm. , 
327 ( 1 – 2 ), 153 – 159 . 
128. Wang , Y. , and Wu , W. ( 2006 ), In situ evading of phagocytic uptake of stealth solid lipid 
nanoparticles by mouse peritoneal macrophages , Drug Deliv. , 13 ( 3 ), 189 – 192 . 
129. Chilkoti , A. , Dreher , M. R. , Meyer , D. E. , and Raucher , D. ( 2002 ), Targeted drug delivery 
by thermally responsive polymers , Adv. Drug Deliv. Rev. , 54 ( 5 ), 613 – 630 . 
130. Sakuma , S. , Suzuki , N. , Sudo , R. , Hiwatari , K. , Kishida , A. , and Akashi , M. ( 2002 ), Optimized 
chemical structure of nanoparticles as carriers for oral delivery of salmon calcitonin 
, Int. J. Pharm. , 239 ( 1 – 2 ), 185 – 195 . 
131. Piskin , E. ( 2004 ), Molecularly designed water soluble, intelligent, nanosize polymeric 
carriers , Int. J. Pharm. , 277 ( 1 – 2 ), 105 – 118 . 
132. Neradovic , D. , Soga, O. , Van Nostrum, C. F. , and Hennink, W. E. (2004), The effect of the 
processing and formulation parameters on the size of nanoparticles based on block 
copolymers of poly(ethylene glycol) and poly( N - isopropylacrylamide) with and without 
hydrolytically sensitive groups , Biomaterials , 25 ( 12 ), 2409 – 2418 . 

133. Na , K. , Lee , K. H. , and Bae , Y. H. ( 2004 ), pH - sensitivity and pH - dependent interior 
structural change of self - assembled hydrogel nanoparticles of pullulan acetate/oligo - 
sulfonamide conjugate , J. Controlled Release , 97 ( 3 ), 513 – 525 . 
134. Ma , Z. , Yeoh , H. H. , and Lim , L. Y. ( 2002 ), Formulation pH modulates the interaction 
of insulin with chitosan nanoparticles , J. Pharm. Sci. , 91 ( 6 ), 1396 – 1404 . 
135. Lynn , D. M. , Amiji , M. M. , and Langer , R. ( 2001 ), pH - responsive polymer microspheres: 
Rapid release of encapsulated material within the range of intracellular pH , Angew 
Chem. Int. Ed. Engl. , 40 ( 9 ), 1707 – 1710 . 
136. Potineni , A. , Lynn , D. M. , Langer , R. , and Amiji , M. M. ( 2003 ), Poly(ethylene oxide) - 
modifi ed poly(beta - amino ester) nanoparticles as a pH - sensitive biodegradable system 
for paclitaxel delivery , J. Controlled Release , 86 ( 2 – 3 ), 223 – 234 . 
137. Heffernan , M. J. , and Murthy , N. ( 2005 ), Polyketal nanoparticles: A new pH - sensitive 
biodegradable drug delivery vehicle , Bioconjug. Chem. , 16 ( 6 ), 1340 – 1342 . 
138. Choi , J. S. , MacKay , J. A. , and Szoka , F. C. Jr. ( 2003 ), Low - pH - sensitive PEG - stabilized 
plasmid - lipid nanoparticles: Preparation and characterization , Bioconjug. Chem. , 14 ( 2 ), 
420 – 429 . 
139. Na , K. , Lee , K. H. , Lee , D. H. , and Bae , Y. H. ( 2006 ), Biodegradable thermo - sensitive 
nanoparticles from poly( l - lactic acid)/poly(ethylene glycol) alternating multi - block 
copolymer for potential anti - cancer drug carrier , Eur. J. Pharm. Sci. , 27 ( 2 – 3 ), 115 – 
122 . 
140. Sun , S. , Liu , W. , Cheng , N. , Zhang , B. , Cao , Z. , Yao , K. , Liang , D. , Zuo , A. , Guo , G. , and 
Zhang , J. ( 2005 ), A thermoresponsive chitosan - NIPAAm/vinyl laurate copolymer vector 
for gene transfection , Bioconjug. Chem. , 16 ( 4 ), 972 – 980 . 
141. Weng , H. , Zhou , J. , Tang , L. , and Hu , Z. ( 2004 ), Tissue responses to thermally - responsive 
hydrogel nanoparticles , J. Biomater. Sci. Polym. Ed. , 15 ( 9 ), 1167 – 1180 . 
142. Devalapally , H. , Shenoy , D. , Little , S. , Langer , R. , and Amiji , M. ( 2007 ), Poly(ethylene 
oxide) - modifi ed poly(beta - amino ester) nanoparticles as a pH - sensitive system for 
tumor - targeted delivery of hydrophobic drugs: Part 3. Therapeutic effi cacy and safety 
studies in ovarian cancer xenograft model , Cancer Chemother. Pharmacol. , 59 ( 4 ), 
477 – 484 . 
143. Brigger , I. , Dubernet , C. , and Couvreur , P. ( 2002 ), Nanoparticles in cancer therapy and 
diagnosis , Adv. Drug Deliv. Rev. , 54 ( 5 ), 631 – 651 . 
144. Mulder , W. J. , Strijkers , G. J. , van Tilborg , G. A. , Griffi oen , A. W. , and Nicolay , K. ( 2006 ), 
Lipid - based nanoparticles for contrast - enhanced MRI and molecular imaging , NMR 
Biomed. , 19 ( 1 ), 142 – 164 . 
145. De , S. , Miller , D. W. , and Robinson , D. H. ( 2005 ), Effect of particle size of nanospheres 
and microspheres on the cellular - association and cytotoxicity of paclitaxel in 4T1 cells , 
Pharm. Res. , 22 ( 5 ), 766 – 775 . 
146. De , S. , and Robinson , D. H. ( 2004 ), Particle size and temperature effect on the physical 
stability of PLGA nanospheres and microspheres containing Bodipy , AAPS Pharm. Sci. 
Tech. , 5 ( 4 ), e53 . 
147. Alonso , M. J. ( 1996 ), Nanoparticulate drug carrier technology , Cohen , S. , and 
Bennstein , H. Eds. Microparticulate Systems for the Delivery of Proteins and Vaccines , 
Marcel Dekkes , New York , pp. 203 – 242 . 
148. Torres , M. P. , Vogel , B. M. , Narasimhan , B. , and Mallapragada , S. K. ( 2006 ), Synthesis and 
characterization of novel polyanhydrides with tailored erosion mechanisms , J. Biomed. 
Mater. Res. A. , 76 ( 1 ), 102 – 110 . 
149. Shen , E. , Pizsczek , R. , Dziadul , B. , and Narasimhan , B. ( 2001 ), Microphase separation in 
bioerodible copolymers for drug delivery , Biomaterials , 22 ( 3 ), 201 – 210 . 
REFERENCES 561

562 BIODEGRADABLE NANOPARTICLES 
150. DiBenedetto , A. K. ( 2003 ), Prediction of the glass transition temperature of polymers: 
A model based on the principle of corresponding states , J. Polym. Sci. Part B: Polym. 
Phys. , 25 ( 9 ), 1949 – 1969 . 
151. Dong , Y. , and Feng , S. S. ( 2006 ), Nanoparticles of poly( d , l - lactide)/methoxy poly(ethylene 
glycol) - poly( d , l - lactide) blends for controlled release of paclitaxel , J. Biomed. Mater. 
Res. A , 78 ( 1 ), 12 – 19 . 
152. Zweers , M. L. , Grijpma , D. W. , Engbers , G. H. , and Feijen , J. ( 2003 ), The preparation of 
monodisperse biodegradable polyester nanoparticles with a controlled size , J. Biomed. 
Mater. Res. B Appl. Biomater. , 66 ( 2 ), 559 – 566 . 
153. Kirby , B. J. , and Hasselbrink , E. F. Jr. ( 2004 ), Zeta potential of microfl uidic substrates: 2. 
Data for polymers , Electrophoresis , 25 ( 2 ), 203 – 213 . 
154. Gessner , A. , Lieske , A. , Paulke , B. , and Muller , R. ( 2002 ), Infl uence of surface charge 
density on protein adsorption on polymeric nanoparticles: Analysis by two - dimensional 
electrophoresis , Eur. J. Pharm. Biopharm. , 54 ( 2 ), 165 – 170 . 
155. Kumar , M. N. , Mohapatra , S. S. , Kong , X. , Jena , P. K. , Bakowsky , U. , and Lehr , C. M. 
( 2004 ), Cationic poly(lactide -co - glycolide) nanoparticles as effi cient in vivo gene transfection 
agents , J. Nanosci. Nanotechnol. , 4 ( 8 ), 990 – 994 . 
156. Reddy , L. H. , Sharma , R. K. , Chuttani , K. , Mishra , A. K. , and Murthy , R. R. ( 2004 ), 
Etoposide - incorporated tripalmitin nanoparticles with different surface charge: Formulation, 
characterization, radiolabeling, and biodistribution studies , AAPS J. , 6 ( 3 ), 
e23 . 
157. Owens , D. E. , 3rd , and Peppas , N. A. ( 2006 ), Opsonization, biodistribution, and pharmacokinetics 
of polymeric nanoparticles , Int. J. Pharm. , 307 ( 1 ), 93 – 102 . 
158. Moghimi , S. M. , and Szebeni , J. ( 2003 ), Stealth liposomes and long circulating nanoparticles: 
Critical issues in pharmacokinetics, opsonization and protein - binding properties , 
Prog. Lipid Res. , 42 ( 6 ), 463 – 478 . 
159. Freiberg , S. , and Zhu , X. X. ( 2004 ), Polymer microspheres for controlled drug release , 
Int. J. Pharm. , 282 ( 1 – 2 ), 1 – 18 . 
160. Siepmann , J. , and Gopferich , A. ( 2001 ), Mathematical modeling of bioerodible, polymeric 
drug delivery systems , Adv. Drug Deliv. Rev. , 48 ( 2 – 3 ), 229 – 247 . 
161. Grassi , M. , and Grassi , G. ( 2005 ), Mathematical modelling and controlled drug delivery: 
Matrix systems , Curr. Drug Deliv. , 2 ( 1 ), 97 – 116 . 
162. Kanjickal , D. G. , and Lopina , S. T. ( 2004 ), Modeling of drug release from polymeric 
delivery systems — a review , Crit. Rev. Ther. Drug Carrier Syst. , 21 ( 5 ), 345 – 386 . 
163. Fu , K. , Harrell , R. , Zinski , K. , Um , C. , Jaklenec , A. , Frazier , J. , Lotan , N. , Burke , P. , 
Klibanov , A. M. , and Langer , R. ( 2003 ), A potential approach for decreasing the burst 
effect of protein from PLGA microspheres , J. Pharm. Sci. , 92 ( 8 ), 1582 – 1591 . 
164. Dhoot , N. O. , and Wheatley , M. A. ( 2003 ), Microencapsulated liposomes in controlled 
drug delivery: Strategies to modulate drug release and eliminate the burst effect , 
J. Pharm. Sci. , 92 ( 3 ), 679 – 689 . 
165. Lemoine , D. , Francois , C. , Kedzierewicz , F. , Preat , V. , Hoffman , M. , and Maincent , P. 
( 1996 ), Stability study of nanoparticles of poly(epsilon - caprolactone), poly( d , l - lactide) 
and poly( d , l - lactide - co - glycolide) , Biomaterials , 17 ( 22 ), 2191 – 2197 . 
166. Molpeceres , J. , Aberturas , M. R. , Chacon , M. , Berges , L. , and Guzman , M. ( 1997 ), Stability 
of cyclosporine - loaded poly - sigma - caprolactone nanoparticles , J. Microencapsul. , 
14 ( 6 ), 777 – 787 . 
167. Freitas , C. , and Muller , R. H. ( 1999 ), Correlation between long - term stability of solid 
lipid nanoparticles (SLN) and crystallinity of the lipid phase , Eur. J. Pharm. Biopharm. , 
47 ( 2 ), 125 – 132 . 

168. Freitas , C. , and Muller , R. H. ( 1999 ), Stability determination of solid lipid nanoparticles 
(SLN) in aqueous dispersion after addition of electrolyte , J. Microencapsul. , 16 ( 1 ), 
59 – 71 . 
169. Santander - Ortega , M. J. , Jodar - Reyes , A. B. , Csaba , N. , Bastos - Gonzalez , D. , and Ortega - 
Vinuesa , J. L. ( 2006 ), Colloidal stability of Pluronic F68 - coated PLGA nanoparticles: 
A variety of stabilisation mechanisms , J. Colloid Interface Sci. , 302 ( 2 ), 522 – 529 . 
170. Zobel , H. P. , Werner , D. , Gilbert , M. , Noe , C. R. , Stieneker , F. , Kreuter , J. , and Zimmer , 
A. ( 1999 ), Effect of ultrasonication on the stability of oligonucleotides adsorbed on 
nanoparticles and liposomes , J. Microencapsul. , 16 ( 4 ), 501 – 509 . 
171. Hattori , Y. , Sakaguchi , M. , and Maitani , Y. ( 2006 ), Folate - linked lipid - based nanoparticles 
deliver a NFkappaB decoy into activated murine macrophage - like RAW264.7 cells , 
Biol. Pharm. Bull. , 29 ( 7 ), 1516 – 1520 . 
172. Sahoo , S. K. , Ma , W. , and Labhasetwar , V. ( 2004 ), Effi cacy of transferrin - conjugated 
paclitaxel - loaded nanoparticles in a murine model of prostate cancer , Int. J. Cancer , 
112 ( 2 ), 335 – 340 . 
173. Gao , X. , Tao , W. , Lu , W. , Zhang , Q. , Zhang , Y. , Jiang , X. , and Fu , S. ( 2006 ), Lectin - 
conjugated PEG - PLA nanoparticles: Preparation and brain delivery after intranasal 
administration , Biomaterials , 27 ( 18 ), 3482 – 3490 . 
174. Sokolov , K. , Follen , M. , Aaron , J. , Pavlova , I. , Malpica , A. , Lotan , R. , and Richards - 
Kortum , R. ( 2003 ), Real - time vital optical imaging of precancer using anti - epidermal 
growth factor receptor antibodies conjugated to gold nanoparticles , Cancer Res. , 63 ( 9 ), 
1999 – 2004 . 
175. Garnett , M. C. ( 2001 ), Targeted drug conjugates: Principles and progress , Adv. Drug 
Deliv. Rev. , 53 ( 2 ), 171 – 216 . 
176. Nobs , L. , Buchegger , F. , Gurny , R. , and Allemann , E. ( 2004 ), Current methods for 
attaching targeting ligands to liposomes and nanoparticles , J. Pharm. Sci. , 93 ( 8 ), 
1980 – 1992 . 
177. Kreuter , J. ( 1994 ), Drug targeting with nanoparticles , Eur. J. Drug Metab. Pharmacokinet. 
, 19 ( 3 ), 253 – 256 . 
178. Jain , K. K. ( 2006 ), Nanoparticles as targeting ligands , Trends Biotechnol. , 24 ( 4 ), 
143 – 145 . 
179. Conner , S. D. , and Schmid , S. L. ( 2003 ), Regulated portals of entry into the cell , Nature , 
422 ( 6927 ), 37 – 44 . 
180. Huang , M. , Ma, Z. , Khor , E. , and Lim, L. Y. (2002), Uptake of FITC-chitosan nanoparticles 
by A549 cells , Pharm. Res. , 19 ( 10 ), 1488 – 1494 . 
181. Ma , Z. , and Lim , L. Y. ( 2003 ), Uptake of chitosan and associated insulin in Caco - 2 cell 
monolayers: A comparison between chitosan molecules and chitosan nanoparticles , 
Pharm. Res. , 20 ( 11 ), 1812 – 1819 . 
182. Liu , J. , and Shapiro , J. I. ( 2003 ), Endocytosis and signal transduction: Basic science 
update , Biol. Res. Nurs. , 5 ( 2 ), 117 – 128 . 
183. Steinman , R. M. , Mellman , I. S. , Muller , W. A. , and Cohn , Z. A. ( 1983 ), Endocytosis and 
the recycling of plasma membrane , J. Cell Biol. , 96 ( 1 ), 1 – 27 . 
184. Okamoto , C. T. ( 1998 ), Endocytosis and transcytosis , Adv. Drug Deliv. Rev. , 29 ( 3 ), 
215 – 228 . 
185. Hilgenbrink, A. R. , and Low , P. S. (2005), Folate receptor -mediated drug targeting: From 
therapeutics to diagnostics , J. Pharm. Sci. , 94 ( 10 ), 2135 – 2146 . 
186. Park , I. K. , Seo , S. J. , Akashi , M. , Akaike , T. , and Cho , C. S. ( 2003 ), Controlled release of 
epidermal growth factor (EGF) from EGF - loaded polymeric nanoparticles composed 
REFERENCES 563

564 BIODEGRADABLE NANOPARTICLES 
of polystyrene as core and poly(methacrylic acid) as corona in vitro , Arch. Pharm. Res. , 
26 ( 8 ), 649 – 652 . 
187. Douglas , S. J. , Davis , S. S. , and Illum , L. ( 1987 ), Nanoparticles in drug delivery , Crit. Rev. 
Ther. Drug Carrier Syst. , 3 ( 3 ), 233 – 261 . 

565 
5.5 
RECOMBINANT SACCHAROMYCES 
CEREVISIAE AS NEW DRUG 
DELIVERY SYSTEM TO GUT: IN VITRO 
VALIDATION AND ORAL 
FORMULATION 
St ephanie Blanquet and Monique Alric 
Universit e d ’ Auvergne, Clermont - Ferrand, France 
Contents 
5.5.1 Enginereed Microorganisms as Delivery Vectors to Human Gastrointestinal Tract 
5.5.1.1 What is the “ Biodrug ” Concept? 
5.5.1.2 Medical Applications 
5.5.1.3 Choice of Candidate Host Microorganisms 
5.5.2 Evaluation of Scientifi c Feasibility of Biodrug Concept Using Yeast as Vector 
5.5.2.1 Approach 
5.5.2.2 Yeast Survival Rate in Simulated Gastrointestinal Conditions 
5.5.2.3 Yeast Heterologous Activity in Simulated Gastrointestinal Conditions 
5.5.2.4 Conclusion 
5.5.3 Oral Formulation of Recombinant Yeasts 
5.5.3.1 Freeze Drying of Recombinant Model Yeasts 
5.5.3.2 Immobilization of Recombinant Model Yeasts in Whey Protein Beads 
5.5.4 General Conclusion and Future Developments 
References 
5.5.1 ENGINEREED MICROORGANISMS AS DELIVERY VECTORS TO 
HUMAN GASTROINTESTINAL TRACT 
5.5.1.1 What Is the “ Biodrug ” Concept? 
The development of recombinant deoxyribonucleic acid (DNA) technology has 
allowed the emergence of novel applications such as drug production directly in the 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

566 IN VITRO VALIDATION AND ORAL FORMULATION 
human digestive environment ( “ in situ ” ) by ingested living recombinant microorganisms 
[1 – 3] . 
This new kind of vector offers several advantages over classical dosage forms. 
First, the microorganisms, by protecting the active compounds, can allow the administration 
of drugs known to be sensitive to digestive conditions when given in classical 
pharmaceutical formulations. Second, the regulation of gene expression (e.g., 
using an inducible promoter) makes it possible to target specifi c sites throughout 
the digestive tract (i.e., the absorption or reaction site of the drug) and to control 
drug release. Thus, similar therapeutic effects can potentially be obtained at lower 
doses [4, 5] and the degradation of the active compound by acid or proteases should 
be avoided upstream from its absorption or reaction site. 
In the digestive tract, genetically modifi ed microorganisms can either carry out 
a reaction of bioconversion or produce compounds of interest. The bioconversion 
reaction can lead either to the production of an active product or to the removal of 
undesirable compounds. The active compound produced in situ can be secreted in 
the digestive medium [5] , be bound to the cells [6, 7] , or accumulate inside the cells 
and be released in the digestive environment by cell lysis [8] . 
5.5.1.2 Medical Applications 
The biodrug concept involves the use of orally administered recombinant microorganisms 
as a new drug delivery route to prevent or treat various diseases. The 
potential medical applications are numerous and can be classifi ed in terms of bioconversion 
or production of active compounds. Validation studies have yet been 
conducted in animals (and even sometimes in human being), as described below 
and summarized in Table 1 . 
Bioconversion Three main types of medical applications have been considered. 
First, recombinant microorganisms could be administered to perform “ biodetoxication 
” in the gut [9] . Here, the objective is to increase the body ’ s protection against 
environmental xeniobiotics, mainly those ingested with food (e.g., pesticides, procarcinogens, 
or chemical additives), by ingesting microorganisms expressing enzymes 
that play a major role in the human detoxication system [e.g., phase I xenobiotic 
metabolizing enzymes such as cytochrome P450 or phase II – like glutathione S - 
transferase (GST)]. Therefore, recombinant microorganisms could be used to 
prevent multifactorial diseases that have been associated with anomalies in human 
detoxication processes. For instance, a defi ciency in GST - M1 has been correlated 
with an increased susceptibility to different cancers, endometriosis, and chronic 
bronchitis [10] . 
Second, modifi ed microorganisms could correct errors of metabolism resulting 
from either gastric or intestinal enzyme defi ciencies (e.g., lipase or lactase) [11] or 
organ failure (by removing urea in the case of kidney failure or ammonia in the 
case of liver failure) [12, 13] . This could constitute an alternative to current therapy 
such as renal dialysis, which is time consuming and uncomfortable for the patient. 
The third potential application is the use of recombinant cells to control the 
activation of prodrug into drug directly in the digestive tract. This is of interest when 
the drug, but not the prodrug, is either toxic at high concentrations or damaged by 
digestive secretions [1] . 

TABLE 1 Potential Medical Applications of Biodrug Concept and Their Validation in Animals 
Biodrug 
Concept Applications 
Examples 
Validation 
Recombinant 
Microorganisms 
(Heterologous Gene) Experimental Models 
Effect 
Bioconversion Biodetoxication Removal of 
benzo( a )pyrene [9] 
Escherichia coli 
(P450 
1A1 and glutathione 
S - transferase) 
Stomach and duodenum 
of a mouse combined 
with mutagenesis assay 
(Ames assay) 
Decrease of the mutagenic 
potential of B(a)P 
Correction of 
errors of 
metabolism 
Correction of lipase 
defi 
ciency [11] 
Lactococcus lactis 
(lipase) 
Pig with pancreatic 
defi ciency, oral 
administration 
Increase of lipid 
digestibility 
Correction of urease 
defi 
ciency [13] 
Escherichia coli 
DH5 
(urease) 
Rat with renal failure, 
oral administration 
Decrease of plasma uric 
acid 
In situ 
production 
of active 
compounds 
Synthesis of 
biological 
mediators 
Secretion of 
Interleukine [5] 
Lactococcus lactis 
(IL - 10) 
Mouse treated with 
dextran sulfate sodium, 
oral administration 
Reduction of colitis 
symptoms 
Oral vaccines Vaccination against 
bacteria [17] 
Lactobacillus plantarum 
( Helicobacter pylori 
urease B subunit) 
Mouse, 
intragastric 
immunization 
Induction of immune 
response (anti - UreB Ig), 
partial protection against 
H. felis 
Vaccination against 
virus [19] 
Attenuated 
Salmonella 
typhimurium 
(SIV 
capsid antigen p27) 
Macaques, 
oral 
administration 
Induction of immune 
response (anti - p27 Ig) 
Control of food 
allergy [20] 
Lactococcus lactis 
(bovine 
. - lactoglobulin) 
Model of mouse allergy, 
oral administration 
Induction of BLG - specifi c 
Th1 response 
567

568 IN VITRO VALIDATION AND ORAL FORMULATION 
In Situ Production of Active Compounds The fi rst (and main) medical application 
derived from the biodrug concept is the development of oral vaccines [14] . In that 
case, the microorganisms will locally deliver the antigens to the digestive mucosa in 
order to stimulate an immune response (production of immunoglobulins) and 
ensure a protection against bacterial [7, 15 – 17] or viral [6, 18, 19] diseases or being 
used in the management of food allergy [4, 20] . For several immunological and 
practical reasons, these new vaccines represent a promising alternative to the traditional 
injectable ones [21] . In particular, oral immunization is the most effi cient way 
to induce a protective local immune response at the site of pathogen contact. 
Recently, clinical trials have shown the vaccinal effi ciency of different recombinant 
strains of attenuated Salmonella typhi in humans, but the patients were presenting 
undesirable side effects (diarrhea) [22] . 
Other medical applications involve the direct production in the digestive medium 
of various biological mediators, such as insulin, cytokines [23] , or growth factors. In 
particular, new approaches to treating infl ammatory bowel diseases (IBD) such as 
Crohn ’ s disease, celiac disease, or ulcerative colitis have been considered [24] . Anti - 
infl ammatory and immunosuppressive therapies are commonly used for the treatment 
of IBD. However, patients are often subject to unpleasant side effects owing 
to the high level of the drug in the body (systemic administration) and some of them 
remain refractory to such treatments. Steidler et al. [5] have investigated the potential 
of alternative therapeutics and shown the interest of delivering interleukin - 10 
(IL - 10), a strong anti - infl ammatory cytokine, in a localized manner by the action of 
recombinant Lactococcus lactis . Oral administration of the strain producing mouse 
IL - 10 led to 50% curing of dextran sulfate sodium – induced colitis and prevented 
the onset of the pathology in mice [5] . The amount of IL - 10 required to achieve the 
healing effect was 10,000 - fold lower when the cytokine was in situ delivered by 
L. lactis compared to systemic treatment with anti - infl ammatory drugs (e.g., dexamethasone). 
A recent study of the same author has reported that the treatment of 
Crohn ’ s disease patients with L. lactis secreting human IL - 10 was safe and allowed 
a decrease in disease activity [25] . 
5.5.1.3 Choice of Candidate Host Microorganisms 
Recombinant bacteria, particularly lactic acid bacteria, have been mostly suggested 
as potential hosts for this new kind of drug delivery system [2, 3] . However, 
yeasts can offer advantages, especially when a eukaryotic environment is required 
for the functional expression of human genes. Moreover, the absence of bacterial 
sequences liable to promote gene transfer to host bacteria can be ensured using 
the effi cient site - targeted homologous recombination machinery of yeasts for 
introduction of the heterologous gene into the yeast genome. Lastly, yeasts are 
not sensitive to antibacterial agents, allowing the simultaneous administration of 
the recombinant microorganisms and antibiotics. In this study, the common 
baker ’ s yeast Saccharomyces cerevisiae was chosen owing to its “ generally recognized 
as safe ” (GRAS) status and its easy culture and genetic engineering. Saccharomyces 
spp. have already been used in humans, mainly in the treatment of 
intestinal functional disorders such as colitis [26] or antibiotic - associated 
diarrhea [27] . 

5.5.2 EVALUATION OF SCIENTIFIC FEASIBILITY OF BIODRUG 
CONCEPT USING YEAST AS VECTOR 
5.5.2.1 Approach 
The scientifi c feasibility of our approach was recently evaluated using recombinant 
S. cerevisiae expressing model genes and an original artifi cial digestive system simulating 
human gastrointestinal conditions. The survival rate and heterologous activity 
of the recombinant model yeasts were followed in this in vitro system. 
Recombinant Model Yeasts Three different yeast strains, all derived from the 
haploid strain W303 - 1B, were used to evaluate the scientifi c feasibility of the biodrug 
concept using yeast as vector. 
The fi rst one, WRP45073A1 (provided by Denis Pompon, CNRS, Gif - sur - Yvette, 
France), expresses the plant P45073A1 when grown in the presence of galactose 
[28] . P45073A1 was chosen as a model for a reaction catalyzed by a P450 owing to 
the nontoxicity and easy quantifi cation of both substrate and product [29] . It 
catalyzes the 4 - hydroxylation of trans - cinnamic acid into p - coumaric acid [cinnamate 
4 - hydroxylase (CA4H) activity]. In this model, the heterologous protein is 
an intracellular enzyme, and it was synthesized (induction of CYP73A1 by galactose 
during the last 12 h of culture) before yeast introduction into the artifi cial digestive 
system. The in situ CA4H activity of yeasts was quantifi ed following the simultaneous 
introduction of recombinant yeasts and trans - cinnamic acid into the in vitro 
system. 
The two other strains — WppGSTV 5 H 6 and WppV 5 H 6 — were genetically engineered 
[30] to secrete (i) a model protein derived from the commonly used reporter 
protein GST, named GST – V 5 H 6 [molecular weight (MW) 31.5 kDa], and (ii) a model 
peptide, peptide – V 5 H 6 (MW 5.6 kDa). The recombinant protein and peptide were 
expressed in fusion with the V 5 epitope (V 5 ) and the polyhistidine (H 6 ) tag to allow 
their immunological detection and make easier their purifi cation, respectively. A 
leader sequence derived from that of the . - factor precursor was used to direct the 
secretion of the heterologous protein compounds into the extracellular medium 
[30] . In that case, recombinant yeasts and galactose, the inductor of the heterologous 
genes, were simultaneously introduced into the artifi cial digestive system to evaluate 
the yeast ability to initiate the synthesis and secrete protein compounds of various 
sizes, directly in the digestive environment. 
Artifi cial Digestive System TIM : Powerful In Vitro Tool The system TIM (TNO 
gastrointestinal tract model) is the in vitro model that at present time allows the 
closest simulation of in vivo dynamic physiological processes occurring within the 
lumen of the stomach and small intestine of humans [31] . It is composed of four 
successive compartments reproducing the stomach and the three parts of the small 
intestine: the duodenum, jejunum, and ileum (Figure 1 ). This dynamic, computer - 
controlled system has been designed to accept parameters and data from in vivo 
studies on human volunteers. The main parameters of digestion, such as pH, body 
temperature, peristaltic mixing and transport, gastric, biliary, and pancreatic secretions, 
and passive absorption of small molecules (e.g., nutrients, drugs) and water, 
BIODRUG CONCEPT USING YEAST AS VECTOR 569

570 IN VITRO VALIDATION AND ORAL FORMULATION 
are reproduced as accurately as possible (Table 2 ). This system has been previously 
described in detail [1, 29 – 31] . 
Compared with animal experiments, this in vitro system offers accuracy, reproducibility, 
easy manipulation, and the possibility of collecting samples at any level 
of the digestive tract and at any time during digestion with no ethical constraint. It 
has been validated by microbial, nutritional, and pharmaceutical studies. For instance, 
validation experiments demonstrate the predictive value of the TIM with regard to 
the survival rate of probiotic bacteria [32, 33] , the digestibility of nutrients [31, 34] , 
and the availability for absorption of minerals [35] , vitamins [36] , food mutagens 
[37] , and drugs such as paracetamol [33, 38] . 
In the present study, the TIM was programmed to reproduce gastrointestinal 
conditions of the adult after the intake of a liquid meal, according to in vivo data 
[31, 39 – 41] . The initial “ meal ” (introduced into the artifi cial stomach at the beginning 
of digestion) was composed of (i) 10 10 WRP45073A1 cells and 200 . mol of trans - 
cinnamic acid or (ii) 3 . 10 10 WppGSTV 5 H 6 or WppV 5 H 6 cells and galactose (40 g/L) 
FIGURE 1 Gastric and small intestinal system TIM. 
Jejunal 
absorption 
Pancreatic juice 
Hollow fibers 
Electrolytes 
pH electrode Stomach
Sodium bicarbonate 
/electrolytes 
Pepsine, lipase 
Sodium 
bicarbonate 
Flexible wall 
Bile salts 
Hydrochloric 
acid/water 
37°C 37°C 
Dialysates 
Pression sensor 
Ileal absorption 
Meal 
Pump 
Sodium 
bicarbonate 
Peristaltic valves 
Ileum 
Ileal delivery 
Duodenum 
Jejunum

TABLE 2 Digestive Parameters Reproduced in Gastrointestinal Model TIM and Their 
Simulation 
pH The pH is computer monitored and continuously controlled in each 
digestive compartment. 
The fall of gastric pH is reproduced by adding hydrochloric acid. 
The pH is kept to 6.5, 6.8, and 7.2 in the duodenum, jejunum, and ileum, 
respectively, by secreting sodium bicarbonate. 
Temperature The compartments are surrounded by water at body temperature (37 ° C). 
Peristaltic 
mixing 
Peristaltic mixing is mimicked by alternate compression and relaxation of 
the fl exible walls containing the chyme, following changes in the water 
pressure. 
Dynamic of 
chyme 
transit 
A mathematical model using power exponential equations [39] is used to 
reproduce gastric and ileal deliveries ( f = 1 . 2 . ( t / t 1/2) . , where f represents 
the fraction of meal delivered, t the time of delivery, t 1/2 the half - time of 
delivery, and . a coeffi cient describing the shape of the curve). 
Chyme transit is regulated by opening or closing the peristaltic valves that 
connect the compartments. 
Volume The volume in each compartment is monitored with a pressure sensor 
connected to the computer. 
Digestive 
secretions 
Simulated gastric, biliary, and pancreatic secretions are introduced into the 
corresponding compartments by computer - controlled pumps. 
Absorption 
of small 
molecules 
and water 
Semipermeable membrane units are connected to the jejunum and ileum 
to remove the products of digestion as well as water. 
in suspension in 300 mL of yeast culture medium. The parameters of in vitro digestion 
are summarized in Table 3 . 
5.5.2.2 Yeast Survival Rate in Simulated Gastrointestinal Conditions 
At the end of in vitro digestion, yeast survival rate was evaluated by comparing the 
total ingested yeasts with the living yeasts recovered in both the ileal effl uents of 
the TIM and the residual digestive content. After 240 or 270 min digestion (depending 
on the strain), 79.5 ± 12.1% ( n = 3), 63.9 ± 2.4% ( n = 3) and 75.5 ± 25.3% ( n = 
4) of the ingested WRP45073A1, WppV 5 H 6 , and WppGSTV 5 H 6 , respectively, were 
recovered in the ileal effl uents (Figure 2 ). When the yeasts remaining in the residual 
chyme were added ( t = Tf), 95.6 ± 10.1% ( n = 3), 83.1 ± 9.6% ( n = 3), and 95.3 ± 
22.7% ( n = 4) survival percentages, respectively, were found, showing the high resistance 
of recombinant yeasts to gastric (pepsin and lipase) and small intestinal (bile 
salts and pancreatic juice) secretions and low gastric pH [29, 30] . This high survival 
rate was confi rmed (Figure 2 ), no signifi cant difference ( p < 0.05) being observed 
during digestion between the ileal recovery profi les of recombinant yeasts (except 
for WppV 5 H 6 ) and that of a nonabsorbable marker, blue dextran, added in the 
artifi cial stomach at the beginning of digestion, as previously described [31] . 
The survival rate of other microorganisms, such as lactic acid bacteria, has also 
been studied in the TIM. At the end of digestion, Marteau et al. [32] found a bacte- 
BIODRUG CONCEPT USING YEAST AS VECTOR 571

572 IN VITRO VALIDATION AND ORAL FORMULATION 
TABLE 3 Parameters of In Vitro Digestion in TIM When Simulating Gastrointestinal 
Conditions of Adult After Intake of Liquid Meal 
Gastric 
compartment 
Initial volume 300 mL 
Time (min)/pH 0/6 
20/4.2 
40/2.8 
60/2.1 
90/1.8 
120/1.7 
Secretions 0.25 mL/min pepsin (590 IU/mL) 
0.25 mL/min lipase (37.5 IU/mL) 
0.25 mL/min HCl 0.5 M if necessary 
Time of half emptying t 1/2 30 min 
. coeffi cient 1 
Duodenal 
compartment 
Volume 30 mL 
pH Maintained at 6.5 
Secretion 0.5 mL/min bile salts (4% during fi rst 
30 min of digestion, then 2%) 
0.25 mL/min pancreatic juice (10 3 USP/mL) 
0.25 mL/min intestinal electrolyte solution 
0.25 mL/min NaHCO 3 1 M if necessary 
Jejunal 
compartment 
Volume 70 mL 
pH Maintained at 6.8 
Secretion 0.25 mL/min NaHCO 3 1 M if necessary 
Dialysis 10 mL/min of jejunal fl uid solution 
Ileal 
compartment 
Volume 70 mL 
pH Maintained at 7.2 
Secretion 0.25 mL/min NaHCO 3 1 M if necessary 
Dialysis 10 mL/min ileal fl uid solution 
Time of half emptying t 1/2 160 min 
. coeffi cient 1.6 
rial cumulative delivery from the ileum between 0 and 25% (depending on the 
tested strain). In this work, under similar experimental conditions, about 75% of 
ingested yeasts were recovered. Until now, the feasibility of the biodrug concept 
had been mainly evaluated with lactic acid bacteria (see Section 5.5.1.2 ). The high 
viability of S. cerevisiae in the digestive tract might favor the choice of yeasts over 
lactic acid bacteria as hosts for the development of biodrugs, particularly if the viability 
of the microorganisms is required for their in situ activity. 
5.5.2.3 Yeast Heterologous Activity in Simulated Gastrointestinal Conditions 
Bioconversion The CA4H activity of WRP45073A1 yeasts was quantifi ed measuring 
p - coumaric acid production by high - performance liquid chromatography. 
Control experiments showed that both trans - cinnamic and p - coumaric acids were 
stable under digestive conditions when no yeast was introduced into the TIM. In 
the presence of yeasts with no CA4H gene in their plasmid, no p - coumaric acid was 
produced, showing the specifi city of the enzymatic reaction catalyzed by the recombinant 
model yeasts. 

FIGURE 2 Survival rate of three recombinant model yeasts in TIM. The cumulative ileal 
deliveries of viable yeasts and that of a nonabsorbable marker, bleu dextran, are represented. 
At the end of digestion, the percentages obtained in the cumulative ileal effl uents (0 – 240 min 
or 0 – 270 min depending on strain) and in the residual digestive content are added ( t = Tf). 
Results are expressed as mean percentages ± SD ( n = 3 for WRP45073A1 and WppV 5 H 6 , 
n = 4 for WppGSTV 5 H 6 ) of intake. 
0 
20 
40 
60 
80 
100 
120 
0 60 120 180 240 300 360 
Time of digestion (min) 
Cumulative ileal delivery of viable yeasts 
(% of intake) 
WRP45073A1 
WppGSTV5H6 
WppV5H6 
marker 
Tf 
At the end of digestion (240 min), 41.0 ± 5.8% ( n = 3) of initial trans - cinnamic 
acid was converted into p - coumaric acid (Figure 3 a ) [29] . By means of a computer 
simulation [29] , in each compartment of the in vitro system, the amount of p - 
coumaric acid resulting from the CA4H activity of yeasts could be dissociated from 
that delivered by the previous compartment. After calculation, trans - cinnamic acid 
conversion rates of 8.9 ± 1.6%, 13.8 ± 3.3%, 11.8 ± 3.4%, and 6.5 ± 1.0% ( n = 3) 
were found in the stomach, duodenum, jejunum, and ileum, respectively (Figure 3 b ). 
The enzymatic reaction occurred throughout the artifi cial gastrointestinal tract, but 
mostly in the duodenum and jejunum. This could be explained by the fact that yeasts 
were no longer stressed by the acid pH of the stomach and could metabolize the 
trans - cinnamic acid that had previously easily entered the cells, owing to the low 
pH ( trans - cinnamic acid is essentially in a cationic form which easily diffuses through 
the yeast membrane [42] ). Also, previous studies have demonstrated that bile salts 
can favor enzymatic reactions [43] . The lower activity in the ileum might result from 
a decrease in the availability of trans - cinnamic acid owing to its previous conversion 
into p - coumaric acid in the upper digestive compartments. The computer simulation 
that was developed here should prove useful in future stages of the development 
of biodrugs, especially if a specifi c level of the digestive tract has to be targeted for 
drug action. 
Further calculations were performed to quantify the specifi c enzymatic activity 
of the WRP45073A1 yeasts. Yeast specifi c activity in the TIM (from 0.05 ± 0.04 . 
10 . 10 to 3.36 ± 0.86 . 10 . 10 . mol/cell/min, depending on the digestive compartment 
and the sampling time [29] ) was close to that observed in classical batch cultures, 
BIODRUG CONCEPT USING YEAST AS VECTOR 573

574 IN VITRO VALIDATION AND ORAL FORMULATION 
FIGURE 3 CA4H activity of WRP45073A1 in TIM. The trans - cinnamic acid conversion 
rate was evaluated in ( a ) overall TIM and ( b ) each compartment of TIM. Results are 
expressed as mean percentages ± SD ( n = 3) of ingested trans - cinnamic acid converted into 
p - coumaric acid. (Reprinted with permission from Blanquet et al., Applied and Environmental 
Microbiology , 69, 2889.) 
0 
10 
20 
30 
40 
50 
0 60 120 180 240 300 
0 60 120 180 240 300 
Time of digestion (min) 
Trans-cinnamic acid conversion (%) 
0
2
4
6
8 
10 
12 
14 
16 
18 
Time of digestion (min) 
Trans-cinnamic acid conversion (%) 
Stomach Duodenum Jejunum Ileum 
(a) 
(b) 
which is very encouraging for a potential use of S. cerevisiae as a biodetoxication 
system to the gut. 
Secretion of Peptides or Proteins The production of the GST – V 5 H 6 and peptide – 
V 5 H 6 in the TIM was examined by Western blotting (data not shown). No signal was 
detectable during control digestions without yeast or with yeasts with no heterologous 
gene in their plasmid. The model protein and peptide were detected as early 
as 90 min after the yeast intake/gene induction in each compartment of the in vitro 
system and remained until 270 min of digestion in the lower part of the small intes

FIGURE 4 Immunoenzymatic (ELISA) measurement of GST – V 5 H 6 produced by WppGSTV 
5 H 6 in ( a ) different compartments of TIM (ng/mL) and ( b ) the overall TIM ( . g). Error 
bars represent standard deviations ( n = 4). (Reprinted with permission from 110, Blanquet 
et al., Journal of Biotechnology , 45, 2006. Copyright 2006 by Elsevier.) 
0
2
4
6
8 
10 
12 
14 
16 
18 
20 
Time of digestion (min) 
GST-V5H6 (ng/mL) 
Stomach 
Duodenum 
Jejunum 
Ileum 
0,0 
0,5 
1,0 
1,5 
2,0 
2,5 
3,0 
3,5 
4,0 
4,5 
Time of digestion (min) 
GST-V5H6 (.g) 
(a) 
(b) 
0 60 120 180 240 300 
0 60 120 180 240 300 
tine [30] . No signal was detectable in the “ meal ” before introduction into the artifi - 
cial stomach, showing the effi cient initiation of protein compound synthesis and 
secretion by galactose in the digestive environment. 
The amount of GST – V 5 H 6 produced in each compartment of the TIM was quanti- 
fi ed by enzyme - linked immunosorbent assay (ELISA). The protein concentrations 
in the digestive medium reached 15 ng/mL, the highest values being found in the 
jejunum and ileum from 150 min to the end of digestion (Figure 4 a ). The GST – V 5 H 6 
concentrations in the digestive environment were close to those measured in stan- 
BIODRUG CONCEPT USING YEAST AS VECTOR 575

576 IN VITRO VALIDATION AND ORAL FORMULATION 
dard batch cultures. Therefore, the low secretion levels of GST – V 5 H 6 may be only 
imputed to the large size of the protein and/or the genetic construction, but not to 
the particular digestive conditions. This hypothesis is consistent with the results of 
Zsebo et al. [44] , who showed that S. cerevisiae is an effi cient host for small polypeptide 
secretion, but not for larger proteins, which accumulate in the periplasmic 
place and cell wall of the yeast. Improved secretion levels might be obtained with 
another heterologous protein or a different expression vector. In the overall in vitro 
system, the total amount of GST – V 5 H 6 regularly increased during digestion to reach 
3.3 ± 0.7 . g ( n = 4) after 270 min digestion (Figure 4 b ). 
To check that the GST – V 5 H 6 recovered in the TIM was truly secreted by living 
recombinant yeasts and did not result from cell lysis, a control strain producing an 
intracellular form of the model protein was contructed (without the leader sequence) 
and tested in similar experimental conditions. Some GST – V 5 H 6 was found in the 
ileum showing that cell lysis occurred in this compartment. At the end of digestion, 
the total amount of GST – V 5 H 6 released by the control strain represented 30% (1 
± 0.2 . g, n = 2) of the protein produced by WppGSTV 5 H 6 , showing a signifi cant 
contribution of cell lysis to the release of the protein in the TIM. 
For the fi rst time, the amount of heterologous proteins secreted by recombinant 
S. cerevisiae was evaluated throughout the upper digestive tract. Until now, the 
secretion effi ciency of recombinant microorganisms had never been directly quanti- 
fi ed throughout the digestive tract. The ability of recombinant strains to produce 
heterologous proteins in the digestive environment had been mainly demonstrated 
indirectly, following the biological effect of the protein: immune response (antibody 
production) [7, 15 – 17, 19, 20] , growth improvement [45] , or reduction in colitis 
symptoms [5] . Nevertheless, the ability of recombinant bacteria to initiate protein 
synthesis in situ has been reported in a few studies. Oozeer et al. [46] have shown 
that engineered Lactobacillus casei was able to initiate the synthesis of luciferase 
during its transit in the digestive tract of a human fl ora - associated mouse model. 
Steidler et al. [5] have demonstrated in mice the in situ synthesis of mouse IL - 10 by 
recombinant L. lactis , the viability of these microorganisms being required to achieve 
their therapeutic effect (see Section 5.5.1.2 ). They have further documented this 
result by showing the de novo synthesis of IL - 10 in the colon of IL - 10 . / . mice. 
Moreover, these authors have quantifi ed the amount of IL - 10 produced by the 
engineered L. lactis in two animal models, but only in a limited part of their digestive 
tract : (i) 7 ng of mouse IL - 10 was recovered in the colon of IL - 10 . / . mice, but 
the interleukin was not detectable in other areas of the gastrointestinal tract [5] and 
(ii) about 470 pg/mL of human IL - 10 was found in an ileal loop of a pig 4 h after 
injection of the recombinant bacteria [47] . Unlike what was previously obtained in 
the TIM with GST – V 5 H 6 , the concentrations of IL - 10 found in the digestive tract 
of the animals were much lower than that recovered in batch cultures [5, 47] . 
5.5.2.4 Conclusion 
For the fi rst time, the ability of engineered S. cerevisiae to carry out a bioconversion 
reaction [29] and initiate the synthesis and secrete protein compounds of various 
sizes [30] was shown throughout the upper gastrointestinal tract in human simulated 
digestive conditions. The CA4H specifi c activity of WRP45073A1 and the secretion 
level of GST – V 5 H 6 were surprisingly similar to that obtained in classical batch cul

ORAL FORMULATION OF RECOMBINANT YEASTS 577 
tures. This is particularly remarkable as the expression strategy of the model genes 
had not yet been adapted to the particular constraints of the digestive environment 
and promising for a future use of recombinant S. cerevisiae as host for biodrug 
development. 
5.5.3 ORAL FORMULATION OF RECOMBINANT YEASTS 
Once the scientifi c feasibility of the new drug delivery system was established, the 
development of pharmaceutical formulations allowing the oral administration of 
the genetically modifi ed S. cerevisiae was considered. Ideally, these oral drug dosage 
forms would improve both the survival and the heterologous activity of yeasts in 
the digestive environment. The following works were carried out only with the strain 
expressing the model P450. In a preliminary step, the effect of a preservation technique 
(lyophilization) and an immobilization procedure (entrapment in whey 
protein beads) on the survival rate and heterologous activity of the model strain 
WRP45073A1 was assessed in simulated digestive conditions. 
5.5.3.1 Freeze Drying of Recombinant Model Yeasts 
Freeze - Drying Conditions Freeze drying is a technique of dehydration commonly 
used for the formulation of drugs containing nonrecombinant Saccharomyces spp. 
[48 – 50] . Standard freeze - drying conditions derived from the literature [51 – 54] and 
our own experiments were applied for the lyophilization of the genetically modifi ed 
model yeasts. The effect of cryoprotectants was further investigated because it 
appears as one of the most important parameters during lyophilization [51, 52] . 
Following galactose induction of the heterologous CYP73A1, yeasts in the beginning 
of their stationary growth phase (10 9 cells/mL) were lyophilized in suspensions 
of trehalose 10% w/v, maltose 10% w/v, lactose 10% w/v, or a mixture of 5% w/v 
milk proteins and 10% w/v trehalose. The parameters of lyophilization are summarized 
in Table 4 [55] . 
Saccharomyces cerevisiae WRP45073A1 survives freeze drying and yeast survival 
rates were dependent on the nature of the cryoprotectants: 13.1 ± 1.8%, 9.5 ± 6.0%, 
TABLE 4 Parameters of Lyophilization Used for Recombinant Model Yeast 
WRP45073A1 
Recombinant yeasts Growth phase: beginning of stationnary growth phase 
Cell concentration in freeze - drying fl asks: 10 9 cells/mL 
Cryoprotectants 10% w/v trehalose 
10% w/v maltose 
10% w/v lactose 
5% w/v milk proteins and 10% w/v trehalose 
Control: physiological water 
Freeze - drying conditions Volume of sample: 5 mL 
Cooling rate: 1 ° C/min 
Condenser plate temperature: . 40 ° C 
Time of lyophilization: 24 h 
Heating temperature (secondary drying): 23 ° C 

578 IN VITRO VALIDATION AND ORAL FORMULATION 
7.7 ± 4.6%, and 7.1 ± 4.0% ( n = 5) for the milk protein – trehalose mix, lactose, 
maltose, and trehalose, respectively [55] . The protective effect of trehalose [51 – 53] 
and maltose [52] (but not that of lactose) compared to physiological water (survival 
rate 0.3 ± 0.2%, n = 5) had already been shown in non genetically modifi ed S. cerevisiae 
. Several mechanisms have been proposed to explain this protective effect. 
One hypothesis is related to the ability of these carbohydrates to form a glassy 
structure during drying, responsible for the long stability of biological materials [56] . 
The milk protein – trehalose mix led to a higher survival compared with trehalose 
alone ( p < 0.05). A similar result had already been observed by Abadias et al. [54] , 
who showed an improvement in the viability of another yeast, Candida sake , from 
7 to 29% when 5% skim milk is used in combination with 10% trehalose. 
Infl uence of Cryoprotectants on Viability and Heterologous Activity of 
Lyophilized Yeasts in Simulated Gastrointestinal Conditions To evaluate the 
infl uence of cryoprotectants on both the survival rate and CA4H activity of 
WRP45073A1 in simulated gastrointestinal conditions, 10 10 viable freeze - dried 
yeasts and 200 . mol of trans - cinnamic acid were simultaneously introduced into the 
TIM. Yeast cells were lyophilized in the presence of the milk protein – trehalose mix, 
trehalose, lactose, or maltose, as previously explained (see above). The freeze - dried 
samples were introduced into the artifi cial stomach suspended in 300 mL of yeast 
culture medium without any storage period. The number of viable cells introduced 
into the TIM was determined from previously obtained survival rates (cf. Section 
5.5.3.1 ). 
Viability of Freeze - Dried Yeasts in TIM Freeze - dried yeasts showed a high tolerance 
to gastric and small intestinal conditions. At the end of digestion (240 min), 
61.5 ± 0.7%, 59.9 ± 3.8%, 56.3 ± 5.4%, and 55.6 ± 6.0% ( n = 3) of the ingested 
cells were recovered in the ileal effl uents of the TIM, following freeze drying in 
the presence of the milk protein – trehalose mix, maltose, lactose, and trehalose, 
respectively (Figure 5 ). When the yeasts remaining in the residual chyme were 
added ( t = Tf), 84.7 ± 3.5%, 87.0 ± 6.4%, 83.7 ± 6.2%, and 70.7 ± 9.2% ( n = 3) 
survival percentages were found, respectively. Nevertheless, whatever the protective 
agent, the cumulative ileal delivery of freeze - dried yeasts remained signifi - 
cantly lower ( p < 0.05) than that of the nonabsorbable marker (see Section 5.5.2.2 ) 
and that of nondried ( “ native ” ) yeasts. The higher sensitivity to digestive conditions 
of freeze - dried cells compared with native ones may be linked to the damage 
caused to cells during drying and rehydration, resulting in increased membrane 
permeability [57] . 
In addition, no signifi cant difference was observed between the ileal recovery 
profi les of yeasts with the various cryoprotectants, showing their lack of infl uence 
on the survival of freeze - dried WRP45073A1 in the TIM. 
A few studies have evaluated the survival rate of freeze - dried S. cerevisiae spp. 
in human volunteers following their oral administration. Nevertheless, comparison 
between in vitro results in the TIM and these in vivo data is hampered by the fact 
that yeast survival had been evaluated only in feces (and not at the end of the ileum) 
after a single or multiple oral administration of the microorganisms. Klein et al. [49] 
found a fecal recovery of 0.12 ± 0.04% ( n = 8) after a single dose of 1 g of S. boulardii 
[10 10.4 colony - forming units (CFU)] to healthy volunteers, and Blehaut et al. [50] 
measured a steady - state fecal recovery of 0.36 ± 0.31% ( n = 8) after oral administra

ORAL FORMULATION OF RECOMBINANT YEASTS 579 
tion of 1 g of S. boulardii for 15 days. These survival rates in feces are much lower 
than those obtained in the ileal effl uents of the TIM. However, a recent work [29] 
has shown that native WRP45073A1 yeasts are very sensitive to colonic conditions: 
Yeast viability in an artifi cial digestive system reproducing the human large intestine 
[58] was only 1.2 ± 0.4% ( n = 3) after 12 h and no more yeast could be detected 
following 24 h fermentation. 
Heterologous Activity of Freeze - Dried Yeasts in TIM The ability of freeze - dried 
recombinant yeasts to catalyze the bioconversion of trans - cinnamic acid into p - coumaric 
acid in the in vitro system was shown whatever the tested protectant. At the 
end of the experiment, conversion rates of 24.2 ± 1.0%, 17.7 ± 2.2%, 15.1 ± 3.3%, 
and 16.5 ± 0.7% ( n = 3) were found for the yeasts lyophilized in the presence of 
milk proteins plus trehalose, maltose, lactose, and trehalose, respectively (Figure 6 ). 
During all the in vitro digestion, the CA4H activity of freeze dried cells remained 
signifi cantly lower ( p < 0.01) than that of native ones. This lower activity could result 
from the P45073A1 damage during freeze drying, probably aggravated by the membranous 
location of the enzyme. Among those tested, the cryoprotectant that allowed 
the highest CA4H activity was the milk protein – trehalose mix ( p < 0.05). 
Conclusion Although the impact of freeze drying on both the survival rate and 
heterologous activity of yeasts in the artifi cial digestive system was found to be 
adverse, lyophilization appears to be a convenient technique for the dehydration of 
recombinant S. cerevisiae . Among the tested cryoprotectants, the association of milk 
proteins and trehalose was the most effi cient to maintain the CA4H activity of 
FIGURE 5 Effect of cryoprotectants on survival rate of WRP45073A1 in TIM. The cumulative 
ileal deliveries of viable freeze - dried and native yeasts and that of the nonabsorbable 
marker are represented. At the end of digestion, the percentages obtained in the cumulative 
ileal effl uents (0 – 240 min) and the residual digestive content are added ( t = Tf). Results are 
expressed as mean percentages ± SD ( n = 3) of intake. Signifi cantly different from the marker 
( t = Tf) at p < 0.05 ( * ), p < 0.01 ( * * ) and p < 0.001 ( * * * ). (Reprinted with permission from 
Blanquet et al., European Journal of Pharmaceutics and Biopharmaceutics , 61, 37, 2006. 
Copyright 2006 by Elsevier.) 
0 
20 
40 
60 
80 
100 
120 
0 60 120 180 240 
Time of digestion (min) 
Cumulative ileal delivery of viable yeasts 
(% of intake) 
Marker Native yeasts 
Milk prot + treh Lactose 
Maltose Trehalose 
Tf
*** ** 
**

580 IN VITRO VALIDATION AND ORAL FORMULATION 
recombinant model yeasts in the stringent digestive conditions. This study also gives 
an example of the usefulness of the TIM in the prescreening of pharmaceutical 
excipients, such as cryoprotectants. 
5.5.3.2 Immobilization of Recombinant Model Yeasts in Whey Protein Beads 
Cell Immobilization Among the available techniques, the entrapment in gel beads 
is frequently used for the immobilization of living cells in food sciences [59, 60] 
because of its simplicity and low cost. This technique has been recently extended to 
microorganisms with probiotic activity with the aim of increasing their survival in 
the human digestive environment and particularly in the stomach [61, 62] . 
Whey proteins have been recently considered a potential alternative to the commonly 
used alginate [59, 61 – 63] for the production of gel beads. They have been 
used to entrap drugs such as retinol [64] and living microorganisms such as bifi dobacteria 
[65] , but until now not yeasts. A new immobilization system using whey 
proteins was then developed for entrapping the recombinant model yeasts 
WRP45073A1 in order to ensure their oral administration [66] . 
The formation of beads is a two - step process based on the cold gelation of whey 
proteins in the presence of divalent cations, such as Ca 2+ [67] . Briefl y, the whey 
protein isolate (WPI) solution (10% w/v in deionized water) was (i) adjusted at pH 
7 to favor the apparition of negative charges implied in ionic bounds with Ca 2+ ions 
and (ii) heated (80 ° C, 45 min) to denaturate the proteins. Recombinant cells in the 
beginning of their stationnary growth phase were suspended in a sterile solution of 
FIGURE 6 Effect of cryoprotectants on CA4H activity of WRP45073A1 in TIM. The 
CA4H activity of freeze - dried and native yeasts was evaluated in the overall TIM. Values are 
expressed as mean percentages ± SD ( n = 3) of initial trans - cinnamic acid converted into 
p - coumaric acid. Signifi cantly different from native yeasts ( t = 240 min) at p < 0.01 ( * * ). Signifi 
cantly different from milk proteins/trehalose group ( t = 240 min) at p < 0.05 ( + ) and p < 
0.01 ( ++ ). (Reprinted with permission from Blanquet et al., European Journal of Pharmaceutics 
and Biopharmaceutics , 61, 38, 2006. Copyright 2006, by Elsevier.) 
0
5 
10 
15 
20 
25 
30 
35 
40 
45 
50 
Time of digestion (min) 
Trans-cinnamic acid conversion (%) 
Native yeasts Milk prot + treh Maltose Lactose Trehalose 
** ++ 
+
+ 
** 
** 
** 
0 60 120 180 240 300

ORAL FORMULATION OF RECOMBINANT YEASTS 581 
10% w/v lactose (fi nal concentration 10 9 cells/mL) and added to denaturated WPI 
solution (7% v/v). The extrusion of the mixture through a needle led to the production 
of droplets forming gel beads in a calcium bath (0.1 M CaCl 2 ). This protocol 
allows the obtaining of spherical beads (diameter 2 605 ± 18 . m, n = 3) with an 
homogeneous distribution of yeasts through the matrix (1.15 . 10 6 viable cells per 
bead) [66] . The lack of infl uence of the immobilizing procedure on the viability of 
yeasts was also shown [66] . 
Gastric Digestion Protocol A most sought - after property of gel beads is their 
potential resistance to gastric conditions. Authors have already shown in vitro 
that beads resulting from the cold - induced gelation of a whey protein – oil emulsion 
[64] or a whey protein – polysaccharide mix [65] were gastroresistant. To further 
investigate the involvement of whey proteins in the gastroresistance of beads, the 
behavior of entrapped WRP45073A1 yeasts was followed in simulated gastric 
conditions. 
The human gastric environment was reproduced using a simple in vitro model 
adapted from that initially developped by Yvon et al. [68] . The main parameters of 
gastric digestion are reproduced according to in vivo data: decrease of pH, pepsin 
supply, body temperature, mixing, and gastric emptying (Figure 7 ). This system was 
validated by studies on the digestability of milk proteins (unpublished data). In the 
present work, it was programmed to reproduce gastric conditions of the adult after 
the intake of a glass of milk. Initially, 10 10 viable entrapped WRP45073A1 cells and 
200 . mol of trans - cinnamic acid were simultaneously introduced into the artifi cial 
stomach, suspended in 300 mL of yeast culture medium. Table 5 summarizes the 
parameters of in vitro gastric digestion. 
Release of Yeasts from Beads in Simulated Gastric Conditions The release of 
entrapped WRP45073A1 cells from whey protein beads was followed in the artifi cial 
FIGURE 7 Gastric digestive system. 
pH 
temps 
pH meter Two-way 
valve 
37°C 
Computer 
system 
Artificial stomach 
in water bath 
Gastric 
effluents 
Pump H2O HCl 0.2 M Pepsin 3804 IU/mL 

582 IN VITRO VALIDATION AND ORAL FORMULATION 
TABLE 5 Parameters of In Vitro Digestion in Artifi cial Stomach When Simulating 
Gastric Conditions of Adult after Intake of Glass of Milk 
Initial “ meal ” 
Volume 300 mL (constant during all digestion) 
Pepsin 75 IU/mL 
Acidifi cation 
Time (min)/pH 0/6.5, 60/2.1 
15/4.4, 75/1.9 
30/3.2, 90/1.7 
45/2.5, 120/1.6 
Flow rate 2 mL/min HCl 0.2 M if necessary 
Exponential base e = 1.04 
Pepsin supply 
Time (min)/pepsine (IU/mL) 0/77.6, 60/139.2 
15/99.7, 75/146.7 
30/117.1, 90/152.8 
45/129.7, 120/163.0 
Flow rate From 13 ( t = 0) to 1 ( t = 120 min) mL/min pepsin 
3804 IU/mL 
Exponential base e = 1.03 
Gastric emptying 
Flow rate From 10 ( t = 0) to 3 ( t = 120 min) mL/min 
Exponential base e = 1.03 
Time of digestion 120 min 
stomach. During the fi rst 60 min of gastric digestion, a few percentage points (2.2 ± 
0.9%, n = 3) of initial entrapped yeasts was recovered in the gastric medium (Figure 
8 ). This low percentage cannot be explained by cell death since control experiments 
showed the high survival rate of free yeasts (about 90%) during all the digestion 
[66] . These results are in agreement with those of Beaulieu et al. [64] , who have 
observed that only 5 – 10% of the incorporated retinol was released from the whey 
protein – oil matrix following 30 min incubation in HCl 0.1 M and pepsin 24 mg/L. 
The low release of yeasts in the fi rst hour of digestion indicates that the beads are 
resistant to acidifi cation until pH 2 (cf. Table 5 ) and pepsin attack, which implies 
that they might cross the gastric barrier in humans. 
From 60 min digestion, the percentage of released yeasts increased regularly to 
reach 39 ± 5% ( n = 3) at 120 min. This phenomenon might be explained by two 
hypotheses: (i) a swelling of beads due to an increase in the acidity of the medium 
or (ii) a degradation of the matrix resulting from a raise in pepsin concentration. 
Complementary studies conducted to further evaluate the effect of pH and pepsin 
on beads have shown that pepsin has no effect on the protein matrix whatever the 
tested pH [66] . As already suggested by other authors [69] , this resistance to enzyme 
attack might be due to the formation of hydrophobic interactions between aromatic 
amino acids of . - lactoglobulin, the major whey protein. On the contrary, incubation 
at pH 2 led to an increase in the diameter of beads, certainly due to high electrostatic 
repulsive forces (between positive charges of protoned amino acids and Ca 2+ ), which 
induced a raise in the pore size. In conclusion, the release of yeasts observed from 

ORAL FORMULATION OF RECOMBINANT YEASTS 583 
60 min digestion is provoked by acidic conditions rather than by enzymatic degradation 
of beads. 
Infl uence of Entrapment on Heterologous Activity of Yeasts in Simulated Gastric 
Conditions In order to evaluate the infl uence of the entrapment process of the 
CA4H activity of WRP45073A1, the heterologous activity of free and entrapped 
yeasts was followed in the gastric system under similar experimental conditions. In 
both cases, p - coumaric acid was detected in the gastric medium as soon as 15 min 
after the beginning of the experiment (Figure 9 ). This implies that both trans - 
cinnamic and p - coumaric acid could diffuse through bead pores. 
During all the digestion, the CA4H activity of entrapped yeasts was signifi cantly 
( p < 0.05) higher than that of free ones (expected for t = 15 min and t = 75 min). At 
120 min, 63.4 ± 1.6% ( n = 3) of initial trans - cinnamic acid was converted into 
p - coumaric acid for immobilized yeasts versus 51.5 ± 1.8% ( n = 3) for control yeasts. 
This phenomenon was particularly marked from 30 to 60 min of digestion when a 
very low amount of recombinant yeasts was released from beads. As suggested by 
Bienaim e et al. [70] , beads might create a microenvironment (e.g., a buffer effect 
toward low pH or enzyme attack) favoring the heterologous activity of yeasts. In a 
similar way, the microenvironment resulting from the presence of an alginate matrix 
improves the invertase activity of recombinant S. cerevisiae in batch cultures [71] . 
Conclusion This preliminary work reveals whey proteins as a convenient material 
for immobilizing recombinant yeasts. Gel beads were resistant to acidifi cation until 
pH 2 and pepsin attack, suggesting that they should cross the gastric barrier in 
humans. Moreover, the presence of the protein matrix seemed to create “ microconditions 
” that favor the heterologous activity of entrapped yeasts. 
FIGURE 8 Release of WRP45073A1 cells from whey protein beads in simulated gastric 
conditions. Results are expressed as mean percentages ± SD ( n = 3) of initial entrapped yeasts. 
(Reprinted with permission from Hebrard et al., Journal of Biotechnology , in press. Copyright 
2006 by Elsevier.) 
0 
0 
10 
20 
30 
40 
50 
Time of digestion (min) 
Viable yeasts released (% of initially entrapped) 
20 40 60 80 100 120 140

584 IN VITRO VALIDATION AND ORAL FORMULATION 
5.5.4 GENERAL CONCLUSION AND FUTURE DEVELOPMENTS 
Using genetically engineered microorganisms as new delivery vehicles to the gut is 
an important challenge for the development of innovative drugs. A potential application 
directly issued from the present work is the development of drug delivery 
systems based on orally administered yeasts carrying out a bioconversion reaction 
or secreting compounds directly in the human digestive tract. 
Soon, the choice of candidate genes as well as the most appropriate dosage forms 
will be made according to the therapeutic target. Oral formulations will be optimized 
in order to (i) control the release of yeasts according to their action site in 
the gastrointestinal tract, (ii) maximize the heterologous activity of yeasts (by addition 
of the appropriate substrate and/or inductor), and (iii) ensure a stability of both 
yeasts and pharmaceutical dosage forms before administration to the patient. Of 
course, heterologous gene expression strategies have to be tailored for a safe use in 
humans, the presence of mobilizable vectors, antibiotic selection markers, and bacterial 
sequences liable to promote gene transfer to host microfl ora being prohibited. 
In addition, environmental confi nement of recombinant cells has to be achieved by 
introducing a suicide process that triggers the elimination of the microorganisms 
upon leaving the digestive tract. Two types of biological containment systems may 
be considered [1, 3] : (i) the active system, which should provide control of the 
recombinant microorganism dissemination through the conditional production of a 
toxic protein [72 – 74] , and (ii) the passive system, which could render the cell growth 
dependent on the complementation of an auxotrophy or other gene defects [75, 76] . 
FIGURE 9 Infl uence of immobilization on CA4H activity of WRP45073A1 in simulated 
gastric conditions. The CA4H activity of entrapped and free yeasts was evaluated in the arti- 
fi cial gastric system. Results are expressed as mean percentages ± SD ( n = 3) of trans - cinnamic 
acid converted into p - coumaric acid. Signifi cantly different from free yeasts at p < 0.05 ( * ). 
(Reprinted with permission from Hebrard et al., Journal of Biotechnology , in press. Copyright 
2006 by Elsevier.) 
0 
10 
20 
30 
40 
50 
60 
70 
Time of digestion (min) 
Trans-cinnamic acid conversion (%) 
Entrapped yeasts 
Free yeasts 
* 
* 
* 
* 
* 
0 20 40 60 80 100 120 140

Steidler et al. [47] have already developed and validated in pigs a passive containment 
system for the L. lactis expressing human IL - 10 by deleting the thymidylate 
synthase gene which is essential for their growth (the resulting strain being dependent 
on thymidine or thymine). 
The present study also shows the particular interest of the TIM in drug development 
and testing. This artifi cial system will constitute a powerful alternative to 
animal experimentation during all preclinical phases of biodrug development. The 
in vitro model can aid in the selection of pharmaceutical formulations, ensuring both 
the release of yeasts directly at their action site and their optimal activity. The effi - 
ciency of newly developed molecular tools (e.g., promoters, selection markers, and 
vectors) can also be evaluated to optimize the functionality of recombinant strains 
in the digestive environment. As the mucosal layer is not involved in the actual 
confi guration of the TIM, this system may be used in combination with intestinal 
cells in culture (e.g., Caco - 2) to study the mucosal transport and metabolism of the 
active compounds produced by recombinant yeasts. Moreover, experiments in a 
large intestinal model [58] , complementary of the gastric and small intestinal system, 
could provide necessary data on the biological safety of engineered microorganisms. 
For example, the potential gene transfer to the human fl ora can be studied and the 
cell death outside of the digestive tract can be checked to ensure there is no dissemination 
in the environment. 
This study opens up new opportunities in the development of new drug delivery 
vectors based on engineered living yeasts for the prevention or treatment of various 
diseases in human. 
REFERENCES 
1. Blanquet , S. , Marol - Bonnin , S. , Beyssac , E. , Pompon , D. , Renaud , M. , and Alric , M. ( 2001 ), 
The Biodrug concept: An innovative approach to therapy , Trends Biotechnol. , 19 , 
393 – 400 . 
2. Corthier , G. , and Renault , P. ( 1999 ), Future directions for research on biotherapeutic 
agents: Contribution of genetic approaches on lactic acid bacteria , in Elmer , G. W. , 
McFarland , L. , Surawicz , C. , Eds., Biotherapeutic Agents and Infection Diseases , Humana , 
Totowa, NJ , pp. 269 – 304 . 
3. Steidler , L. ( 2003 ), Genetically engineered probiotics, Best Practise Res . Clin. Gastroenterol. 
, 17 , 861 – 876 . 
4. Chatel , J. M. , Langella , P. , Adel - Patient , K. , Commissaire , J. , Wal , J. M. , and Corthier , G. 
( 2001 ), Induction of mucosal immune response after intranasal or oral inoculation of mice 
with Lactococcus lactis producing bovine beta - lactoglobulin , Clin. Diag. Lab. Immunol. , 
8 , 545 – 551 . 
5. Steidler , L. , Hans , W. , Schotte , L. , Neirynck , S. , Obermeier , F. , Falk , W. , Fiers , W. , and 
Remaut , E. ( 2000 ), Treatment of murine colitis by Lactococcus lactis secreting 
Interleukin - 10 , Science , 289 , 1352 – 1355 . 
6. Schreuder , M. P. , Deen , C. , Boersma , W. J. A. , Pouwels , P. H. , and Klis , F. M. ( 1996 ), Yeast 
expressing hepatitis B virus surface antigen determinants on its surface: Implications for 
a possible oral vaccine , Vaccine , 14 , 383 – 388 . 
7. Reveneau , N. , Goeffroy , M. C. , Locht , C. , Chagnaud , P. , and Mercenier , A. ( 2002 ), Comparison 
of the immune responses induced by local immunizations with recombinant 
REFERENCES 585

586 IN VITRO VALIDATION AND ORAL FORMULATION 
Lactobacillus plantarum producing tetanus toxin fragment C in different cellular locations 
, Vaccine , 20 , 1769 – 1777 . 
8. Drouault , S. , Corthier , G. , Ehrlich , S. D. , and Renault , P. ( 1999 ), Survival, physiology, 
and lysis of Lactococcus lactis in the digestive tract , Appl. Environ. Microbiol. , 65 , 
4881 – 4886 . 
9. Fahl , W. E. , Loo , D. , and Manoharan , H. ( 1999 ), Chemoprotective bacterial strains, International 
Patent WO 99/27953 . 
10. Baranov , V. S. , Ivaschenko , T. , Bakay , B. , Aseev , M. , Belotserkovskaya , R. , Baranova , H. , 
Malet , P. , Perriot , J. , Mouraire , P. , Baskakov , V. N. , Savitskyi , G. A. , Gorbushin , S. , 
Deyneka , S. I. , Michnin , E. , Barchuck , A. , Vakharlovsky , V. , Pavlov , G. , Shilko , V. I. , 
Guembitzkaya , T. , and Kovaleva , L. ( 1996 ), Proportion of the GSTM1 0/0 genotype in 
some Slavic populations and its correlation with cystic fi brosis and some multifactorial 
diseases , Hum. Genet. , 97 , 516 – 520 . 
11. Drouault , S. , Juste , C. , Marteau , P. , Renault , P. , and Corthier , G. ( 2002 ), Oral treatment 
with Lactococcus lactis expressing Staphylococcus hyicus lipase enhances lipid digestion 
in pigs with induced pancreatic insuffi ciency , Appl. Environ. Microbiol. , 68 , 3166 – 3168 . 
12. Chang , T. M. , and Prakash , S. ( 1998 ), Therapeutic uses of microencapsulated genetically 
engineered cells , Mol. Med. Today , 4 , 221 – 227 . 
13. Prakash , S. , and Chang T. M. S. ( 2000 ), In vitro and in vivo uric acid lowering by artifi cial 
cells containing microencapsulated genetically engineered E. coli DH5 cells , Int. J. Artif. 
Organs , 23 , 429 – 435 . 
14. Pouwels , P. H. , Leer , R. J. , Shaw , M. , Bak - Glashouwer , M. J. H. D. , Tielen , F. D. , Smit , E. , 
Martinez , B. , Jore , J. , and Conway , P. L. ( 1998 ), Lactic acid bacteria as antigen delivery 
vehicles for oral immunization purposes , Int. J. Food Microbiol. , 41 , 155 – 167 . 
15. Robinson , K. , Chamberlain , L. M. , Schofi eld , K. M. , Wells , J. M. , and Le Page , R. W. F. 
( 1997 ), Oral vaccination of mice against tetanus with recombinant Lactococcus lactis , Nat. 
Biotechnol. , 15 , 653 – 657 . 
16. Nayak , A. R. , Tinge , S. A. , Tart , R. C. , McDaniel , L. S. , and Briles , D. E. , 3rd , and Curtiss , 
R. ( 1998 ), A live recombinant avirulent oral Salmonella vaccine expressing pneumococcal 
surface protein A induces protective responses against Streptococcus pneumoniae , Infect. 
Immun. , 66 , 3744 – 3751 . 
17. Corthesy , B. , Boris , S. , Isler , P. , Grangette , C. , and Mercenier , A. ( 2005 ), Oral imunization 
of mice with lactic acid bacteria producing Helicobacter pylori urease B subunit partially 
protects against challenge with Helicobacter felis , J. Infect. Dis. , 192 , 1441 – 1449 . 
18. Barron , M. A. , Blyveis , N. , Pan , S. C. , and Wilson , C. C. ( 2006 ), Human dendritic cell 
interactions with whole recombinant yeasts: Implication for HIV - 1 vaccine development , 
J. Clin. Immunol. , 26 , 251 – 264 . 
19. Steger , K. K. , Valentine , P. J. , Heffron , F. , So , M. , and Pauza , C. D. ( 1999 ), Recombinant, 
attenuated Salmonella typhimurium stimulate lymphoproliferative responses to SIV 
capsid antigen in rhesus macaques , Vaccine , 17 , 923 – 932 . 
20. Adel - patient , K. , Ah - Leung , S. , Creminon , C. , Nouaille , S. , Chatel , J. M. , Langella , P. , and 
Wal , J. M. ( 2005 ), Oral administration of recombinant Lactococcus lactis expressing 
bovine beta - lactoglobulin partially prevents mice from sensitisation , Clin. Exp. Allergy , 
35 , 539 – 546 . 
21. Mercenier , A. , M u ller - Alouf , H. , and Grangette , C. ( 2000 ), Lactic acid bacteria as live 
vaccines , Curr. Issues Mol. Biol. , 2 , 17 – 25 . 
22. Bumann , D. , Hueck , C. , Aebisher , T. , and Meyer , T. F. ( 2000 ), Recombinant live Salmonella 
spp . for human vaccination against pathogens , FEMS Immunol. Med. Microbiol. , 
27 , 357 – 364 . 

23. Steidler , L. ( 2002 ), In situ delivery of cytokines by genetically engineered Lactococcus 
lactis , Ant. Leeuwenh. , 82 , 323 – 331 . 
24. Steidler , L. ( 2001 ), Microbiological and immunological strategies for treatment of infl ammatory 
bowel disease , Microbes Infect. , 3 , 1157 – 1166 . 
25. Braat , H. , Rottiers , P. , Hommes , D. W. , Huyghebaert , N. , Remaut , E. , Remon , J. P. , Van 
Deventer , S. J. , Neirynck , S. , Peppelenbosch , M. P. , and Steidler , L. ( 2006 ), A phase I trial 
with transgenic bacteria expressing interleukin - 10 in Crohn ’ s disease , Clin. Gastroenterol. 
Hematol. , 4 , 754 – 759 . 
26. Lewis , S. J. , and Freedman , A. R. ( 1998 ), Review article: The use of biotherapeutic agents 
in the prevention and treatment of gastrointestinal disease , Aliment. Pharmacol. Ther. , 
12 , 807 – 822 . 
27. Bergogne - Berezin , E. ( 2000 ), Treatment and prevention of antibiotic - associated diarrhea , 
Int. J. Antimicrobial. Agents , 16 , 521 – 526 . 
28. Urban , P. , Werck - Reichhart , D. H. , Teutsch , G. , Durst , F. , Regnier , S. , Kazmaier , M. , and 
Pompon , D. ( 1994 ), Characterization of recombinant plant cinnamate 4 - hydroxylase produced 
in yeast. Kinetic and spectral properties of the major plant P450 of the phenylpropanoid 
pathway , Eur. J. Biochem. , 222 , 843 – 850 . 
29. Blanquet , S. , Meunier , J. P. , Minekus , M. , Marol - Bonnin , S. , and Alric , M. ( 2003 ), Recombinant 
Saccharomyces cerevisiae expressing a P450 in artifi cial digestive systems: A model 
for biodetoxication in the human digestive environment , Appl. Environ. Microbiol. , 69 , 
2884 – 2892 . 
30. Blanquet , S. , Antonelli , R. , Laforet , L. , Denis , S. , Marol - Bonnin , S. , and Alric , M. ( 2004 ), 
Living recombinant Saccharomyces cerevisiae secreting proteins or peptides as a new 
drug delivery system in the gut , J. Biotechnol. , 110 , 37 – 49 . 
31. Minekus , M. , Marteau , P. , Havenaar , R. , and Huis in ’ t Veld , J. H. J. ( 1995 ), A multicompartmental 
dynamic computer - controlled model simulating the stomach and small intestine 
, ALTA , 23 , 197 – 209 . 
32. Marteau , P. , Minekus , M. , Havenaar , R. , and Huis in ’ t Veld , J. H. J. ( 1997 ), Survival of 
lactic acid bacteria in a dynamic model of the stomach and small intestine: Validation and 
the effects of the bile , J. Dairy Sci. , 80 , 1031 – 1037 . 
33. Blanquet , S. , Zeijdner , E. , Beyssac , E. , Meunier , J. P. , Denis , S. , Havenaar , R. , and 
Alric , M. ( 2004 ), A dynamic gastrointestinal system for studying the behavior of orally 
administered drug dosage forms under various physiological conditions , Pharm. Res. , 
21 , 585 – 591 . 
34. Minekus , M. , Jelier , M. , Xiao , J. Z. , Kondo , S. , Iwatsuki , K. , Kokubo , S. , Bos , M. , Dunnewind 
, B. , and Havenaar , R. ( 2005 ), Effect of partially hydrolyzed guar gum (PHGG) 
on the bioaccessibility of fat and cholesterol , Biosci., Biotechnol. Biochem. , 69 , 932 – 938 . 
35. Larsson , M. , Minekus , M. , and Havenaar , R. ( 1997 ), Estimation of the bioavailability of 
iron and phosphorus in cereals using a dynamic in vitro gastro - intestinal model , J. Sci. 
Food Agric. , 74 , 99 – 106 . 
36. Verwei , M. , Arkbage , K. , Havenaar , R. , Van den Berg , H. , Witth o ft , C. , and Schaafsma , G. 
( 2003 ), Folic acid and 5 - methyl - tetrahydrofolate in fortifi ed milk are bioaccessible as 
determined in a dynamic in vitro gastrointestinal model , J. Nutr. , 133 , 2377 – 2383 . 
37. Krul , C. A. M. , Luiten - Schuite , A. , Baan , R. , Verhagen , H. , Mohn , G. , Feron , V. , and 
Havenaar , R. ( 2000 ), Application of a dynamic in vitro gastrointestinal tract model to 
study the availability of food mutagens, using heterocyclic aromatic amines as model 
compounds , Food Chem. Toxicol. , 38 , 783 – 792 . 
38. Souliman , S. , Blanquet , S. , Beyssac , E. , and Cardot , J. M. ( 2006 ), A level A in vitro/in vivo 
correlation in fasted and fed states using different methods: applied to solid immediate 
release oral dosage form , Eur. J. Pharm. Sci. , 27 , 72 – 79 . 
REFERENCES 587

588 IN VITRO VALIDATION AND ORAL FORMULATION 
39. Elashoff , J. D. , Reedy , T. J. , and Meyer , J. M. ( 1982 ), Analysis of gastric emptying data , 
Gastroenterology , 83 , 1306 – 1312 . 
40. Heading , R. C. , Tothill , P. , McLoughlin , G. P. , and Shearman , D. J. C. ( 1976 ), Gastric emptying 
rate measurement in man. A double isotope scanning technique for simultaneous 
study of liquid and solid components of a meal , Gastroenterology , 71 , 45 – 50 . 
41. Chung , Y. C. , Kim , Y. S. , Shadchehr , A. , Garrido , A. , Macgregor , I. L. , and Sleisenger , 
M. H. ( 1979 ), Protein digestion and absorption in human small intestine , Gastroenterology 
, 76 , 1415 – 1421 . 
42. Castelli , F. , Uccella , N. , Trombetta , D. , and Saija , A. ( 1999 ), Differences between coumaric 
and cinnamic acids in membrane permeation as evidenced by time - dependent calorimetry 
, J. Agric. Food Chem. , 47 , 991 – 995 . 
43. Zarate , G. , Chaia , A. P. , Gonzalez , S. , and Oliver , G. ( 2000 ), Viability and beta - galactosidase 
activity of diary propionibacteria subjected to digestion by artifi cial intestinal fl uids , 
J. Food Protection , 63 , 1214 – 1221 . 
44. Zsebo , K. M. , Lu , H. S. , Fieschko , J. C. , Goldstein , L. , Davis , J. , Duker , K. , Suggs , S. V. , 
Lai , P. H. , and Bitter , G. A. ( 1986 ), Protein secretion from Saccharomyces cerevisiae 
directed by the prepro - a - factor leader region , J. Biolog. Chem. , 261 , 5858 – 5865 . 
45. Chen , C. M. , Cheng , W. T. K. , Chang , Y. C. , Chang , T. J. , and Chen H. L. ( 2000 ), Growth 
enhancement of fowls by dietary administration of recombinant yeast cultures containing 
enriched growth hormones , Life Sci. , 67 , 2102 – 2115 . 
46. Oozeer , R. , Goupil - Feuillerat , N. , Alpert , C. A. , Vande Guchte , M. , Anba , J. , Mengaud , J. , 
and Corthier , G. ( 2002 ), Lactobacillus casei is able to survive and initiate protein synthesis 
during its transit in the digestive tract of human fl ora - associated mice , Appl. Environ. 
Microbiol. , 68 , 3570 – 3574 . 
47. Steidler , L. , Neirynck , S. , Huyghebaert , N. , Snoeck , V. , Vermeire , A. , Goddeeris , B. , Cox , 
E. , Remon , J. P. , and Remaut , E. ( 2003 ), Biological containment of genetically modifi ed 
Lactococcus lactis for intestinal delivery of human interleukin - 10 , Nat. Biotechnol. , 21 , 
785 – 789 . 
48. Harms , H. K. , Bertele - Harms , R. M. , and Bruer - Kleis , D. ( 1987 ), Enzyme - substitution 
therapy with the yeast Saccharomyces cerevisiae in congenital sucrase - isomaltase defi - 
ciency , N. Engl. J. Med. , 316 , 1306 – 1309 . 
49. Klein , S. M. , Elmer , G. W. , McFarland , L. V. , Surawicz , C. M. , and Levy , R. H. ( 1993 ), 
Recovery and elimination of the biotherapeutic agent, Saccharomyces cerevisiae , in 
healthy human volunteers , Pharm. Res. , 10 , 1615 – 1619 . 
50. Blehaut , H. , Massot , J. , Elmer , G. W. , and Levy , R. ( 1989 ), Disposition kinetics of Saccharomyces 
boulardii in man and rat , Biopharm. Drug Dispos. , 10 , 353 – 364 . 
51. Berny , J. F. , and Hennebert , G. L. ( 1991 ), Viability and stability of yeast cells and fi lamentous 
fungus spores during freeze - drying: Effects of protectants and cooling rates , 
Mycologia , 83 , 805 – 815 . 
52. Lodato , P. , Segovia de Huergo , M. , and Buera , M. P. ( 1999 ), Viability and thermal stability 
of a strain of Saccharomyces cerevisiae freeze - dried in different sugars and polymer 
matrices , Appl. Microbiol. Biotechnol. , 52 , 215 – 220 . 
53. Diniz - Mendes , L. , Bernardes , E. , De Araujo , P. S. , Panek , A. D. , and Paschoalin , V. M. F. 
( 1999 ), Preservation of frozen yeast cells by trehalose , Biotechnol. Bioeng. , 65 , 572 – 578 . 
54. Abadias , M. , Benabarre , A. , Teixido , N. , Usall , J. , and Vias I. ( 2001 ), Effect of freeze - drying 
and protectants on viability of the biocontrol yeast Candida sake , Int. J. Food Microbiol. , 
65 , 173 – 182 . 
55. Blanquet , S. , Garrait , G. , Beyssac , E. , Perrier , C. , Denis , S. , H e brard , G , and Alric , M. 
( 2005 ), Effects of cryoprotectants on the viability and activity of freeze - dried recombi

nant yeasts as novel oral drug delivery systems assessed by an artifi cial digestive system , 
Eur. J. Pharm. Biopharm. , 61 , 32 – 39 . 
56. Aquilera , J. M. , and Karel , M. ( 1997 ), Preservation of biological materials under desiccation 
, Crit. Rev. Food Sci. Nutr. , 37 , 287 – 309 . 
57. Beker , M. J. , and Rapoport , A. I. ( 1987 ), Advances in Biochemical Engineering/Biotechnology 
, Springer , Berlin , pp. 128 – 171 . 
58. Minekus , M. , Smeets - Peter , M. , Bernalier , A. , Marol - BonnIn , S. , Havenaar , R. , Marteau , 
P. , Alric , M. , Fonty , G. , and Huis in ’ t Veld , J. H. J. ( 1999 ), A computer - controlled system 
to simulate conditions of the large intestine with peristaltic mixing, water absorption and 
absorption of fermentation products , Appl. Microbiol. Biotechnol. , 53 , 108 – 114 . 
59. Yadav , B. S. , Rani , U. , Dhamija , S. S. , Nigam , P. , and Singh , D. ( 1996 ), Process optimization 
for continuous ethanol fermentation by alginate - immobilized cells of Saccharomyces 
cerevisiae HAU - 1 , J. Basic Microbiol. , 36 , 205 – 210 . 
60. Tsen , J. H. , Lin , Y. P. , and King , V. A. E. ( 2004 ), Fermentation of Banana media by using 
. - carrageenan immobilized Lactobacillus acidophilus , Int. J. Food Microbiol. , 91 , 215 – 
222 . 
61. Lee , K. Y. , and Heo , T. R. ( 2000 ), Survival of Bifi dobacterium longum immobilized in 
calcium alginate beads in simulated gastric juices and bile salt solution , Appl. Environ. 
Microbiol. , 66 , 869 – 873 . 
62. Chandramouli , V. , Kailasapathy , K. , Peiris , P. , and Jones , M. ( 2004 ), An improved 
method of microencapsulation and its evaluation to protect Lactobacillus spp. in simulated 
gastric conditions , J. Microbiol. Methods , 56 , 27 – 35 . 
63. Lamas , C. M. , Bregni , C. , D ’ Aquino , M. , Degrossi , J. , and Firenstein , R. ( 2001 ), Calcium 
alginate microspheres of Bacillus subtilis , Drug Dev. Ind. Pharm. , 27 , 825 – 829 . 
64. Beaulieu , L. , Savoie , L. , Paquin , P. , and Subirade , M. ( 2002 ), Elaboration and characterization 
of whey protein beads by an emulsifi cation/cold gelation process: Application for the 
protection of retinol , Biomacromolecules , 3 , 239 – 248 . 
65. Gu e rin , D. , Vuillemard , J. C. , and Subirade , M. ( 2003 ), Protection of bifi dobacteria 
encapsulated in polysaccharide - protein gel beads against gastric juice and bile , J. Food 
Prot. , 66 , 2076 – 2084 . 
66. H e brard , G. , Blanquet , S. , Beyssac , E. , Remondetto , G. , Subirade , M. , and Alric , M. 
( 2006 ) , Use of whey protein beads as a new carrier system for recombinant yeasts in 
human digestive tract. J. Biotechnology , 127 , 151 – 160 . 
67. Hongsprabhas , P. , and Barbut , S. ( 1998 ), Ca 2+ - induced cold gelation of whey protein 
isolate: Effect of two - stage gelation , Food Res. Int. , 30 , 523 – 527 . 
68. Yvon , M. , Beucher , S. , Scanff , P. , Thirouin , S. , and Pelissier , J. P. ( 1992 ), In vitro simulation 
of gastric digestion of milk proteins: Comparison between in vitro and in vivo data , 
J. Agric. Food Chem. , 40 , 239 – 244 . 
69. Remondetto , G. E. , and Subirade , M. ( 2003 ), Molecular mechanisms of Fe 2+ - induced - . - 
lactoglobuline cold gelation , Biopolymers , 69 , 461 – 469 . 
70. Bienaim e , C. , Barbotin , J. N. , and Nava - Saucedo , J. E. ( 2003 ), How to build an adapted 
and bioactive cell microenvironment? A chemical interaction study of the structure of 
Ca - alginate matrices and their repercussion on confi ned cells , J. Biomed. Mater. Res., A , 
67 , 376 – 388 . 
71. Rossi - Alva , J. C. , and Miguez Rocha - Lea o , M. H. ( 2003 ), A strategic study using mutant - 
strain entrapment in calcium alginate for the production of Saccharomyces cerevisiae cells 
with high invertase activity , Biotechnol. Appl. Biochem. , 38 , 43 – 51 . 
72. Molin , S. ( 1993 ), Environmental potential of suicide genes , Curr. Opin. Biotechnol. , 4 , 
299 – 305 . 
REFERENCES 589

590 IN VITRO VALIDATION AND ORAL FORMULATION 
73. Diaz , E. , Munthali , M. , De Lorenzo , V. , and Timmis , K. N. ( 1994 ), Universal barrier to 
lateral spread of specifi c genes among microorganisms , Mol. Microbiol. , 13 , 855 – 861 . 
74. Kaplan , D. L. , Mello , C. , Sano , T. , Cantor , C. , and Smith , C. ( 1999 ), Steptavidin - based 
containment systems for genetically engineered microorganisms , Biomol. Eng. , 16 , 
135 – 140 . 
75. Hols , P. , Defrenne , C. , Ferain , T. , Derzelle , S. , Delplace , B. , and Delcour , J. ( 1997 ), The 
alanine racemase gene is essential for growth of Lactobacillus plantarum , J. Bacteriol. , 
179 , 3804 – 3807 . 
76. Fu , X. , and Xu , J. G. ( 2000 ), Development of a chromosome - plasmid balanced lethal 
system for Lactobacillus acidophilus with thyA gene as selective marker , Microbiol. 
Immunol. , 44 , 551 – 556 . 

591 
5.6 
NASAL DELIVERY OF PEPTIDE AND 
NONPEPTIDE DRUGS 
Chandan Thomas and Fakhrul Ahsan 
Texas Tech University Health Sciences Center, Amarillo, Texas 
Contents 
5.6.1 Introduction 
5.6.2 Nasal Anatomy and Physiology 
5.6.2.1 Structure of Nasal and Olfactory Mucosa 
5.6.2.2 Nasal Vasculature 
5.6.2.3 Enzymes and pH 
5.6.3 Factors Infl uencing Nasal Drug Absorption 
5.6.3.1 Physiological Factors 
5.6.3.2 Nasal Mucociliary Clearance 
5.6.3.3 Pathological Condition of Nose 
5.6.3.4 Dose Volume and Site of Deposition 
5.6.3.5 Physicochemical Properties of Drugs 
5.6.3.6 Type of Delivery Device 
5.6.4 Animal Models for Nasal Absorption Studies 
5.6.5 Enhancement of Intranasal Drug Absorption 
5.6.6 Nasal Delivery of Peptide and High - Molecular Weight Drugs 
5.6.6.1 Insulin 
5.6.6.2 Calcitonin 
5.6.6.3 Low - Molecular - Weight Heparins 
5.6.6.4 Azetirelin 
5.6.6.5 Growth Hormones 
5.6.7 Nasal Delivery of Nonpeptide Molecules 
5.6.7.1 Morphine 
5.6.7.2 Benzodiazepines 
5.6.7.3 Buprenorphine 
5.6.7.4 Hydralazine 
5.6.7.5 Nitroglycerin 
5.6.7.6 Propranolol and Other . - Adrenergic Blocking Agents 
5.6.7.7 Sex hormones 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

592 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
5.6.7.8 17 . - Estradiol (E 2 ) 
5.6.7.9 Testosterone 
5.6.8 Nose: Option for Delivery of Drugs to Central Nervous System 
5.6.9 Nasal Delivery of Vaccines 
5.6.9.1 Nasal Vaccines: Ideal Noninvasive Route 
5.6.9.2 Immunity after Intranasal Immunization 
5.6.9.3 Need for Adjuvants 
References 
5.6.1 INTRODUCTION 
The delivery of drugs via the nasal route has been practiced since ancient times; for 
example, psychotropic and hallucinogenic agents have been used as snuff in many 
parts of the world for hundreds and possibly thousands of years. More recently, 
especially over the past two decades, intranasal drug delivery has shown great 
promise in various fi elds of medical practice. At present, a number of conditions 
(e.g., rhinitis, migraine, nasal congestion, and osteoporosis) are being treated successfully 
with nasal formulations. In addition, an ever - increasing number of nasally 
delivered, systemically acting drugs are in the pipeline. Recently this form of therapy 
received encouragement with the approval of FluMist (MedImmuneVaccines, 
Gaithersburg, MD), an intranasal vaccine against Haemophilus infl uenzae, the infl uenza 
virus. This vaccine is the fi rst to be given by the nasal route as a mist rather 
than by injection. With its approval, many of the pharmaceutical companies, including 
some giants of the pharmaceutical industry, are increasingly looking toward the 
area of nasal drug delivery. The market for such therapy in 2005 is reported to have 
reached $ 2.4 billion. With 16 of the 20 major pharmaceutical companies conducting 
active programs in this area, the fi eld of nasal drug delivery is expected to grow at 
an estimated 33% annually [1] . As reported by Koch [2] , the U.S. drug delivery 
market in 2005 was somewhere in excess of $ 50 billion and has been predicted to 
be around $ 67 billion by the year 2009, whereas the nasal drug delivery market is 
expected to be valued at $ 9 billion by 2008 [1 – 3] . Nasal administration — as compared 
with injection and oral administration — is more feasible and convenient, 
especially in view of the rising number of peptide and protein therapeutics that are 
rapidly being developed. In particular, the possibility of delivering drugs to the brain 
by the nasal route is eliciting increased interest, especially due to the possibility of 
accessing or targeting the local receptors and also of circumventing the blood – brain 
barrier. 
Drug delivery via the nasal route offers a number of advantages, the most important 
of which is the possibility of needle - free treatment. It also means that — in 
addition to the newly developed peptide - and protein - based drugs — this method is 
also suitable for a wide variety and perhaps most of the drugs that are currently in 
use. However, it is not only convenience that sets nasal drug delivery apart: This 
method also provides a rapid onset of action and high bioavailability. 
Because of its rich vasculature and highly permeable structure, the nasal route 
can be used as an alternative to parenteral routes of delivery. It circumvents hepatic 

fi rst - pass metabolism and gut - wall enzyme - mediated degradation. It is also easily 
accessible for self - administration without the help of a health professional and there 
are no associated needle - stick hazards. Other advantages of nasal drug delivery 
systems include a rapid onset of action, reduced risk of overdose, and improved 
patient compliance. However, there are also several disadvantages, including the 
impermeability of the nasal mucosa to lipophilic and high - molecular - weight drugs, 
mucotoxicity associated with long - term use of some formulations, the requirement 
for an expensive delivery device, and possible dose inaccuracy. 
In order to understand the delivery and absorption of drugs by the nasal route 
and appreciate the factors that may affect it, one must begin with a clear picture of 
the anatomy and physiology of the nose. 
5.6.2 NASAL ANATOMY AND PHYSIOLOGY 
5.6.2.1 Structure of Nasal and Olfactory Mucosa 
The human nasal passage is about 12 cm long and runs from the nostrils to the 
nasopharynx (Figure 1 ). The nasal cavity is divided into right and left halves by a 
midline septum, or cartilaginous wall, that extends posteriorly into the nasopharynx 
(E). Each half of the nasal cavity consists of three well - separated regions: (A) the 
vestibule, (B) the olfactory region, and (C) the respiratory region. The vestibule 
is the most anterior part of the nasal cavity, which opens to the face through the 
FIGURE 1 Sagittal section of nasal cavity showing nasal vestibule (A), atrium (B), respiratory 
area and inferior turbinate (C1), middle turbinate (C2) and superior turbinate (C3), 
olfactory region (D), and nasopharynx (E). ( Reproduced from ref. 5 with permission of 
Pharmaceutical Press. ) 
D 
B 
A 
C1 
C2 
C3 
E 
NASAL ANATOMY AND PHYSIOLOGY 593

594 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
nostrils. The respiratory region and the turbinates make up most of the nasal cavity; 
this region has lateral walls that divide it into three chambers, including the superior 
nasal turbinate (C3) at the top, the middle nasal turbinate (C2) below, and the 
inferior nasal turbinate (C1) at the bottom (Figure 1 ). The total volume of nasal 
cavity is 15 mL; its surface area is 150 cm 2 [4, 5] . Air generally travels into the nose 
in an approximately parabolic pattern from the vestibule to the nasopharynx. The 
hard palatine bone and soft palate form the fl oor of the nose and the roof of the 
mouth. The human nasal epithelial surface is covered mainly by stratifi ed squamous, 
olfactory, and respiratory epithelia. The stratifi ed epithelium lies in the anterior part 
of the nose and becomes pesudostratifi ed columnar epithelium at the posterior part, 
which constitutes the respiratory epithelium (Figure 2 ). The olfactory epithelium 
covers the olfactory region in the upper part of the nasal cavity. The nasal epithelial 
surface is covered by a continuous layer of mucus secreted by various mucosal and 
submucosal glands. The mucous layer comprises the gel layer (D) and sol layer (E) 
as shown in Figure 2 . The sol layer is a low - viscosity fl uid surrounding the cilia, and 
the viscous gel layer covers the tips of the cilia — fi ne, hairlike structures that move 
in an organized fashion to ease the fl ow of mucus across the epithelial surface. There 
are also microvilli, and every ciliated cell carries some 100 cilia. Each ciliated and 
nonciliated cell has about 300 microvilli. Figure 2 also shows both nonciliated cells 
and basal cells [4 – 6] . 
The olfactory mucosa is discussed further on in relation to the delivery of drugs 
via the nose to the brain. 
5.6.2.2 Nasal Vasculature 
The nasal surface is supplied with a dense network of blood vessels by the external 
and internal carotid arteries. Blood from the anterior part of the nose is drained 
through the facial vein, but the nose ’ s main blood supply drains through the sphenopalatine 
foramen into the pterygoid plexus or via the superior ophthalmic vein 
FIGURE 2 Cell types of nasal epithelium showing ciliated cell (A), nonciliated cell (B), 
goblet cell (C), gel mucous layer (D), sol layer (E), basal cell (F), and basement membrane 
(G). ( Reproduced from ref. 5 with permission of Pharmaceutical Press. ) 
F G 
A B E D C

[6] . The nasal blood vessels can be greatly dilated with blood to facilitate warming 
and humidifi cation of inspired air in response to prevailing conditions. Nasal blood 
fl ow is very sensitive to a variety of agents applied topically and systemically. 
5.6.2.3 Enzymes and p H 
Nasal secretions contain a mixture of secretory materials from the goblet cells, nasal 
glands, and lacrimal glands. The main constituents of nasal secretions are water, with 
2 – 3% mucin and 1 – 2% electrolytes. Nasal secretions also contain several enzymes, 
including lysozyme, cytochrome P450 – dependent monooxygenases, steroid hydroxylases, 
proteases such as neutral endopeptidase, leucine aminopeptidase, aminopeptidase 
peroxidase, carboxypeptidase N, and protease inhibitors [7] . However, 
because most studies of peptide and protein degradation are carried out in homogenates 
of nasal tissue, the peptides and proteins can be exposed to both intracellular 
and extracellular enzymes; therefore, the data regarding peptide stability must be 
interpreted with caution. The normal pH of nasal secretions in adults ranges from 
about 5.5 to 6.5; in young children, it ranges from 5.0 to 6.7. The nasal pH can vary 
depending on pathological conditions such as allergic rhinitis and environmental 
conditions such as cold and heat [8] . 
5.6.3 FACTORS INFLUENCING NASAL DRUG ABSORPTION 
The nasal absorption of drugs is infl uenced by a multitude of factors, including nasal 
physiology, nasal pathology, physicochemical properties of drugs, dosage forms, and 
delivery method. The presence of pathological conditions such as allergic rhinitis 
and the common cold further complicates nasal drug delivery. Moreover, the intimate 
contact between the nasal mucosa and the atmosphere leads to variability in 
absorption with changes in temperature and humidity. 
5.6.3.1 Physiological Factors 
Nasal Blood Flow The nasal vasculature differs from the tracheobronchial tree 
due to the presence of (a) venous sinusoids, (b) arteriovenous anastosomes, and 
(c) the nasal vasculature, which shows cyclical changes of congestion, hence giving 
rise to the nasal cycle. In the nasal vasculature, the arterioles lack an internal elastic 
membrane, making the endothelial basement membrane continuous with the basement 
membrane of the smooth muscle cells. Also present are the fenestrated type 
of capillaries lying just below the surface epithelium and surrounding the glands. 
Because of this, the capillaries facilitate rapid movement of fl uid through the vascular 
wall, allowing water to escape into the airway lumen. The conditioning of the 
inhaled air is greatly infl uenced by the nasal blood vessels. In essence, air is heated 
and humidifi ed by the fl ow of nasal blood in the opposite direction to the incoming 
airfl ow. The nasal blood fl ow also controls the size of the nasal passage ’ s lumen. 
Changes in ambient temperature and humidity, nasal administration of vasoactive 
drugs, nasal trauma, and compression of large veins in the neck may adversely affect 
blood fl ow in the nose [9, 10] . Other factors such as mood changes, hyperventilation, 
and even exercise can have an effect on the nasal blood fl ow and hence the nasal 
FACTORS INFLUENCING NASAL DRUG ABSORPTION 595

596 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
passages [10 – 12] . In conclusion, any change in blood fl ow can alter the absorption 
of a nasally administered drug. 
Nasal Enzymes The enzymes present in the nasal cavity may be involved in the 
extensive enzymatic degradation of drugs administered nasally. The presence of the 
enzymes in the nasal epithelium acts as a defensive mechanism or a barrier against 
the entry of xenobiotics [9] . Examples include nasal decongestants, essences, anesthetics, 
nicotine, and cocaine, which are metabolized by the nose ’ s P450 - dependent 
monooxygenase system [13, 14] . The nasal mucosa also includes oxidative phase I 
and conjugative phase II enzymes. The phase I enzymes include aldehyde dehydrogenase, 
carboxyl esterase, and carbonic anhydrases; phase II enzymes include glucuronyl, 
sulfate, and glutathione transferases. 17 . - Estradiol has shown signifi cantly 
more conjugation when administered via the nasal route as compared to the intravenous 
route. A variety of other drugs have been shown to be metabolized by nasal 
enzymes, including progesterone, testosterone, and insulin [15, 16] . 
5.6.3.2 Nasal Mucociliary Clearance 
Nasal mucociliary clearance is the transport of the mucous layer covering the nasal 
epithelium toward the nasopharynx by ciliary beating for its eventual discharge into 
the gastrointestinal tract. Nasal mucociliary clearance plays a very important role 
in the upper respiratory tract in preventing various noxious agents such as allergens, 
bacteria, viruses, and toxins from reaching the lungs. The ciliated cells of the nasal 
mucosa drive the movement of the mucus, and hence the physiological control of 
the ciliated cells and the rheological properties of the mucus determine the effi - 
ciency of the nasal mucociliary clearance system. In humans the normal mucociliary 
transit time is reported to be 12 – 15 min, and transit times of 30 min or more are 
likely to be an indication of impaired mucociliary clearance. Impairment of mucociliary 
clearance has been associated with longer contact times of various noxious 
agents as well as drugs with the nasal mucosa. On the other hand, increases in the 
mucociliary clearance rate decrease the contact between drug and the epithelium 
and consequently reduce drug absorption. Therefore nasal drug absorption can be 
augmented by the use of bioadhesive polymers or microspheres or by increasing 
the viscosity of the drug formulation [5, 17] . Hydroxypropyl methylcellulose, polyacrylic 
acid, and hyaluronan all enhance absorption by increasing nasal residence 
time [18–20] . The effect of mucociliary clearance may vary depending on the site of 
drug deposition. Ciliated epithelium is present in the middle and posterior parts of 
the turbinates, but there is little or no ciliary epithelium in the anterior regions 
of the nasal cavity [21, 22] . This is one of the reasons why a drug deposited in the 
posterior part of the nose is washed away more quickly than a drug deposited in 
the anterior site of the nasal cavity [23] . 
5.6.3.3 Pathological Condition of Nose 
As mentioned earlier, the presence of nasal pathological conditions — such as allergic 
rhinitis, polyposis, and common colds — infl uences nasal drug absorption to a 
great extent. The majority of pathological conditions of the nose show bleeding, 
excessive secretion of mucus, nasal blockage, and crusting. It has been reported that 

excessive nasal secretion may wash away a nasally administered drug before it can 
be absorbed [24] . Nasal drug absorption and distribution are also infl uenced by the 
presence of nasal polyps and blockage. Several studies however have suggested that 
the presence of nasal pathological conditions do not affect nasally administered 
peptide drugs. For example, buserelin and desmopressin absorption studies have 
shown similar nasal absorption profi les in normal subjects and in those suffering 
from colds or rhinitis [25, 26] . 
5.6.3.4 Dose Volume and Site of Deposition 
A dose of 25 – 200 . L per nostril is what can be maximally accommodated by the 
human nose. A dose higher than this will be drained off and hence shows lower 
absorption. Some studies have reported that a 100 - . L volume resulted in a larger 
deposition area. Hence, taking into account the volume of administration becomes 
very important for manufacturers of nasal drug delivery systems. The site of deposition 
of the nasal formulation may also affect the nasal absorption of drugs since the 
anterior part of the nose provides greater contact between the nasal epithelium and 
drug, but the mucociliary clearance mechanism of the posterior tends to remove 
drug more rapidly [27] . It has been found that the permeability of the posterior area 
is greater than that of the anterior portion, and hence, based on the formulation, 
drugs may be administered in either the anterior or posterior parts of the nose. The 
nasal adapter ’ s spray - cone angle defi nes the width of the nasal spray pattern and 
thus plays an important role in determining the site of deposition in the nasal cavity. 
Changes in the cone angle of the adapter from 60 ° to 35 ° or 30 ° can produce a larger 
and more posterior deposition and therefore higher drug deposition in the ciliated 
area [10] . 
5.6.3.5 Physicochemical Properties of Drugs 
The absorption of a drug across the nasal mucosa is a function of its physicochemical 
properties, such as molecular weight, lipophilicity, and water solubility, as seen with 
most of the mucosal routes of delivery. The majority of studies on the effects of drug 
lipophilicity on nasal absorption are rather confl icting. The effect of lipophilicity on 
the nasal absorption of barbituric acids has been investigated. It was found that drug 
absorption through the nasal mucosa increases with an increase in the partition 
coeffi cient. Interestingly, there was only a fourfold increase in absorption between 
phenobarbital and barbital despite the fact that the partition coeffi cient of phenobarbital 
was 40 - fold higher than that of barbital [28] . Similarly, increases in nasal 
absorption have been seen for hydrocortisone, testosterone, and progesterone with 
increases in the partition coeffi cient. However, a hyperbolic — rather than a linear — 
relationship was observed between the in vivo nasal bioavailability of a series of 
progesterone derivatives and their octanol – water partition coeffi cients [29] . In contrast 
to this, Kimura et al. [30] showed that, for a series of quaternary ammonium 
compounds structurally related to tetraethylammonium chloride, nasal absorption 
was inversely related to the partition coeffi cient. All these studies suggest that a 
drug ’ s lipophilicity may not be an appropriate indicator of the extent of its nasal 
absorption. Besides the drug lipophilicity another important factor most studied for 
its infl uence on nasal absorption is the aqueous solubility of a drug. This is because 
FACTORS INFLUENCING NASAL DRUG ABSORPTION 597

598 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
the nasal mucosa is constantly kept moist by the nasal secretions and is well perfused 
with blood vessels. In addition to limiting drug absorption, it also is a limiting factor 
for the formulation of the drug. Further it is also important to understand the relation 
between the saturation solubility of the drug and drug absorption. However, 
the infl uence of drug solubility on absorption of drugs via the nasal route has not 
been signifi cantly explored and needs much more attention. The relative effectiveness 
of nasal atropine and hyoscine was compared by Tonndorf et al. [31] by measuring 
each drug ’ s capacity to arrest salivary secretion; they found that 0.65 mg of 
hyoscine, 40 times more soluble in water than atropine, is equivalent to 2 mg of 
atropine. Some authors have suggested that the aqueous pores in the nasal mucosa 
play a major role in absorption of hydrophilic drugs [32] . When nasal formulations 
are administered as inhaled powders or suspensions, drug dissolution rate becomes 
an important factor. Formulations deposited in the nostrils require proper dissolution 
for better absorption. Nasal mucociliary clearance may remove the drug if it 
remains undissolved in the nostrils. The size and shape of a drug molecule can affect 
its nasal absorption. Molecules with an average molecular weight less than 1000 Da 
are better absorbed nasally than higher molecular weight drugs whereas linear 
molecules are less effectively absorbed than compact ones [33] . The nasal and oral 
absorption of polyethylene glycol 600, 1000, and 2000 have been studied by Donovan 
et al. [34] in relation to molecular weight; they found that the greater the molecular 
weight, the less effective the absorption. This pattern of absorption was seen in the 
case of both the nasal and oral routes. The effect of water - soluble compounds such 
as 4 - oxo - 4 H - 1 - benzopyran - 2 - carboxylic acid, p - aminohippuric acid, sodium cromoglycate, 
inulin, and dextran showing different molecular weights on the nasal absorption 
was studied by Fisher et al. [35] . A 43 - fold decrease in the nasal absorption of 
the these compounds was observed with a 368 - fold increase in the molecular weight 
[9] . Similarly, studies with 13 di - iodo - l - tyrosine - labeled dextran showed an inverse 
relationship between the percentage absorbed after nasal administration and the 
weight of the molecule; in fact, a 36 - fold increase in molecular weight produced an 
88 - fold reduction in nasal absorption [36] . The effect of molecular weight on the 
nasal absorption of fl uorescein isothiocyanate and diethylaminoethyl dextrans has 
been studied by Maitani et al. [37] . It was found that an inverse relationship between 
absorption and molecular weight existed for these compounds. However, since the 
nasal absorption of these compounds was low, enhancers were used; hence it is dif- 
fi cult to rule out the infl uence of the enhancers used on the extent of absorption 
obtained. The absorption of a drug after nasal administration is also infl uenced by 
the pH of a drug formulation as well as that of the nasal cavity — along with the p Ka 
of the drug substance. Biological membranes form a major barrier to the transport 
of drugs into the bloodstream. There are a number of transport mechanisms by 
which drugs are transported across the biological membranes. These include transcellular, 
paracellular, and carrier - mediated transport mechanisms. The most important 
factors that infl uence the above - mentioned mechanisms are the pH, p Ka , and 
partition coeffi cient of the drug. The pH of a nasal formulation should be in the 
range of 4.5 – 6.5 in order to minimize nasal irritation. However, the drug ’ s p Ka must 
also be taken into account so as to maximize the drug ’ s concentration in un - ionized 
form. The effect of pH on the nasal absorption of benzoic acid was studied by 
Hussain et al. [28] , who showed that the absorption of benzoic acid is pH 
dependent. 

5.6.3.6 Type of Delivery Device 
Both the type of drug delivery system and the specifi c type of delivery device can 
affect drug absorption via the nasal route. The choice of delivery system depends 
mainly on the physiochemical properties of the drug, its desired site of action, and, 
more importantly, patient compliance and marketing aspects. The formulations most 
commonly used in nasal delivery are solutions, suspensions, gels, dry powders, and, 
most recently, nanoparticulate formulations. 
Solutions are most commonly used for intranasal drug delivery. Such solutions 
may be used when the active ingredient is soluble in water or in some other vehicle 
approved by the Food and Drug Administration. At present, nasal solutions are 
available in the form of drops and sprays. Drops are the simplest and the most 
convenient nasal dosage form, and they are also easy to manufacture. However, 
their major drawback is that exact dosages cannot be administered with them. 
Another disadvantage is that they — like most solution - based medications — are vulnerable 
to microbial contamination; therefore, preservatives must often be added. 
These, in turn, have further disadvantages, as they may both cause irritation and 
hamper mucociliary clearance, thus decreasing compliance. Chemical stability is also 
often an issue with nasal drops. 
Since the introduction of metered - dose inhalers, nasal solutions have increasingly 
been formulated as nasal sprays. Initially, aerosol - based systems containing chloro- 
fl uorocarbons were employed; however, the Montreal Protocol put an end to this. 
Thereafter, mechanical pumps or actuators were employed to deliver nasal formulations 
as sprays. These devices, using actuators, can precisely deliver as little as 25 . L 
and as much as 200 . L of a formulation. However, various factors must be considered 
in formulating the spray; these include viscosity, particle size, and surface 
tension, all of which may affect the accuracy of the dose administered. 
Suspensions may also be used to deliver nasal formulations, though only rarely, 
since a number of complicating factors (e.g., particle size and morphology) must be 
considered. Suspensions offer the advantage of increasing residence time in the 
nasal cavity, thus possibly augmenting nasal bioavailability. 
Gels are thickened solutions that may sometimes be used to deliver drugs via the 
nose, since they offer a number of advantages, such as reducing postnasal drip into 
the back of the throat and hence reducing the loss of the drug from the nasal cavity, 
anterior leakage, and the associated irritation. The use of gels is also reported to 
improve absorption and to mask the irritation associated with some ingredients by 
the addition of soothing agents and emollients. A vitamin B 12 (cyanocobalamin) nasal 
gel, Nascobal (Nastech Pharmaceutical, Kirkland, WA), is available in a metered - 
dose formulation. Several other drugs, such as insulin, are being studied with a view 
to formulating them as nasal gels [9, 27, 38] . Although nasal powders are more stable 
than other formulations, they are rarely used because they tend to irritate the nasal 
tissue. However, a powder form may be useful when the active ingredient cannot be 
formulated as a solution or suspension. With the development of refi nements in 
technology, many researchers are exploring the use of nanoparticle - based formulations 
to deliver drugs nasally. The main advantage of these state - of - the - art formulations 
is that they ensure increased absorption as well as better compliance. 
Microsphere - and liposome - based formulations are being increasingly tested. Some 
of these studies are discussed in Sections 5.6.6 and 5.6.7. 
FACTORS INFLUENCING NASAL DRUG ABSORPTION 599

600 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
The type of nasal device employed in delivering a drug formulation plays a major 
role in the effi cacy of the treatment. In general, two types of delivery systems are 
used: mechanical pumps and pressurized aerosol containers. The properties of the 
drug to be used infl uence the selection of the system. The various types of delivery 
devices are described in the following sections. 
Unit-Dose Containers The unit - dose container offers a number of advantages. It 
is easy and convenient to carry and also does away with the need for preservatives, 
thus greatly increasing patient compliance. The unit - dose container is more accurate 
than the multidose container; metered - dose nasal sprays are still more accurate. The 
volume of drug held by such a container is usually determined by its fi lling volume, 
which greatly infl uences its accuracy. Another type of unit - dose container is based 
on an actuator and consists of a nasal adapter, or a small chamber that contains a 
piston [9] . Unit - dose containers are used mainly in emergencies (e.g., to manage 
pain), although they are not restricted to such use and may also be employed 
in instances where a single administration is required (e.g., vaccination). An 
example of a product that employs unit - dose containers is Imitirex (sumatriptan) 
(GlaxoSmithKline, Research Triangle Park, NC). 
Squeeze Bottles This is a smooth plastic bottle with a jet outlet. When the bottle 
is pressed, a certain volume of its contents is atomized as the air inside the bottle 
is pushed out. This type of device is vulnerable to contamination as ambient air 
rushes into it following the release of pressure. The squeeze bottle is used mainly 
to deliver decongestants and not vasoconstrictors, as in the latter case the dose 
administered would be diffi cult to control [9] . 
Metered-Dose Nasal Pump Sprays Metered - dose pumps are the most widely used 
devices for the delivery of formulations via the nose. A number of commercially 
available products use this technology. The accuracy of the delivered dose is fairly 
high and makes it possible to administer dose volumes ranging from 25 to 200 . L. 
A metered - dose pump is made up of a container as well as a pump, valve, and actuator. 
The characteristics of the spray delivered will differ depending on the properties 
of the drug, the precompression mechanism, and the valve and pump selected. The 
length of the actuator is an important factor determining the deposition of the drug 
in the nose; the collection of residual drops on its tip will affect correct dosing. 
Airless and Preservative -Free Sprays There are now pumps that prevent the entry 
of air into a dispensing device after use, thus increasing the stability of numerous 
compounds that are vulnerable to oxidation; this innovation has also minimized the 
use of preservatives. The working principle of these pumps is operation against a 
vacuum using a collapsible bag and a sliding piston. This is possible because the 
vacuum created when a dose is dispensed is accompanied by a reduction in the 
volume of the container, either by deforming the container itself or by dragging 
the sliding piston out of it. These maneuvers have no infl uence on the system ’ s effi - 
ciency and, in fact, provide an advantage in that the container can be held in any 
position without signifi cantly compromising the accuracy of the dose dispensed. This 
system is particularly suitable for use with children and bedridden hospitalized 
patients. 

Whenever systems without preservatives are used for single or double doses, they 
pose little risk of contamination. However, multidose systems are generally used 
over a longer period of time; therefore, unless their formulations also include preservatives, 
the chances of contamination are increased. Scientists from Erich Pfeiffer 
and Qualis Laboratorium (both in Germany) have reviewed the latest trends in 
preservative - free nasal sprays and report that it is possible to prevent microbial 
contamination via the orifi ce in two ways. The fi rst is by introducing a chemical 
additive, such as a bacteriostatic agent, into the nasal actuator so that it comes into 
contact with both the medication and the environment. However, in the case of an 
open system, the formulation within the actuator can still be contaminated. The 
second approach is the use of a mechanism whereby the system is sealed behind 
the orifi ce, thus preventing microorganisms gaining access [9, 39] . 
Some innovative technologies are being developed by a variety of pharmaceutical 
fi rms. The following section touches briefl y on the latest of these. 
Kurve Technology Kurve Technology has developed a unique system of controlled 
particle dispersion (CPD) by which it is possible to deliver drugs to the entire nasal 
cavity as well as the olfactory region and the paranasal sinuses. It uses the principle 
of vortical fl ow, by which inherent airfl ows of the nasal cavity are disrupted (Figure 
3 a and b ). Its advantages include optimization of the size and trajectory of droplets, 
which makes it possible to saturate the nasal cavity. CPD also increases nasal residence 
time and reduces the deposition of compounds in the lungs and stomach, thus 
making the treatment more effective and effi cient. ViaNase ID (Kurve Technology, 
Bothell, WA) is a CPD - powered electronic atomizer also developed by Kurve. Its 
advantages include generation of narrow droplet distribution between 3 and 50 . m 
and control of the atomization rate (i.e., the rate at which the droplets are generated 
and how rapidly they exit the device). CPD technology can be used to deliver both 
solutions and suspensions; currently, work is in progress to apply this principle to 
dry powders. Testing is also under way for the delivery of small and large molecules 
as well as peptides and proteins. Finally, CPD technology makes it possible to 
provide preservative - free packaging; unit - dose ampules; targeted deposition, as 
mentioned earlier; and monitoring of doses and compliance [40 – 42] . 
OptiNose OptiNose AS (Oslo, Norway) has introduced the novel idea of bidirectional 
intranasal drug delivery, which delivers a drug while the patient exhales and 
thus is said to prevent lung deposition. It utilizes the concept that exhalation against 
resistance leads to closure of the soft palate, thus separating the nasal cavity from 
the mouth as well as cutting off communication between the cranial surface of the 
soft palate and the posterior margin of the nasal septum. When this occurs, the air 
can enter one nostril through the sealing nozzle, turn 180 ° , and fi nally exit through 
the other nostril in the reverse direction. This concept is utilized in breath - actuated 
bidirectional delivery; that is, the air is blown out of the container and the sealing 
nozzle is used to direct its fl ow of air into the nose. When this approach was compared 
with conventional nasal drug delivery in 16 healthy subjects using 99m Tc - 
labeled nebulized particles, it was found that bidirectional nasal delivery did, in fact, 
prevent deposition in the lungs. The single - use device is already developed and 
undergoing clinical testing for the delivery of a variety of compounds; a multidose 
liquid reservoir and powder delivery device are also being developed. The technol- 
FACTORS INFLUENCING NASAL DRUG ABSORPTION 601

602 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
FIGURE 3 ( a ) Vertical droplet fl ow created by controlled particle dispersion used in 
ViaNase ID (Kurve Technology, Bothell, WA). ( b ) Deposition pattern produced by controlled 
particle dispersion. ( Reproduced from ref. 42 with permission from Drug Delivery 
Technology. ) 
Controlled particle dispersion 
Transistion from aperture to 
nasal cavity 
Vestibule 
Nasal cavity 
Delivery path of the droplet 
Velocity profile of air steam 
a: Nasal vestibule 
b: Palate 
c: Inferior turbinate 
d: Middle turbinate 
e: Superior turbinate 
f: Nasopharynx 
(a) 
(b) 
ogy is also being tested in connection with vaccination by the nasal route, and vaccines 
against diphtheria and infl uenza have already been shown to improve local 
and systemic immune responses. Another area being explored is the delivery of 
drugs to the brain via the nose. A phase I clinical trial of midazolam has been carried 
out and has shown an onset and level of sedation comparable to that of intravenous 
administration. The duration of sedation was found to be longer with the OptiNose 
technology, but the bioavailability of the drug was found to be only 68%, as 
compared with 100% with intravenous administration. Figures 4 a and b show the 
Optinose multidose liquid device and multiuse powder device being developed 
respectively [43, 44] . 
DirectHaler The DirectHaler (Direct - Haler A/S, Copenhagen, Denmark) nasal 
delivery device takes advantage of the nasal anatomy and an innovation in device 
technology in order to improve nasal drug delivery and patient compliance. Similar 

to OptiNose, this novel drug delivery device avoids lung deposition. It takes advantage 
of the fact that, when air is blown out of the mouth against a particular resistance, 
the oral and nasal cavity airway passage closes on its own. Hence when a 
patient blows air into the DirectHaler Nasal device, the nasal dry powder dose is 
delivered into the nostril (Figure 5 ). The DirectHaler is reported to solve most of 
the problems that are associated with existing drug delivery devices, including dripping 
of the liquid dose out of the nostril following its delivery, swallowing of the 
dose after the administration and hence low absorption, and other problems of 
contamination associated with the liquid and multidose formulations. The device is 
quite easy and cost effective to manufacture, fi ll, and assemble using the latest high - 
speed technology. The tube of the device is made by using extrusion and roll forming 
while the device cap is manufactured by injection molding. A modifi ed high - speed 
capsule - fi lling machine is used to carry out the powder dose fi lling. DirectHaler has 
also been developed for the combination of oral and nasal drug delivery. For 
pulmonary delivery DirectHaler Pulmonary has been developed. Further, for the 
FIGURE 4 ( a ) Optinose multidose liquid device and ( b ) Optinose multiuse powder device. 
( Reproduced with permission of Per Gisle Djupesland by personal communication. ) 
(a) (b) 
FIGURE 5 DirectHaler Nasal delivery device. ( Reproduced with permission of Troels 
Keldmann by personal communication. ) 
Pharma blister pack 
avaiable, extra 
protection barrier 
Flow valve 
for control 
PowderWhirl chamber 
for turbulent dispersion 
of the dose 
Inhaler cap 
for moisture 
protection 
Mouth piece 
for generation 
of air flow 
Transparency of 
device for visibility 
of dose 
FACTORS INFLUENCING NASAL DRUG ABSORPTION 603

604 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
complete targeting of the whole respiratory system, Novel DirectHaler Compliance 
system has been designed. The device has successfully undergone clinical trials for 
its use in nasal drug delivery and has shown patient compliance [45] . 
5.6.4 ANIMAL MODELS FOR NASAL ABSORPTION STUDIES 
Nasal absorption studies can be carried out in mainly two types of animal models: 
the whole - animal model and the isolated organ perfusion model. 
In vivo nasal absorption has been studied in a variety of mammals, including the 
rat, rabbit, dog, sheep, and monkey. Rats are the most widely used animals for testing 
intranasal drug delivery. They are easy to handle and simple and inexpensive to 
maintain. Use of the rat model for the nasal delivery of insulin was fi rst described 
by Hirai and co - workers more than 25 years ago [46, 47] . The rat has been used in 
two basic models, in vivo and in situ — the latter also referred to as the ex vivo nasal 
perfusion model. Both models require anesthesia, though an in vivo model not 
requiring surgery has also been reported and is discussed later. In early studies of 
nasal absorption in rats, the animals were anesthetized and the passage of the nasopalatine 
tract was then sealed with acrylic glue to prevent the drainage of drug 
solution from the nasal cavity. The trachea was cannulated with a polyethylene tube, 
with another tube being inserted through the esophagus toward the posterior part 
of the nasal cavity. The drug solution was then delivered to the nose through either 
the nasal cavity or the esophageal tubing [8, 47] . Blood samples were then collected 
from the femoral vein. 
However, a less traumatic and much more feasible rat model has recently been 
proposed by Pillion et al. and other groups [48, 49] . In this model, the rats are anesthetized 
and kept on their backs; then the drug solution is administered directly into 
the nose by inserting a pipette about 3 – 5 mm into each nostril. In some cases the 
drug is administered into only one nostril so as to prevent blockage. The rats then 
remain on their backs long enough for the formulation to come into contact with 
the nasal mucosa. Blood samples are then collected from the tail vein. 
In support of the earlier models, it was argued that, by sealing the nasopalatine 
tract, the drug would necessarily be fully absorbed and transported into the circulation 
via permeation through the nasal mucosa. However, such nasal absorption with 
blockage of drainage from the nose to the mouth is not the normal physiological 
condition. Although the rat model has been used extensively by investigators 
throughout the world, application of the results of such studies to humans is very 
limited because of the small body size of these animals and signifi cant interspecies 
differences. In fact, signifi cant variability between the rat and the human was 
observed in studies of the bioavailability of insulin administered intranasally [50] . 
Furthermore, the use of anesthesia has raised concerns because of its potential to 
confound the test results. 
In the rabbit model, drug solution is delivered by spray instillation into a nostril, 
keeping the rabbit ’ s head in an upright position and allowing the rabbit to breathe 
normally. Blood samples are then collected from the ear vein. New Zealand White 
and Japanese White rabbits are most commonly used in such research. One of the 
advantages of the rabbit model is that the blood volume of these animals is large 
enough for multiple sampling and pharmacokinetic analysis [51] . 

Dogs, sheep, and monkeys can be kept conscious during nasal delivery to mimic 
the human [51] . Sheep, because of their large nostrils and docile nature, serve as 
excellent models for studies of this kind. 
5.6.5 ENHANCEMENT OF INTRANASAL DRUG ABSORPTION 
Lipophilic drugs or compounds have consistently been shown to be completely 
absorbed across the nasal mucosa; frequently, nasal absorption of these compounds 
is identical to that obtained with intravenous administration. In some reports, bioavailability 
after nasal administration reached almost 100% and Tmax was similar to 
that obtained with intravenous administration. For example, lipophilic drugs such 
as propranolol, naloxone, buprenorphine, testosterone, and 17 . - ethynylestradiol 
have been reported to be completely or almost completely absorbed after nasal 
administration in animal models. However, the same is not true of polar molecules 
and certain low - molecular - weight drugs and also high - molecular - weight peptides 
and proteins. In these instances absorption enhancers have been employed, and this 
is the method most widely used to improve nasal drug absorption [52 – 54] . Such 
absorption enhancers belong to a variety of different chemical groups and may have 
one or multiple ways of enhancing the absorption. Absorption enhancers work by 
(1) altering the mucous layer by decreasing its viscosity or disrupting it; (2) altering 
the tight junctions by sequestering extracellular calcium ions, which are reported to 
be essential in maintaining the integrity of these junctions; (3) inhibiting mucosal 
enzymatic degradation; (4) reverse - phase micelle formation — in certain cases, 
reverse - phase micelles may be formed within the cell membranes, thus creating an 
aqueous pore through which the drug can pass; and (5) altering membrane fl uidity, 
which can be achieved when there is disorder in the membrane phospholipid component 
or leaching of proteins from the membrane or by a combination of these 
mechanisms [52 – 56] . Table 1 lists some of the selected absorption enhancers based 
on the mechanisms mentioned above. 
5.6.6 NASAL DELIVERY OF PEPTIDE AND 
HIGH - MOLECULAR - WEIGHT DRUGS 
Protein and peptide delivery by means other than injection is currently receiving 
enormous attention due to the increasing number of biotechnology - based products 
being developed. There have been numerous attempts to design systems for oral 
peptide, protein, and gene delivery, but these have unfortunately met with limited 
success, thus providing an impetus for exploring alternative noninvasive delivery 
methods. As mentioned earlier, more and more research has been directed to nasal 
drug delivery because of the numerous advantages it offers. The following section 
deals with the delivery of peptide and protein drugs. 
5.6.6.1 Insulin 
Insulin is produced by the . cells of the islets of Langerhans in the pancreas. 
It is made up of two peptide chains, which have 21 and 30 amino acid residues, 
NASAL DELIVERY OF PEPTIDE AND HIGH-MOLECULAR-WEIGHT DRUGS 605

606 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
TABLE 1 Enhancement of Nasal Absorption 
Type of 
Compound 
(Absorption 
Enhancers ) Examples 
Mechanism of 
Absorption 
Enhancement 
Bile salts and 
derivatives 
Sodium deoxycholate, sodium glycocholate, 
sodium taurocholate, sodium 
taurodihydrofusidate, sodium 
glycodihydrofusidate 
Alteration of tight 
junctions, membrane 
disruption, inhibition 
of mucosal enzymatic 
degradation, mucolytic 
activity 
Chelating 
agents 
Citric acid, EDTA, enamines, N - acyl 
derivatives of collagen, salicylates 
Alteration of tight 
junctions 
Enzyme 
Inhibitors 
Amastatin, bestatin, camostat mesylate, 
boroleucine 
Enzyme inhibition 
Fatty acids and 
derivatives 
Acylcarnitines, acylcholine, caprylic acid, 
capric acid, oleic acid, phospholipids, 
mono - and diglycerides, sodium laurate 
Membrane disruption 
Surfactants Sodium lauryl sulfate, saponin, 
polyoxyethylene - 9 - lauryl ether, 
polyoxyethylene - 20 - lauryl ether, 
alkylmaltosides such as tetradecylmaltoside, 
dodecylmaltoside, and decylmaltoside 
Membrane disruption 
respectively. These two chains are held together by disulfi de linkages between cysteine 
residues. Insulin is an anabolic and anticatabolic peptide hormone with a 
molecular weight of about 6000. It was fi rst used in the successful treatment of diabetes 
mellitus in 1922. Today insulin is widely used in the treatment of both insulin - 
dependent and non - insulin - dependent diabetes mellitus [57 – 59] . A nonparenteral 
formulation of insulin has been approved only recently; before this development, 
insulin treatment required the use of painful injections. Exubera (Pfi zer Labs, New 
York, NY) is the fi rst insulin and fi rst biotechnology - based medicine for the treatment 
of a systemic disorder that can be administered without an injection. It was 
developed by Nektar (San Carlos, CA) and is now a registered trademark of Pfi zer 
(New York, NY). It still poses a few concerns, especially its effect on the lungs of 
patients with asthma or chronic obstructive pulmonary disease. Because of the 
problems associated with the parenteral injection of insulin, many diabetic patients 
fl atly refuse to accept insulin therapy. As to subcutaneous injections, these fail to 
attain a physiological pattern of insulin owing to their adverse pharmacokinetics, 
and normoglycemia is often not achieved [47] . Of all the routes so far studied apart 
from delivery via the lungs, the nasal route would appear to be the most advantageous 
for the delivery of insulin. 
The nasal delivery of insulin was demonstrated as early as 1922 by Woodyatt [59] . 
Since then, numerous studies have focused on this methodology. Some of the early 
studies included absorption of insulin from the nasal mucosa in human diabetics, 
the use of an insulin sprayer that contained saponin, and insulin in ethylene glycol 
or trimethylene glycol applied to the nose in the form of drops or sprays; the last 

of these methods demonstrated a signifi cant fall in blood glucose levels in normal 
rabbits, dogs, and diabetic humans [60 – 62] . 
The enhancement of nasal absorption of insulin by hydrophobic bile salts has 
also been investigated. It was found that minor differences in the number, position, 
and orientation of the nuclear hydroxyl groups as well as alterations to side - chain 
conjugation can improve the adjuvant potency of bile salts. Moreover, the absorption 
of insulin positively correlated with an increase in the hydrophilicity of the 
steroid nucleus of the bile salts. In the presence of bile salts, nasal absorption of 
insulin reached peak levels within about 10 min, and some 10 – 20% of the dose was 
found to have been absorbed into the circulation. Marked increases in serum insulin 
levels were seen with sodium deoxycholate, the most lipophilic of the bile salts, 
whereas the least elevation — as well as least lowering of blood glucose levels — was 
seen with the most hydrophobic bile salt, sodium ursodeoxycholate [63] . 
Morimoto et al. [64] studied the nasal absorption of insulin using polyacrylic acid 
gel. When insulin was formulated with 0.1% w/v polyacrylic acid gel base (pH 6.5), 
the maximum hypoglycemic effect was seen 30 min following intranasal administration; 
in 1% w/v gel base, however, it took 1 h to reach the maximum effect. There 
was no effect of the pH (4.5, 6.5, and 7.5) of 0.1% w/v polyacrylic acid gel on the 
extent of nasal absorption. 
Pillion and his group studied alkylmaltosides differing in chain length for their 
abilities to lower blood glucose levels when formulated with insulin [65] . Tetradecylmaltoside 
(TDM) was the most effective agent in producing the hypoglycemic 
effect, followed by dodecylmaltoside (DDM) and decylmaltoside (DM), all at concentrations 
of 0.060%. The onset of hypoglycemic action using these nasal drops 
was seen within 30 min and the maximum effect was obtained within 60 – 120 min. It 
was also demonstrated that insulin plus TDM at concentrations as low as 0.03% 
induced a hypoglycemic effect; however, insulin plus octylmaltoside (OM) failed to 
produce any hypoglycemic effect even at OM concentrations as high as 0.50%. 
Dodecylsucrose, which differs from DDM by only one carbohydrate residue, had a 
similar effect on blood glucose; however, decylsucrose was found to be less potent, 
and nonglucosides were able to enhance the nasal absorption of insulin only at 
concentrations . 0.50% [65] . 
Insulin formulated with 0.06 or 0.125% hexadecylmaltoside produced a pronounced 
and rapid dose - dependent decrease in blood glucose levels after nasal 
administration. The effects of seven different alkylmaltosides were studied, and all 
the reagents (Figure 6 ) showed a similar maximal enhancement of insulin uptake 
when a concentration of 0.125% was employed. The fi gure demonstrates that TDM 
showed the greatest effect when concentrations of 0.03 and 0.06% were used. 
Similar experiments were carried out using sucrose esters in nasal insulin formulations 
(Figure 7 ). It was observed that tetradecanoylsucrose and tridecanoylsucrose 
were more effective in stimulating insulin absorption as compared with decanoylsucrose 
and dodecanoylsucrose. But — compared with TDM at concentrations of 
0.03% — the sucrose esters were less effective in promoting nasal absorption [66] . 
Sucrose cocoate (SL - 40) is produced by the chemical esterifi cation of coconut oil 
with sucrose; it has frequently been used in cosmetic and dental preparations as an 
excipient. When this excipient was formulated with insulin at 0.125, 0.25, and 0.5% 
concentrations, the associated plasma levels of insulin increased rapidly; whereas 
there was no enhancement of insulin plasma levels when insulin in saline was admin- 
NASAL DELIVERY OF PEPTIDE AND HIGH-MOLECULAR-WEIGHT DRUGS 607

608 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
FIGURE 6 Changes in area under the curve (AUC 0 – 120 ) for blood glucose values in rats 
that received 2 U insulin in presence of alkylmaltosides. Data represent mean change in 
AUC 0 – 120 in arbitrary units (AU) ± standard error of the mean (SEM) compared with rats 
that received insulin formulated without alkylmaltoside ( n = 3, 4). ( Reproduced from ref. 66 
with permission of John Wiley & Sons. ) 
Octyl 
Decyl 
Dodecyl 
Tridecyl 
Tetradecyl 
Pentadecyl 
Hexadecyl 
Concentration (%) 
Alkylmaltosides 
AUC Change (AU) 
10
9
8
7
6
5
4
3
2
1
0
0.00 0.02 0.04 0.06 0.08 0.10 0.12 0.14 
istered. The levels of insulin in plasma increased from a baseline value of 10 . U/mL 
to a level of 200 . U/mL after the nasal administration of sucrose cocoate - containing 
formulations; the T max was found to be 10 min (Figure 8 ) [67] . 
Chitosan is a linear cationic polysaccharide made up of copolymers of glucosamine 
and N - acetylglucosamine. It is commercially obtained by alkaline deacetylation 
of chitin [53, 68] and has been used for the nasal delivery of a number of 
drugs. The usefulness of chitosan in the enhancement of nasal absorption was 
reported fi rst by Illum [69] . Later, Illum and his group also published experimental 
results indicating that solution formulations with 0.5% chitosan promoted the 
absorption of nasally administered insulin in rat and sheep [70] . 
The use of chitosan nanoparticles in the enhancement of the nasal absorption 
of insulin has also been investigated in rabbits. Chitosan nanoparticles were prepared 
by ionotropic gelation of chitosan and pentasodium tripolyphosphate (TPP). 
Two types of chitosan were used in the hydrochloride salt form (Seacure ® 210 Cl 
and Protasan ® 110 Cl). Insulin loaded in chitosan 210 Cl produced a signifi cant 
increase in systemic absorption and also the greatest decline in the level of blood 
glucose, as much as 60% of basal levels; this result was found to be signifi cantly 
different from that obtained with the insulin control solution as well as the insulin – 
chitosan solution [71] . A novel chitosan nanoparticle formulation was prepared by 
again employing ionotropic gelation of TPP and chitosan glutamate (A1), and 
postloaded insulin – chitosan nanoparticles (A2) were also prepared. Both these 

FIGURE 7 Changes in area under the curve (AUC 0 – 120 ) for blood glucose values in rats 
that received 2 U insulin in presence of alkanoylsucroses. Data represent mean change in 
AUC 0 – 120 in arbitrary units (AU) ± SEM compared to rats that received insulin formulated 
without alkanoylsucroses ( n = 3, 4). ( Reproduced from ref. 66 with permission of John Wiley 
& Sons. ) 
Decyl 
Dodecyl 
Tridecyl 
Tetradecyl 
Concentration (%) 
AUC Change (AU) 
10 
9
8
7
6
5
4
3
2
1
0
0.00 0.02 0.04 0.06 0.08 0.10 0.12 0.14 
Alkanoylsucroses 
novel formulations were tested in rat and sheep models and also compared with 
the insulin – chitosan solution formulation (A3) and subcutaneous injection of 
insulin. In the rat model, it was found that A3 performed better than either A1 or 
A2. A1 showed a minimum blood glucose concentration ( C min ) and time to reach 
C min ( T min ) of 40% and 90 min, respectively. The F dyn — calculated as individual area 
over the curve (AOC IN or SC . dose SC /mean AOC SC . dose IN or SC ) . 100 — was found to 
be around 48%, whereas the insulin – chitosan nanoparticles showed F dyn values of 
38 and 37 for the A1 and A2 formulations, respectively. 
Since the concentrations of insulin to be administered in the sheep model would 
have been large, the insulin - loaded chitosan nanoparticles were not investigated in 
that model. However, the pharmacodynamics and pharmacokinetics of various 
insulin – chitosan preparations were compared with postloaded insulin – chitosan 
nanoparticles. It was found that chitosan solution and chitosan powder formulations 
were far better, with the chitosan powder formulation showing a bioavailability of 
17% as against 1.3 and 3.6% for the chitosan nanoparticles and chitosan solution 
[72] . The effects of the concentration and osmolarity of chitosan and the presence 
of absorption enhancers in the chitosan solution on the permeation of insulin 
across the rabbit nasal mucosa in vitro and in vivo were investigated, and the same 
NASAL DELIVERY OF PEPTIDE AND HIGH-MOLECULAR-WEIGHT DRUGS 609

610 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
investigation was also carried out in rats. The results suggested that, by increasing 
the concentrations of chitosan from 0 to 1.5%, insulin permeation in vitro could be 
increased. As compared with insulin without chitosan, there was a 25 - fold increase 
in the permeability coeffi cient of insulin with 1.5% chitosan. A similar increase in 
permeability was seen in the case of a hyperosmotic solution, and there was also an 
FIGURE 8 Changes in ( a ) plasma insulin levels and ( b ) blood glucose levels after nasal 
administration of 0.5 U insulin formulated in saline (  ) or 0.125% ( • ), 0.25% (  ), and 0.5% 
(%) sucrose cocoate. Blood glucose concentrations at time 0 (250 – 350 mg/dL) were normalized 
to a value of 100% in each animal. Data represent mean ± SEM, n = 3. Inserts represent 
changes in plasma insulin AUC 0 – 60 ( a ) and changes in blood glucose AUC 0 – 120 ( b ). 
( Reproduced from ref. 67 with permission of Elsevier. ) 
250 
200 
150 
100 
160 
140 
120 
100 
80 
60 
40 
0 20 40 60 80 100 120 
50
0
0 10 20 30 40 50 60 
10
8
6
4
2
0
0 0.1 0.2 0.3 0.4 0.5 0.6 
12 
10
8
6
4
2
0
0.0 0.10.2 0.3 0.4 0.50.6 
Plasma insulin (.U/mL) 
Time (minutes) 
AUC change (AU) AUC change (AU) 
Concentration (%) 
Concentration (%) 
Initial blood glucose (%) 
Time (minutes) 
(b) 
(a)

increase in permeability when insulin was formulated in deionized water as compared 
with phosphate buffer 7.4 (Figure 9 ). An increase in permeability was also 
seen when 5% hydroxypropyl . - cyclodextrin (HP . - CD) plus 1% chitosan was 
included with insulin as compared with chitosan alone. However, there was no 
statistical difference in permeability when insulin was formulated with 0.1% 
ethylenediaminetetraacetic acid (EDTA) and 1% chitosan. In the in vivo studies 
FIGURE 9 Effect of ( a ) concentrations, ( b ) osmolarity, and ( c ) medium of chitosan solution 
on mean serum glucose concentrations after nasal administration of 10 IU/kg insulin to rats. 
Bars represent the standard deviation (SD) of fi ve experiment. ( Reproduced from ref. 73 
with permission of Elsevier. ) 
0% chitosan 
0.5% chitosan 
1.0% chitosan 
1.5% chitosan 
Control: plain 
saline 
SC 
Hypo-osmolarity 
Iso-osmolarity 
Hyperosmolarity 
Time (h) 
Time (h) 
Time (h) 
Water 
pH7.4PBS 
Serum glucose 
(% of initial value) 
Serum glucose 
(% of initial value) 
Serum glucose 
(% of initial value) 
140 
140 
120 
100 
80 
60 
40 
20
0 
120 
100 
80 
60 
40 
20
0
0 1 2 3 4 5 
0 1 2 3 4 5 
120 
100 
80 
60 
40 
20
0
0 1 2 3 4 5 
(a) 
(b) 
(c) 
NASAL DELIVERY OF PEPTIDE AND HIGH-MOLECULAR-WEIGHT DRUGS 611

612 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
in rats, insulin without chitosan did not show any reduction in blood glucose levels, 
but as the concentrations of chitosan were increased, a hypoglycemic effect was 
seen. With a formulation of 0.5% chitosan, the nadir in glucose levels was observed 
within 1 h after administration; with 1 and 1.5% chitosan – insulin formulations, the 
lowest levels were reached in about 2 h. In the case of isoosmotic formulations, the 
lowering of blood glucose levels was weaker as compared with the hypo - or hyperosmolar 
solutions; with deionized water and EDTA, effects similar to those of the 
permeability studies were seen in terms of lower serum glucose levels. When insulin 
was formulated with 1% chitosan, there was also a decrease in serum glucose levels, 
and a similar effect was seen with both 1% chitosan and 0.1% EDTA. A similar 
effect was also seen in the case of 5% Tween 80. Insulin formulated with 5% HP . - 
CD and 1% chitosan was more effective in reducing serum glucose levels than when 
5% HP . - CD or 1% chitosan was used alone. These studies suggested that the concentrations, 
osmolarity, medium, and inclusion of absorption enhancers in chitosan 
solution infl uence absorption following the nasal delivery of insulin [73] . 
Varshosaz and co - workers recently explored the use of chitosan microspheres 
and chitosan gels in the nasal delivery of insulin [74] . They prepared microspheres 
of chitosan, and insulin was loaded into the microspheres. The formulations were 
administered through the nasal cavity by the method described earlier by Hussain 
et al. [47] . Varshosaz and colleagues found marked differences in the AUC of blood 
glucose reduction and the AUC of insulin concentration between the untreated 
controls and those animals that were treated either by intravenous administration 
and insulin - loaded chitosan (using ascorbyl palmitate as the cross - linking agent) or 
with microspheres. Serum glucose reduction in diabetic rats with nasal insulin – 
chitosan microspheres was around 67% in the intravenous group, and absolute 
bioavailability ( F abs ) was around 44% [74] . Furthermore, the same researchers prepared 
insulin – chitosan gels containing different enhancers and investigated their 
nasal absorption. As seen in Figures 10 and 11 , when EDTA was employed as an 
absorption enhancer in the chitosan gels and administered nasally, a signifi cant 
increase in insulin absorption and a decrease in serum glucose levels by as much as 
FIGURE 10 Serum glucose level in four groups of diabetic rats ( n = 6): A , untreated control 
group; B , intravenous administration of 4 IU/kg insulin; C , nasal administration of blank gel 
base; D , nasal administration of 100 . L/kg chitosan gel containing 4000 IU/dL insulin. 
( Reproduced from ref. 75 with permission of Taylor & Francis. ) 
450 
400 
350 
300 
250 
200 
150 
100 
50 
200 150 100 50 0 
0 
250 300 
Time (min) Serum glucose (mg/dL) 
A
B
C
D 
*** 
** * 
*** *** 
*** *** 
* 
***

46% compared to the intravenous route of administration were obtained. The 
authors suggested that this formulation would be benefi cial in the controlled delivery 
of insulin by the nasal route [75] . 
5.6.6.2 Calcitonin 
Calcitonin is a 32 – amino - acid peptide with a molecular weight of 3418 that is 
cleaved from a larger prohormone. It has a single disulfi de bond, which causes the 
amino terminus to assume a ring shape. Calcitonin is a hormone that participates 
in calcium and phosphorus metabolism. The major source of calcitonin in mammals 
is the parafollicular or C cells in the thyroid gland; it is also synthesized in other 
tissues, including the lungs and intestinal tract. When serum calcium levels are elevated, 
calcitonin is released from the thyroid gland. Salmon calcitonin (sCT) is more 
potent and longer lasting than the mammalian form and hence is used clinically. 
Calcitonin ’ s main action is to reduce the plasma concentration of calcium. At pharmacological 
doses, calcitonin brings about reduction in bone resorption. It is indicated 
for the treatment of postmenopausal osteoporosis in women with low bone 
mass relative to healthy premenopausal women. The marketed version of intranasal 
salmon calcitonin is Miacalcin (calcitonin - salmon) Nasal Spray (Novartis Pharma 
AS, Huningue, France). Up to now, this calcitonin treatment has been approved only 
for treatment in women. It has been reserved as a second - line treatment, since it 
reduces fracture risk less than do other available treatments for osteoporosis. 
Miacalcin Nasal Spray, fi rst manufactured by Sandoz Pharmaceuticals, was approved 
by the FDA in 1995 and is now distributed by Novartis Pharmaceuticals in the 
United States. 
Pontiroli et al. [76] looked at the intranasal absorption of calcitonin in normal 
subjects. Their study included six healthy volunteers who had no family history of 
endocrine or metabolic diseases. Human calcitonin (Cibacalcin; Ciba - Geigy) was 
administered intravenously or mixed with sodium glycocholate, a surfactant, in distilled 
water and instilled as nose drops. Plasma concentrations of calcitonin were 
found to be consistently higher when compared with intranasal administration of 
FIGURE 11 Serum insulin levels of four groups of diabetic rats ( n = 6): A , untreated control 
group; B , intravenous administration of 4 IU/kg insulin; C , nasal administration of blank gel 
base; D , nasal administration of 100 . L/kg chitosan gel containing 4000 . g/dL insulin. 
( Reproduced from ref. 75 with permission of Taylor & Francis. ) 
31,600 
31,200 
30,800 
4,000 
3,600 
3,200 
2,800 
2,400 
2,000 
1,600 
1,200 
800 
400
0 
Serum insulin (pmol/L) 
0 50 100 150 200 250 
AB
C
D 
Time (min) 
* 
*** 
*** 
*** *** 
** * 
* * 
* * 
** 
NASAL DELIVERY OF PEPTIDE AND HIGH-MOLECULAR-WEIGHT DRUGS 613

614 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
the same dose of calcitonin. However, it was found that intranasal administration 
effected a reduction in the plasma concentrations of calcium to a similar extent as 
that seen when a similar dose of intravenous calcitonin was given. The data also 
showed that although a higher dose of calcitonin by the intranasal route brought 
about a higher plasma calcitonin concentration, there was no difference in the 
decrease in plasma concentrations when calcitonin was given at doses of 500 and 
1000 . g. 
Polyacrylic acid aqueous gel enhances the absorption of calcitonin after nasal as 
well as rectal administration. When [Asu1,7] - eel calcitonin (10 U/kg) was administered 
nasally in polyacrylic acid gel at a concentration of 0.1% w/v, a prominent 
hypocalcemic effect was seen in the fi rst 30 min. Nasal administration of [Asu1,7] - eel 
calcitonin in saline had no hypocalcemic effect at the same dose when given by the 
nasal route. In addition to this, the effect of [Asu1,7] - eel calcitonin in the dose range 
of 1 – 10 U/kg has also been studied. The resulting data showed that a rapid reduction 
in plasma calcium concentrations can be achieved at doses of 5 and 10 U/kg; however, 
at doses of 1 U/kg only a small reduction in the plasma calcium concentration was 
observed, suggesting that polyacrylic acid gel can be used for the intranasal administration 
of peptides such as calcitonin. The possible side effects, however, were not 
known at the time the study was performed [76 – 78] . 
Commercially available sCT has been used mainly in the treatment of bone - 
related diseases such as Paget ’ s disease, hypercalcemia, and osteoporosis. It can 
be used to alleviate pain due to its analgesic properties; hence there may be a 
need for its frequent administration. As reported in the literature, owing to chemical 
and enzymatic degradation, the polypeptide has a short half - life of only about 
14 min. Hence its use calls for measures to improve its in vivo stability and overcoming 
the problem of rapid clearance. Polyethylene glycol (PEG) has been 
extensively used for this purpose in association with a variety of agents. Lee at 
al. [79] have prepared and characterized PEG - modifi ed sCT and studied how 
blood clearance is affected with and without PEGylation. Succinimidyl carbonate 
mono - methoxy - polyethylene glycol (SC - mPEG) was prepared using mPEG, which 
has a molecular weight of 12,000, and bis( N - succinimidyl) carbonate as per the 
procedure of Miron and Wilcheck [80] . There are three main reactive sites for 
the activated PEG in sCT: the N - terminal amino group (Cys 1 ) and the . - amino 
groups of two lysine residues (Lys 11 and Lys 18 ), as shown in Figure 12 . The site 
of conjugation of the PEG on sCT determines the stability of the sCT in the face 
of enzymatic attacks; hence the three possible mono - PEG - sCTs — Cys 1 - PEG - sCT, 
Lys 11 - PEG - sCT, and Lys 18 - PEG - sCT — can withstand the effects of proteolytic 
enzymes. It was found that, on PEGylation, the plasma half - life improved to 11.2 
and 54.0 min for the mono - PEG - sCT and the di - PEG - sCT as compared with 
the non - PEGylated sCT, which showed a plasma half - life of about 4.7 min 
(Figure 13 ). 
FIGURE 12 Primary structure of salmon calcitonin. Possible PEGylated sites are Cys 1 , 
Lys 11 , and Lys 18 . ( Reproduced from ref. 79 with permission of Taylor & Francis. ) 
Cys1-Ser-Asn-Leu-Ser-Thr-Cys-Val-Leu-Gly-Lys11-Leu-Ser-Gln-Glu-Leu-His-Lys18-Leu-Gln-Thr-Tyr- 
Pro-Arg-Thr-Asn-Thr-Gly-Ser-Gly-Thr-Pro-NH2

The same group — Shin et al. [81] — did nasal absorption studies of a low - 
molecular - weight PEG (2000) instead of the 5000 and 12,000 they tried previously. 
They also used commercially available succinimidyl - propionated monomethoxy - 
poly(ethylene glycol) - 2000 (SP - mPEG) for the chemical modifi cation instead of 
synthesizing it, as in their previous studies. The PEGylation of sCT was done by 
mixing SC - mPEG and sCT, and this mixture was shaken at 25 o C. The reaction 
mixture was stopped by using an excess of 1.0 M glycine solution. This conjugated 
mixture was then subjected to size exclusion chromatography. Radioiodination of 
the sCT and the PEG - sCT were carried out and the radiolabeled 125 I - sCT and 
125 I - mono - PEG2000 - sCT were then used for the nasal absorption studies. 
Tissue distribution studies were also done in rats after nasal administration. As 
seen in Figure 14 and Table 2 , it was found that the elimination half - life of the 
unmodifi ed sCT was 199 min, whereas the SP - mPEG2000 - modifi ed sCT showed an 
increased terminal elimination with a half - life of 923 min. It was also found that the 
SP - mPEG2000 - modifi ed sCT took a signifi cantly longer time to reach its maximum 
concentration, 520 min, as compared with the 77 min for the unmodifi ed sCT, and 
the AUC was found to be 20,638 . g/min/mL, which is much higher than the 3650 . g/ 
min/mL for the unmodifi ed sCT. The authors reported that the increase in the terminal 
half - life observed could be due to a fl ip - fl op phenomenon. Also, when the 
tissue distribution of the formulation was examined 12 h after administration, the 
highest radioactivity was found in the liver. The details of the biodistribution studies 
are as shown in Table 3 . 
The same group [82] further studied the stability of these mono - PEG2000 - sCT 
and the unmodifi ed sCT in the rat nasal mucosa. It was found that PEGylated sCT 
exhibited signifi cant resistance against trypticlike and nonspecifi c enzymatic degradation. 
Ahsan et al. [49] showed that when sCT was formulated with alkylglycosides, 
bioavailability was enhanced following both nasal and ocular administration. Miacalcin 
(Novartis Pharma AS, Huningue, France) was used to prepare the formulation 
FIGURE 13 Blood clearance of PEGylated salmon calcitonins in rat. ( Reproduced from 
ref. 79 with permission of Taylor & Francis. ) 
100 
80 
60 
40 
20
0 
0 10 20 30 40 50 
Time (min) 
60 
Remaining radioactivity % 
Native sCT 
Mono-PEG-sCT 
Di- PEG-sCT 
NASAL DELIVERY OF PEPTIDE AND HIGH-MOLECULAR-WEIGHT DRUGS 615

616 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
FIGURE 14 Average serum concentration of intact sCT – time curves following nasal 
administration of unmodifi ed sCT ( • ) and Mono - PEG2K - sCT (  ) in rats ( n = 9 each). 
( Reproduced from ref. 81 with permission of The Pharmaceutical Society of Japan. ) 
10
1
0 500 1000 1500 2000 2500 
Time (min) 
Serum concentration (ng/mL) 
TABLE 2 Pharmacokinetic Parameters (Mean ± SD) of Unmodifi ed sCT and 
Mono - PEG2k - sCT obtained after nasal administration to rats ( n = 9 each) 
Parameter Unmodifi ed sCT Mono - PEG 2k - sCT 
C max (ng/mL) 10.5 ± 4.7 12.9 ± 3.0 
t max (min) 77 ± 22 520 ± 167 a 
t 1/2, . z (min) 199 ± 97 923 ± 389 a 
CI/F (mL/min) 7.4 ± 5.2 1.3 ± 1.0 a 
V ss / F (mL) 1802 ± 811 1392 ± 450 
AUC (ng · min/mL) 3650 ± 1894 20638 ± 9486 a 
AUC/D (ng · min/mL/ng) 0.18 ± 0.09 1.03 ± 0.48 a 
MRT . a (min) 314 ± 131 1505 ± 560 a 
Source : From ref. 74 . 
a Signifi cantly different from unmodifi ed sCT ( p < 0.05). 
TABLE 3 Extent of Total Radioactivity (Mean ± SD) 
in Various Body Organs after Nasal Administration 
of Unmodifi ed sCT and Mono - PEG2k - sCT to Rats 
( n = 9 each) 
Tissue 
Radioactivity (%) in Whole Organ a 
Unmodifi ed SCT Mono - PEG 2k - sCT 
Liver 0.80 ± 0.41 1.03 ± 0.65 
Kidney 0.30 ± 0.14 0.52 ± 0.24 
Lung 0.18 ± 0.12 0.20 ± 0.12 
Heart 0.06 ± 0.03 0.13 ± 0.05 b 
Spleen 0.04 ± 0.02 0.10 ± 0.05 
Thyroid 0.04 ± 0.03 0.05 ± 0.02 
Source : From ref. 74 . 
a Determined 12 h after administration. 
b Signifi cantly different from unmodifi ed sCT ( p < 0.05). 

in solutions containing different concentrations of tetradecylmaltoside and octylmaltodise. 
These formulations were administered as described by Ahsan et al. [49] . 
It was found that when calcitonin was formulated in saline, absorption after administration 
by the nasal route was negligible; a similar result was seen with 0.125% 
OM. However, when calcitonin was formulated with 0.125% TDM, absorption was 
found to be increased, and maximal absorption ( Tmax ) was achieved after 10 min. 
The AUC 0 – 40 was found to be fourfold higher than with saline and OM formulations. 
The bioavailability was found to be 53% as compared with intravenously administered 
calcitonin at the same dose of 2.2 U. The AUC 0 – 40 was found to be 6250 pg/ 
mL · min as compared with 3500 pg/mL · min when the concentration of TDM was 
increased to 0.25% from 0.125%. It was also found that in the absence of the absorption 
enhancer increasing the amount of calcitonin from 2.2 to 22 U did not increase 
absorption by the nasal route. However, when formulated in the presence of 0.25% 
TDM, there was a 23 - fold increase in the relative bioavailability of calcitonin. Also, 
when calcitonin was formulated with 0.25% TDM and given as nose drops, there 
was a signifi cant reduction in the plasma calcium concentration as compared with 
the saline formulation. 
5.6.6.3 Low - Molecular - Weight Heparins 
Low - molecular - weight heparins (LMWHs) are fragments of natural heparin that 
are obtained by either enzymatic degradation or chemical depolymerization of 
unfractionated heparins (UFHs). The molecular weight of LMWHs, a heterogeneous 
mixture of sulfated glycosaminoglycans, is about one - third that of UFHs. 
Owing to the variations in the distribution of molecular weights, they show differences 
in their affi nity for plasma proteins, activity against factor Xa, and thrombin, 
as well as duration of activity. They have been proven to be useful in the treatment 
and prevention of deep vein thrombosis. Of late, LMWHs have been preferred to 
therapy with conventional heparin. However, one of the main disadvantages of the 
use of LMWHs on a regular, noninstitutional basis is that they must be delivered 
by the subcutaneous or intravenous route. There have been concerns regarding 
patient compliance, longer hospital stays, and the need for skilled health professionals 
for therapeutic drug monitoring and administration. This has prompted a number 
of researchers to seek alternative forms of delivery. 
The nasal administration of LMWHs was investigated in Sprague - Dawley rats 
using TDM as an absorption enhancer [83] . TDM was used at concentrations of 
0.125 and 0.25%. Lovenox (Aventis Pharmaceuticals, Bridgewater, NJ) (enoxaparin 
sodium injection), a commercially available LMWH, was prepared with 0.25% TDM 
and the nasal absorption was compared with and without TDM. It had been previously 
reported that a plasma anti – factor Xa level of 0.20 U/mL or higher is required 
for an antithrombotic effect to be considered therapeutic [83] . It was found that 
enoxaparin when formulated only in saline did not produce a therapeutic anti – factor 
Xa level. However, when enoxaparin was formulated with 0.25% TDM, a signifi cant 
increase in the AUC and Cmax for the anti – factor Xa level was observed. Dalterparin, 
another commercially available LMWH, showed a similar response. When UFH was 
formulated with TDM, it produced an increase in the anti – factor Xa levels as compared 
with saline, but it was not in the therapeutic range (Figure 15 ). 
The bioavailability ( Fabs ) of enoxaparin achieved by the subcutaneous route was 
found to be 83% of that achieved by the intravenous route. In the absence of TDM, 
NASAL DELIVERY OF PEPTIDE AND HIGH-MOLECULAR-WEIGHT DRUGS 617

618 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
the nasal F abs was 4.0%, but in the presence of 0.25% TDM, the F abs was 19% compared 
with the intravenous route. The relative bioavailability ( F rel ) of nasal enoxaparin 
plus 0.25% TDM was found to be 23% compared with the subcutaneous 
route. 
The use of alkanoylsucroses in the enhancement of nasal absorption of LMWHs 
was investigated by our group [84] . As seen in Figure 16 a , enoxaparin plus 0.125% 
octanoylsucrose showed no signifi cant increase in anti – factor Xa levels; even when 
the concentration was increased to 0.5%, it barely reached the therapeutic level of 
0.2 U/mL. Similar results were reported for 0.125% decanoylsucrose; however, when 
the concentration was increased to 0.25 or 0.5%, there was an appreciable and rapid 
rise in anti – factor Xa levels (Figure 16 b ). The inability of octanoylsucroses and 
0.125% decanoylsucrose to increase anti – factor Xa levels is attributed to the critical 
micellar concentration as reported by the authors [84] . In the case of dodecanoylsucroses, 
all the concentrations produced anti – factor Xa levels well above the therapeutic 
level required for an antithrombotic effect (Figure 16 c ). 
Our group also compared the effi cacy and potency of alkanoylsucroses with those 
of the well - known absorption enhancer 1% sodium glycocholate (Figure 16 d ); the 
results were similar to those seen with 0.5% dodecanoylsucrose. Also, 0.5% 
dodecanoylsucrose showed the highest increase in C max , and it was found that an 
increase in the concentration of alkanoylsucroses led to a subsequent increase in 
the C max . When the absolute and relative bioavailabilities of nasal LMWH plus 0.5% 
dodecanoylsucrose were compared with those of 1% sodium glycocholate, similar 
profi les were found [84] . 
FIGURE 15 ( a ) Nasal administration of 100 U of ( A ) enoxaparin, ( B ) dalteparin, or 
( C ) UFH formulated with ( • ) and without (  ) 0.25% tetradecylmaltoside. Data represent 
mean ± SEM, n = 3. Asterisks indicate results that are signifi cantly different from those 
obtained with the drug formulated with saline, P < 0.05. ( b ) Administration of 100 U of 
enoxaparin via the subcutaneous (  ), intravenous (  ), and nasal (  ) routes. Nasal administration 
was performed with a formulation that included 0.25% TDM. Data represent mean 
± SEM, n = 3. ( Reproduced from ref. 83 with permission of John Wiley & Sons. ) 
A. Enoxaparln B. Dalteparin C. UFH 
Anti–factor Xa activity (U/mL) 
0.55 
0.50 
0.45 
0.40 
0.35 
0.30 
0.25 
0.20 
0.15 
0.10 
0.05
0
0 0 0 120 120 120 240 240 240 360 360 360 480 480 480 
Time (min) 
Anti–factor Xa activity (U/mL) 
3.5 
3.0 
2.5 
2.0 
1.5 
1.0 
0.5 
0.0 
0 60 120 180 240 300 360 420 480 
Time (min) 
(a) (b)

FIGURE 16 Changes in anti – factor Xa activity after nasal administration of enoxaparin 
formulated in saline or in presence of different concentrations of the following: ( a ) octanoylsucrose; 
( b ) decanoylsucrose; ( c ) dodecanoylsucrose; ( d ) sodium glycocholate and dodecanoylsucrose 
to anesthetized rats (enoxaparin dose, 330 U/kg). Data represent mean ± SEM, 
n = 3, 5. ( Reproduced from ref. 84 with permission of Pharmaceutical Press. ) 
(a) 
(c) 
(b) 
(d) 
0 100 200 300 400 
0.0 
0.1 
0.2 
0.3 
0.4 
0.5 
0.6 
Anti–Factor Xa activity (U/mL) 
Time (min) 
0.5% Decanoylsucrose 
0.25% Decanoylsucrose 
0.125% Decanoylsucrose 
Saline 
0 100 200 300 400 
0.0 
0.1 
0.2 
0.3 
0.4 
0.5 
0.6 
Anti–factor Xa activity (U/mL) 
Time (min) 
0.5% Octanoylsucrose 
0.25% Octanoylsucrose 
0.125% Octanoylsucrose 
Saline 
0 100 200 300 400 
0.0 
0.1 
0.2 
0.3 
0.4 
0.5 
0.6 
Anti–Factor Xa activity (U/mL) 
Time (min) 
0.5% Dodecanoylsucrose 
0.25% Dodecanoylsucrose 
0.125% Dodecanoylsucrose 
Saline 
0 100 200 300 400 
0.0 
0.1 
0.2 
0.3 
0.4 
0.5 
0.6 
Anti–Factor Xa activity (U/mL) 
Time (min) 
1% Sodium glycocholate 
0.5% Dodecanoylsucrose 
Saline 
The authors also studied the infl uence of the chain length of alkylmaltosides on 
the nasal absorption of enoxaparin. The results indicated that increases in the concentration 
of alkylmaltosides increased the AUC for plasma anti – factor Xa; it was 
found that the absolute and relative bioavailabilities of enoxaparin increased by 
twofold with an increase in alkyl chain length from 8 to 14 carbons. Of all the alkylmaltosides, 
TDM was found to be the most potent absorption enhancer [85] . 
Furthermore, we have also shown the effi cacy of cyclodextrins in enhancing 
absorption following the nasal delivery of LMWHs. Three different cyclodextrins 
were employed: . - cyclodextrins ( . - CD), hydroxypropyl . - CD (HP . - CD), and 
dimethyl . - CD (DM . - CD). . - CD showed therapeutic levels of anti – factor Xa only 
at 2.5 and 5% . - CD, but there was no signifi cant difference between the two concentrations, 
which was attributed to their solubility limit. In the case of HP . - CD, 
neither 1.25 nor 2.5% produced an appreciable increase in anti – factor Xa levels; 
NASAL DELIVERY OF PEPTIDE AND HIGH-MOLECULAR-WEIGHT DRUGS 619

620 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
only 5% HP . - CD showed levels above 0.2 U/mL, which, however, was not signifi - 
cant. Unlike the other two cyclodextrins, 1.25 and 2.5% DM . - CD showed a fourfold 
increase in AUC profi les. The studies reported that 5% DM . - CD produced the 
greatest increase in the bioavailability of LMWHs, with an eightfold increase in the 
AUC profi le. It was also found that the reduction in transepithelial electrical resistance 
(TEER) and changes in tight junction protein ZO - 1 distribution facilitated 
by 5% DM . - CD were much greater than with . - CD or HP . - CD [86] . Recently our 
group has shown that positively charged polyethylenimines (PEI) increases the 
nasal absorption of LMWHs by reducing the surface negative charge of the drug. 
When PEI 1000 kDa was employed at a concentration of 0.25%, a four - fold increase 
in the absolute and relative bioavailabilities was observed in comparison with the 
control formulation of LMWH plus saline [87] . 
5.6.6.4 Azetirelin 
Azetirelin is a novel analog of the tripeptide thyrotropin - releasing hormone 
(TRH). It was discovered in 1969 when two different groups of researchers, led by 
Guillemin and Schally, showed that the hypothalamic substance that causes the 
anterior pituitary gland to release thyrotropin (thyroid - stimulating hormone, or 
TSH) is l - pyroglutamyl - l - histidyl - l - prolineamide ( l - pGlu - l - His - l - ProNH2), now 
called TRH. In azetirelin the pyroglutamyl moiety of the TRH is replaced by an 
(oxo - azetidinyl) carbonyl moiety. It has been reported that the inhibition of 
pentobarbital - induced sleep and reserpine - induced hypothermia due to azetirelin 
in mice, as opposed to TRH, are about 10 – 100 times more effective as well as 8 – 36 
times longer lasting. Azetirelin is stable in plasma and degrades much more slowly 
than TRH in brain homogenates, thus showing improved pharmacological potency 
as well as effi cacy over TRH. It is highly potent when given intravenously; however, 
when administered by the oral route, it shows very low bioavailability of only 2% 
[88 – 91] . 
Kagatani et al. [90] studied the effect of acylcarnitines as drug absorption enhancers 
for the nasal delivery of azetirelin in a rat model. A buffered azetirelin sample 
solution was administered intranasally, as described previously [47] . The nasal and 
oral absorptions of azetirelin were then compared. The Fabs after nasal absorption 
was found to be 17.1%, which was 21 times greater than the 0.8% after oral administration. 
As reported above, a pilot study of oral azetirelin showed a bioavailability 
of about 2%. A bioavailability of about 20% was seen in the case of nasally administered 
TRH in humans as well as rats. The authors predicted that since azetirelin 
is an analog of TRH, its pharmacokinetic properties after nasal delivery in humans 
could also be about 20% [90, 91] . 
5.6.6.5 Growth Hormones 
Recombinant human growth hormone (hGH) is a 22 - kDa protein drug having 191 
amino acids. It has been used to treat a number of conditions, including short stature 
in children, Turner syndrome, and chronic renal failure. It is said to play an important 
role in the metabolism of proteins, carbohydrates, and fats as well as electrolytes 
and hence infl uences weight and height. It has been reported that hGH secretion 
in humans is pulsatile, showing low basal serum levels in between peaks. It has been 

suggested that this secretory pattern of hGH can be mimicked by the nasal route 
as opposed to painful subcutaneous injections [8, 92, 93] . 
In a pharmacokinetic - based study by Hedin et al. [94] , hGH was administered 
with a nasal permeation enhancer, sodium tauro - 24, 25 - dihydrofusidate (STDHF), 
in patients defi cient in growth hormone (GH) using a reprocessed lyophilized form 
of hGH. The lyophilized material was formulated with STDHF and all the subjects 
received the formulation by both the nasal and subcutaneous routes. The dose given 
by the subcutaneous route was a standard dose of 0.1 IU/kg body weight (BW), 
whereas three different doses (of 0.2, 0.4, and 0.8 IU/kg BW) of the nasal formulation 
were given. As compared with the subcutaneous route, all three nasal formulations 
showed a rapid increase in the plasma levels of hGH, with Tmax being reached 
15 – 25 min after administration, as compared with 3 – 4 h in the case of the subcutaneous 
route. However, the Cmax was higher in the case of the latter route, and the nasal 
formulations touched baseline after 3 – 4 h, as compared with 14 – 18 h after subcutaneous 
delivery. 
Several studies have shown that frequent doses of hGH are more benefi cial than 
the total amount given as a single dose and that higher peaks of plasma hGH with 
low troughs are found in taller children; hence the nasal therapy for defi ciencies in 
hGH would not only be more convenient but also offer advantages, including a rapid 
decrease of the peaks to zero levels and a mimicking of the pulsatile pattern of the 
endogenous hormone [94 – 97] . Nasal irritation studies also were carried out, indicating 
that the nasal formulations showed only local short - term irritation. 
Didecanoylphosphatidylcholine (DDPC) and . - cyclodextrin ( . - CD) were used 
as enhancers and reversibility studies were carried out in vivo in rabbits. Three different 
combinations were used: DDPC, . - CD, and DDPC plus . - CD for the nasal 
administration of hGH. Vermehren et al. [98] used intravenous hGH as the reference. 
When hGH was administered with . - CD as a powder, 23.6% bioavailability 
was seen, as compared with 18.1% Frel when given at the same time as two powders. 
When hGH only was given, a bioavailability of 8.3% was attained. DDPC plus hGH 
together showed a Frel of 22.3 and 21.5% when given as two powders, while simultaneous 
administration of DDPC plus . - CD and hGH as two powders showed a 
Frel of 14.3%. When dosed as one single powder, it showed a Frel of 31.9%. Reversibility 
of the enhancer effect was seen when a dose of hGH in enhancer - free formulation 
was given 30 min after dosing of the test formulations; this resulted in 
reduction in the AUC and Cmax by half. 
Another group studied the enhancer effect of DDPC on the pharmacokinetics 
and the biological activity of nasally administered hGH in GH - defi cient patients. 
Three different doses — 0.05, 0.10, and 0.20 IU/kg with DDPC — were given by the 
nasal route and 0.10 and 0.015 IU/kg were given by the subcutaneous and intravenous 
routes. A short - lived serum GH peak was seen in the intravenous treatment, 
showing a peak value of around 128 . g/L, whereas the subcutaneous route showed 
a peak level of 13.98 . g/L and nasal doses showed peaks of about 3.26, 7.07, and 
8.37 . g/L for the three treatment doses. The bioavailabilities of the nasal doses were 
found to be 7.8, 8.9, and 3.8%, respectively, as compared with the Fabs of 49.5% for 
the subcutaneous dose. It was also found that the serum insulinlike growth factor 1 
(IGF - 1) increased only upon subcutaneous administration There was no change in 
the serum IGF protein binding protein 3 levels in any of the nasal doses or in the 
subcutaneous or intravenous doses [92] . 
NASAL DELIVERY OF PEPTIDE AND HIGH-MOLECULAR-WEIGHT DRUGS 621

622 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
Leitner et al. [93] tried to overcome the limitations posed by the low bioavailability 
of nasally delivered hGH due to the drug ’ s high molecular weight and 
hydrophilicity. The strategy employed was to use dry polymer particles to enhance 
absorption, taking advantage of the fact that these polymers would form a gellike 
layer and hence be cleared slowly, giving a longer circulating drug. Due to the presence 
of covalent immobilization of sulfhydryl groups on the backbone of the thiolated 
polymers or thiomers, the permeation - enhancing properties of these thiomers 
were improved. They have also been reported to have mucoadhesive and enzyme - 
inhibitory properties, and the addition of glutathione (GSH) was found to increase 
their permeation - enhancing properties further [93, 99 – 103] . A polycarbophil – 
cysteine (PCP – Cys) microparticulate system was prepared using GSH and hGH, 
and this was added to the formulation. The in vitro release profi les of the PCP – Cys 
and unmodifi ed PCP containing hGH proved to be similar. Three different formulations 
were tested for in vivo nasal absorption: PCP – Cys/GSH/hGH microparticles, 
PCP/hGH microparticles, and mannitol/hGH powder against a subcutaneous hGH. 
The Frel for mannitol/hGH powder was around 2.40%, the PCP – Cys/GSH/hGH 
microparticles showed a Frel of 8.11%, and PCP/hGH showed 2.70%, which represents 
a threefold increase in nasal uptake when thiomer/GSH is used in the formulation. 
The microparticulate formulation of PCP – Cys/GSH/hGH showed a sixfold 
higher plasma concentration when compared to the PCP – Cys/GSH/hGH gel 
formulation. 
5.6.7 NASAL DELIVERY OF NONPEPTIDE MOLECULES 
5.6.7.1 Morphine 
Morphine is a potent narcotic analgesic used preoperatively and as an anxiolytic 
agent in pediatric patients; it is also used in the management of postoperative pain 
as well in moderate to severe pain in cancer. Oral morphine in solution, immediate - 
release, or controlled - release formulations shows a bioavailability of only about 
20%. Morphine absorption in humans is poor, and only 10% bioavailability is 
obtained when it is given as a solution rather than intravenously. Chitosan has been 
reported to be a potent absorption enhancer and greatly improves the absorption 
of small polar molecules and peptides. Intranasal administration was carried out in 
a sheep model; various formulations were also tested in humans and comparisons 
versus the intravenous route were made. The sheep model was used for nasal delivery 
because it has been reported that testing in sheep is highly predictive of results 
in humans [104, 105] . In sheep studies, when morphine HCl solution was given 
nasally (control), the Cmax obtained was limited: 151 n M with a Frel of 10%, with a 
Tmax of about 20 min, suggesting a slow rate of absorption. When 0.5% chitosan was 
formulated with morphine as a solution, the Cmax increased to 657 n M , and Frel was 
26.6%. The rate of absorption was also increased, as evidenced by a Tmax of 14 min. 
With the formulation of chitosan into microspheres, the Cmax was 1010 n M, Tmax 
about 8 min, and Frel about 54.6%. A further increase in nasal absorption was seen 
when morphine was formulated as a powder consisting of starch microspheres and 
L - . - lysophosphatidylcholine (LPC), with Cmax being 1875 n M, Tmax about 10 min, and 
Frel about 75% (Figure 17 ). In the case of human phase I clinical trials, a dose of 

10 mg morphine sulfate led to a mean C max of 336 n M following 30 min of intravenous 
administration. A 0.5% chitosan and morphine HCl solution led to a C max of 98 n M 
and a T max of about 16 min. The plasma half - life ( t 1/2 ) in the case of nasal administration 
was found to be 2.98 h, as compared with 1.67 h via the intravenous route. The 
mean bioavailability with the nasal solution of morphine plus chitosan was 56%. 
Furthermore, the powder formulation comprising chitosan and morphine HCl 
showed a C max of 92 n M, T max of 21 min, t 1/2 of 2.72 h, and F rel of about 56% (Figure 
18 ). The nasal formulations were reported to be well tolerated. 
5.6.7.2 Benzodiazepines 
Diazepam has been the standard or preferred option for the treatment of all types 
of seizures in both children and adults. However, it has disadvantages, including a 
short duration of action, so that in some cases diazepam must be given rectally in 
order to manage prolonged seizures. Moreover, its use in the community is restricted 
because of the need for privacy, especially in the case of adult patients. Finally, the 
intravenous route is also reported to be inconvenient, since nonprofessional caregivers 
may not be comfortable enough to administer the drug in this way. Midazolam 
has been reported to be clinically effective with both intravenous and oral administration 
for the induction of sedation and reduction of anxiety. Owing to the drawbacks 
mentioned above, intranasal formulations of benzodiazepines could be highly 
benefi cial [106, 107] . 
FIGURE 17 Morphine plasma concentration after nasal administration of morphine formulations 
in sheep: Mor Sol, morphine solution; Mor Chi Sol, morphine solution containing 
chitosan; Mor Chi PWD, morphine chitosan powder; Mor SMS LPC, starch microspheres 
with lysophosphatidylcholine and morphine as a freeze - dried powder. ( Reproduced from 
ref. 105 with permission of the American Society for Pharmacology and Experimental 
Therapeutics. ) 
1500 
1000 
500
0 
0 20 40 60 80 100 120 
Time (min) 
Morphine plasma concentration (nmol/L) 
Mor Sol (F2) 
Mor Chi Sol (F3) 
Mor Chi PWD (F4) 
Mor SMS LPC (F5) 
NASAL DELIVERY OF NONPEPTIDE MOLECULES 623

624 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
Midazolam, Triazolam, and Flurazepam The feasibility of intranasal administration 
of midazolam, fl urazepam, and triazolam has been studied and compared with 
oral absorption in dogs. There was a 3.4 - fold increase in the C max after nasal administration, 
from 5.5 – 8.7 ng/mL to 17.4 – 30.0 ng/mL. The mean t 1/2 showed comparable 
values for both routes. The T max obtained after nasal administration of midazolam 
was found to be 15 min, as compared with the 15 – 45 min observed for oral dosing, 
while the C max after nasal administration was 6.5 – 20.3 ng/mL, as compared with 
3.0 – 8.6 ng/mL observed for the oral route. Like midazolam and triazolam, fl urazepam 
also showed a shorter half - life, 15 min, as compared with 15 – 45 min with oral 
administration. The C max for oral administration was 0.14 – 0.59 ng/mL; after nasal 
administration it was in the range of 2.6 – 11.1 ng/mL, a 16.4 - fold increase. Since the 
gastrointestinal tract at bedtime is likely to be in the fed state, causing a twofold 
decrease in the absorption of midazolam and triazolam, the nasal route may be a 
better option for the treatment of amnesia, since these drugs cross the nasal mucosa 
effectively without the use of an absorption enhancer, as shown in these studies 
[108] . 
In situ nasal absorption studies of midazolam were carried out in rats. The effects 
of solution concentration, osmolality, and pH on nasal absorption were studied using 
the in situ perfusion technique. The absorption of midazolam was reported to be 
prevented at osmolalities in the range of 142 – 450 mOsm/kg; however, a hypoosmotic 
3 - mOsm/kg solution resulted in signifi cant absorption, where the pH rose from 3.3 
to 6.5. No lag time in absorption was observed when the solutions were buffered at 
a pH of either 5.5 or 7.4; however, at pH 3.3, no absorption was seen, suggesting 
FIGURE 18 Morphine plasma concentration in human volunteers after intravenous administration 
of morphine and after nasal administration of morphine as chitosan solution and 
powder formulations: Mor Chi Sol, morphine solution containing chitosan; Mor Chi PWD, 
morphine – chitosan powder; IN, intranasal. ( Reproduced from ref. 105 with permission of the 
American Society for Pharmacology and Experimental Therapeutics. ) 
IV Mor 
IN Mor Sol 
IN Mor PWD 
Morphine plasma concentration (nmol/L) 
1000 
100 
10
1 
0 2 4 6 8 10 12 
Time (h)

that pH was the main factor determining the absorption of midazolam (Figure 19 ) 
[109] . 
The pharmacokinetics and pharmacodynamics of midazolam after nasal administration 
were investigated in healthy volunteers in two different studies in comparison 
with the intravenous route [110, 111] . Studies reported in 1997 demonstrated 
that intranasal midazolam was rapidly absorbed, with maximal concentration 
attained in the range of 10 – 48 min, with a mean of 25 min. These results were only 
the maximum concentration achieved and the time taken to reach this maximum. 
The maximum concentration reported after intranasal administration was in the 
range of 91.0 to 224.3 ng/mL, with a mean of 147 ng/mL. Bioavailability reported in 
this study [110] was about 50%, in line with the results obtained with oral administration, 
as reported in an earlier study [112] . Knoester et al. [111] also carried out 
similar studies using a concentrated intranasal spray in healthy volunteers. The 
concentrated preparation was prepared by mixing midazolam HCl in a mixture of 
water and propylene glycol, pH 4. A Spruyt Hillen (IJsselstein, Netherlands) intranasal 
device was used to deliver the dose. Besides nasal irritation lasting 10 min and 
teary eyes, no other discomfort was reported. Midazolam was rapidly absorbed on 
nasal administration, showing a maximum concentration of 72 ng/mL within 14 min. 
A mean bioavailability of 0.89 was obtained. It was reported that intraindividual 
basal electroencephalogram (EEG) activity after intranasal administration was 
comparable with that after intravenous administration (Figures 19 and 20 ) . 
Diazepam As mentioned earlier, because of shortcomings of rectal administration, 
the nasal delivery of diazepam has gained interest. The nasal bioavailability of 
diazepam in sheep was estimated and further compared with results obtained earlier 
in humans and rabbits [106] ; in this study, human and rabbit nasal bioavailability 
for the fi rst 30 min was reported to be 37 and 54%, respectively [113] . Diazepam 
solubilized in PEG 300 was used for nasal administration via a modifi ed nasal device, 
a Pfeiffer unit dose (Princeton, NJ). The sheep received the nasal formulations in a 
fi xed standing position such that the head was slightly tilted back. It was found that 
the serum concentration after administration of a 7 - mg solution of diazepam was 
FIGURE 19 Fit of composite model to concentration – time data for midazolam and 1 - 
hydroxymidazolam in one volunteer. Solid lines indicate the time course of midazolam concentrations 
(  ) and 1 - hydroxymidazolam concentrations (  ) after intravenous administration. 
Dotted lines indicate the time course of midazolam concentrations (  ) and 1 - hydroxymidazolam 
concentrations (  ) after intranasal administration. ( Reproduced from ref. 111 with 
permission of Blackwell Publishing. ) 
1000 
100 
10
1 
0.1 
0 60 120 180 240 300 360 420 480 540 
Time (min) 
Concentration (.g/L) 
NASAL DELIVERY OF NONPEPTIDE MOLECULES 625

626 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
lower than that obtained with a 3 - mg solution, suggesting a low nasal bioavailability. 
The bioavailability after the initial 30 min was found to be 15%, as compared to the 
earlier mentioned bioavailability in human and rabbit, and a C max of 934 ng/mL with 
a T max of 5 min. The difference in bioavailability between animal and human was 
larger when periods shorter than 0 – 60 min were used in the calculations. When the 
results among the three species were compared, it was found that the bioavailability 
in sheep was higher than that in humans during the early or initial phases, after 
which the reverse was observed. Lindhardt et al. [113] used a profi le that took into 
account the observation period with respect to the rate of bioavailability and found 
that similar profi les with respect to rate were observable in relation to all of the 
three nasal formulations given in humans; moreover, an optimal correlation between 
sheep and rabbit was observed. The authors suggested that use of the jugular vein 
in sampling blood from sheep could have resulted in the low bioavailability. 
5.6.7.3 Buprenorphine 
Buprenorphine is a derivative of the morphine alkaloid thebaine and is a partial 
opioid antagonist. It exerts an agonistic effect on the . - muscarinic receptors and an 
antagonistic effect on the . type. Buprenorphine at lower doses produces suffi cient 
agonist effect to enable opioid - addicted individuals to discontinue the use of opioids 
without experiencing withdrawal symptoms. It has been reported that an intravenous 
dose of 0.3 mg of buprenorphine is equivalent to 10 mg of morphine and that 
oral delivery of buprenorphine results in a bioavailability of only about 15% due 
to fi rst - pass metabolism. In addition to the intravenous formulation, there is a sublingual 
formulation offering the advantage of avoiding the fi rst - pass metabolism 
effect. A clinical trial of nasally administered buprenorphine was reported in 1989 
[114] . Buprenorphine is highly lipophilic and hence easily absorbed across the nasal 
epithelium. The buprenorphine formulation used in the clinical trial was prepared 
in 5% dextrose solution and a Pfeiffer atomizing pump operated manually was used 
to deliver it. The mean T max and C max for the intranasal dose were 30.6 min and 
1.77 ng/mL, respectively. A relative nasal bioavailability of about 48.2% was attained. 
FIGURE 20 Individual plasma concentration – time curves for midazolam (solid lines) and 
1 - hydroxymidazolam (broken lines) after intranasal administration of 5 mg midazolam. The 
bold curves represent the mean pharmacokinetic model fi t to the data. ( Reproduced from 
ref. 111 with permission of Blackwell Publishing. ) 
1000 
100 
10
1 
0.1 
Concentration (.g/L) 
0 60 120 180 240 300 360 420 480 540 
Time (min)

Butorphanol, an analog of buprenorphine, showed a nasal bioavailability of 70% 
and also a much lower Tmax after nasal absorption as compared with the sublingual 
and buccal routes [115] . Lindhardt et al. [106] compared buprenorphine formulated 
in 30% PEG - 300 in sheep with that of the 5% dextrose formulation mentioned 
earlier. A unit - dose Pfeiffer device was again used to administer the formulation. It 
was found that nasal bioavailability in sheep was about 70% when buprenorphine 
was formulated in PEG - 300 and approximately 89% when it was formulated with 
5% dextrose. The rate of absorption was reported to be very fast, with a Tmax of 
10 min; the Cmax was found to be 37 and 48 ng/mL for PEG - 300 and dextrose, respectively. 
In sheep, the pharmacokinetics of buprenorphine showed a two - compartment 
model as compared to a three - compartment model in humans. 
5.6.7.4 Hydralazine 
Hydralazine is a vasodilator used in the treatment of malignant hypertension and 
hypertensive emergencies and is generally used in conjunction with other antihypertensive 
agents. Although the oral absorption is good, there is low oral bioavailability 
due to fi rst - pass metabolism. Nasal absorption of hydralazine has been 
studied in rats using both in vivo nasal absorption and in situ nasal perfusion 
methods; the effect of surfactant and solution pH has also been reported. It was 
found that the nasal absorption of hydralazine was increased in the presence of 
surfactants such as sodium glycocholate and polyoxyethylene - 9 - lauryl ether. The 
nasal absorption of hydralazine was reported to be a pH - dependent passive process, 
with the absorption increasing as the pH was increased from 3.0 to 6.6. In nasal 
absorption studies in rats, peak levels of hydralazine were reached in 30 min at pH 
3.0. The in situ absorption of hydralazine as a function of perfusion pH was also 
evaluated. The results of the in situ nasal perfusion studies demonstrated that 
hydralazine is eliminated from the nasal cavity and the perfusate by fi rst - order 
kinetics. Even in the ionized form, the drug was well absorbed, and it was suggested 
that the aqueous channels in the nasal mucosa played an important role in the 
transport of hydralazine [116, 117] . 
5.6.7.5 Nitroglycerin 
Nitroglycerin is delivered across the mucosal membranes in the management of 
acute ischemic conditions. Nitroglycerin carries out this function by arterial vasodilatation 
and venodilation, which leads to a decrease in both the preload and afterload 
and also improved coronary blood fl ow. The intranasal action of nitroglycerin, 
also called glyceryl trinitrate, appears to be similar [118] and brings about a reduction 
in myocardial oxygen consumption. Like that of hydralazine, the oral bioavailability 
of nitroglycerin is low; hence alternative routes of delivery and innovative 
delivery systems have been preferred, such as sublingual patches, ointments, or 
transdermal patches. The intravenous route ensures a rapid onset of action, but its 
preparation and standardization procedures make it costly. Intranasal nitroglycerin 
in various operative experiences has been found to have a rapid onset of action with 
predictable and consistent therapeutic effects. A peak level of nitroglycerin is 
reached 1 – 2 min after intranasal administration; it is barely detectable after 16 min. 
These studies were carried out in fi ve patients who were undergoing elective coro- 
NASAL DELIVERY OF NONPEPTIDE MOLECULES 627

628 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
nary artery bypass surgery. The plasma levels were reported to be similar with 
intravenous administration and better than with sublingual administration [118 – 
120] . The pressor response to endotracheal intubation in both normotensive and 
hypertensive patients can be attenuated by intranasal nitroglycerin in operative 
settings. It has been reported that when nitroglycerin was given 30 s before the 
induction sequence in 40 hypertensive patients treated with . blockers, there was a 
blunted pressor response to intubation. In comparison to the placebo control group, 
the group that received nitroglycerin had a lower mean arterial pressure at 1, 3, and 
5 min after induction. Thus intranasal nitroglycerin can be employed in the selective 
control of hypertension [118, 121, 122] . 
5.6.7.6 Propranolol and Other . - Adrenergic Blocking Agents 
Propranolol is a nonselective . - adrenergic receptor blocking agent. It is clinically 
used in the management of hypertension and the treatment of angina pectoris. When 
given orally in humans, it has led to considerable variation in plasma drug levels. 
This, as well as its subsequently low bioavailability, is believed to be due to its extensive 
metabolism in the gut and in the liver. A study in the late 1970s showed that 
nasal absorption of propranolol at a dose of 1 mg in rats produced blood levels 
similar to those achieved with intravenous administration; however, the same dose 
administered orally resulted in very low blood levels [47, 123] . The feasibility of 
nasal absorption of propranolol in solutions and sustained - release formulations in 
rats and dogs was studied. The procedure described by Hirai et al. [46] was used to 
carry out the surgical operation in rats. In dogs, the formulations were given intranasally 
using a micropipette and syringe. The mean blood levels of propranolol by 
the nasal route were compared with those of the oral and intravenous routes in rats 
and dogs. As in the case of the results obtained in humans, oral administration of 
propranolol solutions resulted in low and variable drug levels in rats and dogs, 
whereas the nasal administrations of propranolol solution showed plasma drug 
levels that were similar to those achieved with intravenous administration. In the 
case of sustained - release formulations, it was found that there was an initial low 
level of drug; however, these levels were maintained for a longer time. The bioavailabilities 
obtained from the AUC were found to be identical, although the maximum 
blood levels in the case of sustained - release formulations were found to be much 
lower than with the propranolol solutions. A propranolol formulation containing 
2% methylcellulose gels in humans was studied by the same group. Identical 
serum drug profi les were obtained after nasal administration as with intravenous 
administration [124] . 
The effect of intranasal propranolol on exercise parameters with the Bruce protocol 
[118, 125] was studied in 16 patients with chronic, stable, effort - induced angina 
pectoris. Propranolol was given as a single 5 - mg/puff nasal spray to the patients. A 
mean plasma level of 20 ng/mL was obtained, and a signifi cant increase in total 
exercise time was seen, from 460 to 530 s. This led to an increase in the time to 1 mm 
ECG as well as to the onset of angina. Both maximum heart rate and systolic blood 
pressure were lower than with placebo. This study demonstrated that propranolol 
in the form of a nasal spray elicited immediate . blockade and was useful in treating 
patients with angina pectoris, who showed improvement in exercise tolerance after 
receiving the drug [118, 125] . 

Although no adverse reactions have been reported with intranasal administration 
of propranolol, complications may occur, as ocular administration has produced 
some systemic side effects [118] . The infl uence of substrate lipophilicity on drug 
uptake by the nasal route was reported in humans. Alprenolol and metoprolol, . 
blockers with varying degrees of lipophilicity, were used. The fi ndings from these 
studies demonstrate that the more hydrophilic drugs showed a lower bioavailability. 
Alprenolol showed rapid uptake into the systemic circulation by the nasal route and 
also a higher bioavailability [126, 127] . 
5.6.7.7 Sex Hormones 
The low oral bioavailability of hormone - replacement drugs due to intestinal and 
fi rst - pass metabolism requires the use of higher doses of these drugs, which are 
associated with many side effects. Parenteral administration of sex steroids as well 
as use of the transdermal route has been viewed as an alternative. However, the 
transdermal route has certain limitations, such as the visibility and palpability of the 
patch as well as possible skin irritation. These drawbacks have limited the use of 
this route. The intranasal route has therefore been considered as an alternative 
[128] . 
5.6.7.8 17 b - Estradiol ( E 2 ) 
The most common form of estrogen in clinical use is 17 . - estradiol. It is reported 
to reduce bone turnover, prevent postmenopausal bone loss, and decrease the risk 
of fracture in both early and late postmenopausal women. Lipophilic drugs such 
as sex steroids pose the problem of going into solution, thus leading to low estrogen 
levels. Hence a number of studies have attempted to solubilize 17 . - estradiol 
using DM . - CD for its intranasal delivery [129, 130] . An E 2 spray has been developed 
(S21400 or Aerodiol, Institut de Recherches Internationales Servier, France), 
which is estradiol that has been solubilized in water with randomly methylated . - 
CD. Estradiol delivered intranasally is rapidly absorbed by the nasal mucosa and 
shows maximum plasma levels within 10 – 30 min. Plasma levels return to 10% of 
the maximum plasma concentration within 2 h of administration and to untreated 
postmenopausal levels within 12 h. Hence after intranasal administration, estradiol 
has a pulsatile profi le as compared to the more sustained plasma profi le seen with 
the oral and transdermal routes. However, whether sustained levels are required 
for effi cacy has not been determined, although intranasally delivered estradiol does 
increase serum estradiol to the same extent as is seen with oral administration 
[128, 130 – 134] . 
The short - and long - term effects of intranasal 17 . - estradiol on bone marrow 
turnover and serum IGF - 1 were studied in a double - blind placebo - controlled clinical 
trial and compared with oral 17 . - estradiol. Some 425 Caucasian postmenopausal 
women with climacteric symptoms were studied. The nasal effi cacy of estradiol was 
assessed using the Kupperman index (KI), which is a weighted evaluation of the 
incidence and severity of 11 menopausal symptoms summarized in a menopausal 
index as follows: hot fl ushes, the most heavily weighted ( . 4); night sweats ( . 2); and 
sleep disturbances and nervousness (each . 2). The lower weighted symptoms 
include depression, irritability, vertigo, fatigue, arthralgia, headache, tachycardia, 
NASAL DELIVERY OF NONPEPTIDE MOLECULES 629

630 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
and vaginal dryness (each . 1). The highest possible score is 51 [128, 135 – 137] . 
Various markers of bone resorption — such as urinary type I collagen telopeptides, 
the formation of serum osteocalcin, serum type I collagen, N – terminal extension 
propeptide (PINP), and serum bone marrow alkaline phosphatase (BAP) — were 
determined at baseline and after 1, 3, and 15 months. Urinary - type collagen C telopeptides 
were considerably reduced in all the treated groups within 1 month, and 
this reduction continued even at 3 months. Neither serum osteocalcin nor PINP 
showed any change at 1 month; however, they were reduced at 3 months with oral 
dosage. There was an increase in the bone formation parameters at 1 month for the 
higher doses of intranasal estradiol, but no reduction was seen at 3 months. No signifi 
cant change from placebo - treated groups was observed at the end of 3 months 
in the case of circulating IGF - I after intranasal estradiol, but a signifi cant decrease 
was seen with oral estradiol. After a year of treatment with intranasal estradiol at 
a dose of 300 . g/day, resorption and formation markers decreased to premenopausal 
levels. This study concluded that normalization of bone turnover to premenopausal 
levels can be achieved following 1 year of treatment with intranasal 
17. - estradiol [131] . 
5.6.7.9 Testosterone 
Testosterone, the most potent natural male sex hormone, is generally given intramuscularly. 
It is absorbed well orally but is extensively metabolized in the liver 
and the gastrointestinal tract. Owing to fl uctuations in serum levels of testosterone 
esters, other viable routes of delivery are being explored. Two transdermal patches 
have become available commercially. However, due to the limitations associated 
with this route, it is not preferred by patients, particularly because the site of 
application is the scrotum [8, 138] . In order to improve systemic bioavailability, 
the nasal absorption of testosterone has been evaluated versus intravenous and 
intraduodenal administration in rats. When given nasally, the concentration of 
testosterone in the circulation increased and peak levels were reached within 
2 min; blood levels were similar to those seen with intravenous administration. 
Intraduodenal administration produced low blood levels. A bioavailability of 99% 
at 25 . g/dose was seen and 90% at 50 . g, but the intraduodenal route showed a 
very low bioavailability of 1% [8] . Hussain et al. [139] showed in rats that testosterone 
can be absorbed intranasally, and an elimination half - life of about 40 min 
was obtained. However, since the problem of solubility is an obstacle to preparing 
the formulation for nasal administration, the use of a prodrug was evaluated by 
the same group. A water - soluble ester of testosterone, testosterone 17 . - N, N - 
dimethylglycinate hydrochloride (TSDG), is completely absorbed when given 
intranasally at much lower doses as compared with testosterone itself. After 
absorption, conversion of the prodrug to testosterone begins almost immediately, 
and the terminal elimination half - life of testosterone was found to be 55 min, 
which is similar to that obtained after intravenous administration. Peak plasma 
concentration was reached within 12 min for the lower dose (equivalent to 
25 mg/kg of TS) and 20 min for the higher dose (50 mg/kg) by both routes. The 
AUC also showed similarity, suggesting the complete absorption of the nasally 
administered prodrug. 

5.6.8 THE NOSE: OPTION FOR DELIVERY OF DRUGS TO CENTRAL 
NERVOUS SYSTEM 
The possibility of delivering agents to the central nervous system (CNS) via the nose 
has long been known, an example being the sniffi ng of cocaine in order to produce 
a sense of euphoria, which is attained within 3 – 5 min [53] . In short, nasal administration 
is not only an exciting possibility in the fi eld of drug delivery but may also be 
the means of solving delivery problems for the innumerable agents that cannot cross 
the blood – brain barrier (BBB). Such agents are therefore being developed by 
nanoparticle - based systems or by formulating them as prodrugs. The literature 
offers numerous examples demonstrating ways of delivering such drugs to the brain. 
In particular, use of a direct pathway, as in the case of cocaine, from the nasal cavity 
to the CNS has been suggested. Illum [140] and Chow et al. [141] have shown in 
animal models that, in the early time points after nasal administration, the brain 
concentration of cocaine was higher than when the drug was given by the intravenous 
route. The specifi c site through which nose - to - brain delivery is believed to take 
place is the olfactory region. In the early 1900s it was shown that the olfactory region 
was responsible for the uptake (or rather entry) of viruses, in particular the poliomyelitis 
virus, into the brain [142 – 146] . Further work in support of this theory then 
demonstrated the presence of the poliomyelitis virus in the cerebrospinal fl uid 
(CSF) and also in the systemic lymphatics of the olfactory mucosa [147 – 150] . In the 
period from 1970 to 1990, there were many reports of nose - to - brain delivery across 
the olfactory epithelium for a number of different agents, including metals and 
tracer materials such as colloidal gold, cadmium, potassium ferricyanide, and iron 
ammonium citrate [151 – 154] . 
There are certain aspects of drug delivery that must be clearly understood in 
designing a nose - to - brain drug delivery system. For example, lipophilic drugs are 
absorbed across the nasal epithelium almost immediately and effi ciently to enter 
the systemic circulation. Therefore such drugs will show little sign of direct nose - to - 
brain delivery. Drugs that are on the hydrophilic side or polar molecules will not be 
readily absorbed across the nasal mucosa; these molecules generally undergo paracellular 
transport as compared to transcellular transport in the case of lipophilic 
drugs. Such molecules have a higher chance of being taken up by the olfactory 
mucosa for delivery to the brain. In general, drugs travel from the nose to the brain 
via (1) drug internalization into the olfactory epithelium ’ s primary neurons followed 
by the intracellular axonal transport to the olfactory bulb or (2) drug absorption by 
paracellular or transcellular pathways across the olfactory sustentacular epithelial 
cells, following which the drug enters the CSF or CNS. In the human nervous system, 
the olfactory region is the only site in direct contact with the surrounding environment. 
It has been reported that the intracellular axonal pathway takes a longer time 
to deliver drugs to the brain [140, 155 – 157] . Table 4 gives an account of some of the 
drugs/molecules that have been delivered from the nose to the brain in human and 
animal models. The direct delivery of drugs to the brain or CSF via the olfactory 
epithelium is discussed in the following paragraph , briefl y describing the olfactory 
mucosa. 
The olfactory region is mainly involved in the detection of smell, and the makeup 
and organization of the epithelial layer enhance the accessibility of air to the 
OPTION FOR DELIVERY OF DRUGS TO CENTRAL NERVOUS SYSTEM 631

632 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
TABLE 4 Nose - to - Brain Delivery of Agents in Different Species 
Drug/Molecule Path Followed Reference 
Humans 
Arginine - vasopressin — a 158 
Adrenocorticotrophin — a 159 
Cholecystokinin — a 160 
Diazepam — a 161 
Insulin — a 158 , 162 
technetium - 99m - 
diethylenetriaminepentacetic 
acid 
Direct from nose to brain 163 
Apomorphine Nasal cavity > CSF 164 
Melatonin/hydroxycobalamin Nasal cavity > CSF 165 
Melanocortin, vasopressin, and 
insulin 
Nasal cavity > CSF 166 
Rats 
Zidovudine Nasal cavity > CSF, nasal cavity > systemic 
circulation 
167 
Dextromethorphan HCl Direct from nose to brain 168 
Cephalexin Nasal cavity > CSF, nasal cavity > systemic 
circulation 
169 
Sulfonamides Nasal cavity > CSF, nasal cavity > systemic 
circulation 
170 , 171 
Dextran Nasal cavity > olfactory mucosa > CNS and 
also nasal cavity > systemic circulation 
172 
Dopamine Direct from nose to brain 173 
Cocaine Direct from nose to brain 141 
Dihydroergotamine Direct from nose to brain 174 
Insulin Direct from nose to brain 175 
Dopamine Nasal cavity > CSF 173 
Methotrexate Nasal cavity > CSF 176 
Nerve growth factor Direct from nose to brain via olfactory 
pathway 
177 
WGA – HRP (wheat germ 
agglutinin – horseradish 
peroxidase) 
Direct from nose to brain via olfactory nerve 
and bulb 
178 
Zolmitriptan Direct from nose to brain 179 
Leptin Direct from nose to brain 180 
Morphine Direct nose to brain via olfactory pathway 181 
Nimodipine Direct nose to brain via olfactory bulb and 
nasal cavity > CSF 
182 
Meptazinol hydrochloride Nasal cavity > CSF 183 
Estradiol Nasal cavity > CSF via olfactory neurons 184 
a Facilitated transport to brain based on functional evidence in humans. 
neuronal components comprising the odorant detectors. The olfactory region is 
mainly located on the nasal septum and partly on the superior and middle turbinates. 
It occupies only a small region in humans of about 10 cm 2 in the roof of the 
nasal cavity, as compared to around 150 cm 2 in dogs. The olfactory epithelium is a 

pseudostratifi ed epithelium comprising three types of cells: olfactory receptor cells, 
supporting cells, and basal cells. The receptor cells are elongated bipolar neurons 
located in the middle stratum of the epithelium, interspersed among the sustentacular 
cells; the microvilli cover the supporting cells, which extend from the mucosal 
surface to the basal membrane; while the basal cells are located in the basal surface 
of the neuroepithelium. These basal cells go on to differentiate, becoming new 
receptor cells [140, 185, 186] . The surface of the nasal cavity measures about 180 cm 2 
in humans as compared to about 10 cm 2 in rats, and the olfactory region is reported 
to constitute about 3% of the nasal cavity in humans and 50% in rats. Some other 
differences include the fact that in adult humans the volume of CSF is about 160 mL 
while it is only about 150 . L in rats; also, the CSF is replaced about three times daily 
in humans, whereas in rats it is replaced hourly. Hence, though there is suffi cient 
evidence regarding nose - to - brain delivery, especially in rats and in some cases in 
humans, the impact of these factors on the interpretation of the results between the 
two species could be signifi cant [155, 187] . 
5.6.9 NASAL DELIVERY OF VACCINES 
The discovery of vaccines for smallpox, cholera, and typhoid and the variety of vaccines 
now available have led to a signifi cant reduction in the mortality and morbidity 
due to many diseases, with smallpox being the fi rst to have been completely eradicated 
and poliomyelitis targeted to be the next. At present, the World Health Organization 
is working toward the complete elimination of poliomyelitis throughout 
the world [188, 189] . However, since Jenner discovered the vaccine for smallpox 
more than two centuries ago [190] , only some 50 vaccines have been approved for 
use, and few additional vaccines have been discovered. Most of those in current use 
are administered parenterally; they can induce only a systemic immune response, 
not mucosal immunity. Obviously the latter is very important in the prevention and 
treatment of infectious diseases, be they due to viral, bacterial, or parasitic pathogens 
that attack via the mucosal surfaces [190] . 
The criteria to be met in designing a vaccine formulation include the following: 
The vaccine should have the capacity to produce lifelong immunity, be able to act 
against the different strains and variants or the subtypes of the organisms, be effective 
in all age groups, be able to act quickly and also to induce immunity in the fetus 
when the mother is treated, be able to act effectively after a single treatment, and 
ideally be administered noninvasively. Finally, such a vaccine must be relatively 
inexpensive and remain active under a variety of conditions, especially not requiring 
the cold chain [191] . 
The following section addresses the need for needle - free vaccines with formulations 
based on safer adjuvant and delivery systems. 
5.6.9.1 Nasal Vaccines: Ideal Noninvasive Route 
When we talk about targeting the pathogens entering through the mucosal surfaces, 
the route that usually comes to mind is the oral route. However, this route has its 
drawbacks for several reasons, such as the fact that the antigen used is degraded 
along with the gastric contents; furthermore, there is also the diffi culty of reaching 
the antigen - presenting cells [192] . In comparison to the oral route, nasal vaccination 
NASAL DELIVERY OF VACCINES 633

634 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
has been shown to require a lower antigen dose, which is essential considering the 
cost of the recombinant agents that may be used as antigens. Nasal administration 
has been shown to induce immune responses in the respiratory and genital surfaces. 
When compared with the other mucosal routes such as vaginal or rectal administration, 
the nasal route is much more acceptable in terms of both accessibility and 
overall convenience. Hence the nasal route is increasingly being seen as ideal for 
the administration of vaccines. 
The nasal route has traditionally been used as an effective route in the treatment 
of respiratory infections, the rationale having been to target the infectious agents 
at their port of entry. Since most infectious disease pathogens enter at various 
mucosal sites, the nasal route has attracted increased attention as an alternative 
route for the delivery of vaccines. A further advantage offered by the nasal route 
in that it is capable of inducing both systemic and mucosal immunity as compared 
to the parenteral route, which brings about only systemic immunity [190, 193, 194] . 
Nasal immunization can result in distant as well as local mucosal immunity because 
of the mucosal immune system ’ s common properties. This means that it is possible, 
via nasal immunization, to attain adequate immunity at other mucosal sites such as 
the respiratory, intestinal, and genital; hence vaccines administered nasally will have 
an important role in the prevention of respiratory infections and, more importantly, 
in the treatment of sexually transmitted diseases [190 – 196] . 
The earliest vaccines were live attenuated, inactivated toxins, or inactivated 
toxoids. But with advances in molecular biotechnology, it has become possible to 
produce extremely pure vaccines. However, the main drawbacks of these vaccines 
are their poor immunogenicity, so that the use of adjuvants is often required to 
attain the necessary immunogenicity. A vaccine adjuvant is especially important in 
subunit vaccines, which is how most of the vaccines available today are supplied. 
Adjuvants can be best defi ned as particular agents that increase the immunity produced 
when they are coformulated and delivered with the vaccine antigen. The 
adjuvants are formulated in the vaccines so as to produce a longer lasting immunity. 
The more time the adjuvant takes to be eliminated from the system, the longer it 
is able to induce the required or intended lasting immune response. 
Vaccine adjuvants may be classifi ed operationally as delivery or immunostimulating 
adjuvants. Vaccine delivery adjuvants simply act as delivery agents, that is, they 
present the vaccine antigens to the antigen - presenting cells (APCs), whereas the 
immune - stimulating agents act by stimulating the APCs to elicit an appropriate 
immune response. This results from the stimulation of the Toll - like receptors, present 
on macrophages and dendritic cells, the activation of which shows increased antigen 
presentation and cytokine release. The combination of both types of adjuvants may 
lead to better formulations for nasal vaccines [196 – 199] . 
5.6.9.2 Immunity after Intranasal Immunization 
An understanding of the respiratory tract and the immune response following nasal 
vaccination is necessary to understand how the antigen used in the vaccine interacts 
with the surfaces of the human body and how the different adjuvants may interact, 
modify, and aid in generating an immune response. The nose is a component of the 
upper respiratory tract, which is composed of the mouth, nasopharynx, and larynx. 
The nasal passages have an extensive surface area which is richly vascularized. 

However, the nasal epithelium has little ability to break down drugs. The extensive 
mucosal surface of the nose has a lining of pseudostratifi ed epithelium as well as 
cilia and the goblet cells involved in the secretion of mucus. The lymphoid tissue 
primarily involved in the mucosal immune responses is the mucosal - associated 
lymphoid tissue (MALT). The different regions of the respiratory tract that play an 
infl uencing role in the immune system are as follows: 
• The epithelial surface, which comprises immunocompetent cells in the connective 
tissue 
• The lymphoid tissues linked to the respiratory tract, which are categorized into 
three parts: larynx - associated lymphoid tissue (LALT), nose - associated lymphoid 
tissue (NALT), and bronchus - associated lymphoid tissue 
• The lymph nodes that drain the respiratory tract 
The NALT, which is the organized lymphoid structure in the nasal passages and 
occurs in abundance in the nasal mucosa, plays a signifi cant role in the mucosal 
surface ’ s defense against invading pathogens. The NALT is equivalent to Waldeyer ’ s 
ring, which is made up of the adenoids or tonsils situated in the roof of the nasopharynx, 
bilateral lymphoid bands, the palatine tonsils, and the tonsil at the base of 
the tongue (the lingual tonsil). The NALT is the main target site for vaccine antigens 
in humans [193, 194, 197, 200, 201] . As the mucosal immune system develops, the 
NALT is involved in a variety of important functions in relation to the host defense 
mechanism, from being an impediment to drug absorption, serving as a guard 
against attacking pathogens and antigens, facilitating the uptake of antigens, eliciting 
the secretory antibody response, and inducing the immune response in the other 
distant mucosal surfaces due to the function of the common mucosal immune 
system. Owing to mucosal tolerance, it is also involved in preventing serious allergic 
responses to inhaled antigens [194, 201, 202] . 
On administration, the antigen interacts fi rst with the inductive tissue of the 
MALT, thus initiating a primary response. The IgA serum cells are found in the 
effector sites of the MALT, and local immunity results from the production of 
the secretory IgA (s - IgA) response. As mentioned earlier, the NALT and the tonsils 
form the main inductive sites in rodents and humans. These are composed of M cells 
involved in the uptake of antigen and its presentation to the underlying lymphoid 
tissues, the antigen being taken up by the M cells and the APCs, consisting of dendritic 
cells, macrophages, and B cells; all these cells together with the T cells produce 
the cellular and humoral immune responses [194] . 
Vaccination by the nasal route produces a mucosal protection using mucus, the 
epithelial surface, and both innate and acquired immune responses. The innate 
defense mechanism plays a very important role in that it infl uences the type of 
acquired immune mechanism, which mainly responds on the basis of “ immune 
memory. ” The ability to attain these responses is the main principle of attaining 
protection from infection. 
5.6.9.3 Need for Adjuvants 
As mentioned earlier, the subunit vaccines in particular require the use of adjuvants 
in order to initiate an immune response leading to protection. The subunit vaccines 
NASAL DELIVERY OF VACCINES 635

636 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
TABLE 5 Nasal Vaccination Delivery Systems Studied 
Delivery 
System/Adjuvant Antigen Employed Remarks Reference 
Chitosan Diphtheria toxin Increased local as well as 
systemic effects 
208 
Infl uenza virus Immune response 
comparable to that 
of intramuscular 
administration 
209 
Filamentous hemagglutinin 
and recombinant pertussis 
toxin (single or bivalent 
vaccine) 
Chitosan stimulates 
mucosal and systemic 
effects 
209 , 210 
IL - 12 Tetanus toxoid IL - 12 - induced IgA 
response 
211 
Infl uenza hemagglutinin and 
neuraminidase 
Induces protective 
immunity 
212 
CpG motifs Hepatitis B surface antigen 
(HBsAg) 
Potent enhancement of 
systemic and mucosal 
immune responses 
213 
Formalin - inactivated 
infl uenza virus 
Enhances the serum IgG 
and s - IgA responses 
214 
Liposomes Infl uenza subunit vaccine Induction of systemic IgG 
and s - IgA responses 
215 
Inactivated measles virus Stimulation of mucosal 
and systemic responses 
216 
Bovine serum albumin 
(BSA) 
— 217 
Streptococcus mutans — 218 
Infl uenza hemagglutinin and 
neuraminidase (when 
used in combination with 
heat - labile toxin (HLT) 
Good response in 
presence of HLT 
219 
Infl uenza, hepatitis B, 
tetanus toxoid 
— 220 
Yersinia pestis — 221 
ISCOMs 
(immune 
stimulating 
complex) 
Infl uenza subunit Protective immunity to 
challenge 
222 
Measles nucleoprotein Induces cytotoxic T - cell 
response 
223 
Echinococcus surface 
antigen 
— 224 
Respiratory syncytial virus 
(RSV) envelope antigen 
225 
Poly(lactic - co - 
glycolic acid 
(PLGA) 
microparticles 
HBsAg Strong systemic and 
mucosal immune 
responses 
226 
Cationic 
nanoparticles 
(SMBV) 
HBsAg and . - galactosidase Strong mucosal as well as 
systemic antibody and 
CTL responses 
227 

REFERENCES 637 
available today, which are administered intramuscularly or subcutaneously, involve 
alum as an adjuvant. The drive for initiating further research into vaccine adjuvants 
has been stimulated by many factors, among the chief of which is that the aluminum - 
based adjuvants currently available have failed in many candidate vaccines or have 
not achieved the necessary immunity or induced a cytotoxic T - cell response. Nasally 
formulated vaccines, mainly subunit vaccines, currently being purifi ed are less immunogenic 
and also cannot elicit the necessary T - cell response. As a result, research is 
now focusing on fi nding newer adjuvants for nasal DNA and subunit vaccines in 
order to attain specifi c immune responses as well as the necessary antibody subtype 
response. In addition to this, an adjuvant can help to reduce the dose of antigen 
required and also the number of doses needed to achieve mucosal immunity [203 – 
207] . Despite the extensive research going on in the fi eld of vaccine adjuvants, the 
only FDA - approved adjuvant for human use is alum. There are several other adjuvants, 
such as monophosphoryl lipid A (MPL), that have been approved in the 
European market; another, Corixa, is used as an adjuvant in Fendrix, the hepatitis 
B vaccine of GlaxoSmithKline Biologicals. The main hindrance to the approval of 
many adjuvants that reach clinical trials is their potential to elicit toxic side effects 
in clinical use. It has been reported that aluminum salts do induce some allergies in 
humans. As more purifi ed and target - oriented or specifi c vaccines obtained by 
recombinant technology are being launched, it becomes more diffi cult for vaccine 
antigens alone to induce the necessary immune responses, as these recombinant 
antigens or synthetic peptides cannot jump start an immune response. 
A number of adjuvants are awaiting approval for human use. The main impediment 
to the successful development of vaccine adjuvants is that their mechanism of 
action is not clearly understood. Table 5 offers a list of available nasal drug delivery 
systems and the various adjuvants that have been used in the development of nasal 
vaccines. 
REFERENCES 
1. Southall , J. , and Ellis , C. ( 2000 ), Developments in nasal drug delivery , Innovat. Pharm. 
Technol. , 110 – 115 . 
Delivery 
System/Adjuvant Antigen Employed Remarks Reference 
HBcAg HBsAg Stimulates strong Th1 
response 
228 
Cholera toxin Group B streptococci High levels of IgA in 
cervicovaginal 
secretions 
229 
Haemophilus infl uenzae Effective nasal 
vaccination 
230 
Infl uenza virus — 231 
Synthetic peptide of RSV Complete protection 232 
TABLE 5 Continued

638 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
2. Koch, M. (2003), The growing market for nasal drug delivery , Pharmatech , 1 – 3 . 
3. Ruppar , D. ( 2006 ), Intranasal delivery: Sniffi ng out new sources for growth , Drug Deliv. 
Technol. , 6 , 0 – 33 . 
4. Illum , L. , and Fisher , A. N. ( 1997 ), Intranasal delivery of peptides and proteins , in Adjei , 
A. L. and Gupta , P. K. , Eds., Inhalation Delivery of Therapeutic Peptides and Proteins , 
Marcel Dekker , New York , pp. 135 – 184 . 
5. Ugwoke , M. I. , Verbeke , N. , and Kinget , R. ( 2001 ), The biopharmaceutical aspects of 
nasal mucoahdesive drug delivery , J. Pharm. Pharmacol. , 53 , 3 – 21 . 
6. Illum , L. ( 1999 ), Bioadhesive formulations for nasal peptide delivery , in Mathiowitz , E. , 
Chickering , D. E. , and Lehr , C. - M. , Eds., Bioadhesive Drug Delivery Systems, Fundamentals, 
Novel Approaches and Development , Marcel Dekker , New York , pp. 507 – 539 . 
7. Washington , N. , Washington , C. , and Wilson , C. G. ( 2001 ), Physiological Pharmaceutics, 
Barriers to Drug Absorption , Taylor and Francis , London . 
8. Chien , Y. W. , Su , K. S. E. , and Chang , S. - F. ( 1989 ), Nasal Systemic Drug Delivery , Marcel 
Dekker , New York . 
9. Behl , C. R. , Pimplaskar , H. K. , Sileno , A. P. , deMeireles , J. , and Romeo , V. D. ( 1998 ), 
Effects of physicochemical properties and other factors on systemic nasal drug delivery , 
Adv. Drug Deliv. Rev. , 29 , 89 – 116 . 
10. Mygind , N. , and Dahl , R. ( 1998 ), Anatomy, physiology and function of the nasal cavities 
in health and disease , Adv. Drug Deliv. Rev. , 29 , 3 – 12 . 
11. Olson , P. , and Bende , M. ( 1985 ), Infl uence of environmental temperature on human 
nasal mucosa , Ann. Otol. Rhinol. Lryngol. , 94 , 153 – 157 . 
12. Paulsson , B. , Bende , M. , and Ohlin , P. ( 1985 ), Nasal mucosal blood fl ow at rest and during 
exercise , Acta Otolaryngol. (Stockh.) , 99 , 140 – 144 . 
13. Hadley , W. M. , and Dahl , A. R. ( 1983 ), Cytochrome P450 - dependent mono - oxygenase 
in nasal membrane of six species , Drug Metab. Dispos. , 11 , 275 – 279 . 
14. Longo , V. , Pacifi ci, G. M. , Panattoni, G. , Ursino , F. , and Gervasi, P. G. (1989), Metabolism 
of diethylnitrosamine by microsomes of human respiratory nasal mucosa and liver , 
Biochem. Pharmacol. , 38 , 1867 – 1869 . 
15. Brittebo , E. G. , and Rafter , J. J. ( 1984 ), Steroid metabolism by rat nasal mucosa: Studies 
on progesterone and testosterone , Steroid. Biochem. , 20 , 1147 . 
16. Smith , E. L. , Hill , R. L. , and Borman , A. ( 1958 ), Activity of insulin degraded by leucine 
aminopeptidase , Biochem. Biophys. Acta. , 29 , 207 – 214 . 
17. Marttin , E. , Schipper , N. G. M. , Verhoef , J. C. , and Merkus , F. W. H. M. ( 1998 ), Nasal 
mucosal clearance as a factor in nasal drug delivery , Adv. Drug Deliv. Rev. , 29 , 13 – 38 . 
18. Nagai , T. , Nishimoto , Y. , Nambu , N. , Suzuki , Y. , and Sekine , K. ( 1984 ), Powder dosage 
form of insulin for nasal administration , J. Controlled Release , 1 , 15 – 22 . 
19. Nagai , T. , and Machida , Y. ( 1990 ), Bioadhesive dosage forms for nasal administration , 
in Lenaerts , V. , and Gurny , R. , Eds., Bioadhesive Drug Delivery Systems , CRC Press , 
Boca Raton, FL , pp. 169 – 178 . 
20. Pritchard , K. , Lansely , A. B. , Martin , G. P. , Helliwell , M. , Marriott , C. , and Benedetti , 
L. M. ( 1996 ), Evaluation of bioadhesive properties of hyaluronan derivatives, detachment 
weight and mucociliary transport rate studies , Int. J. Pharm. , 129 , 137 – 145 . 
21. Marttin , E. , Verhoef , J. C. , Romeijn , S. G. , Zwart , P. , and Merkus , F. W. H. M. ( 1996 ), 
Acute histopathological effects of bezalkonium chloride and absorption enhancers on 
rat nasal epithelium in vivo , Int. J. Pharm. , 141 , 151 – 160 . 
22. Ennis , R. D. , Borden , L. , and Lee , W. A. ( 1990 ), The effects of permeation enhancers on 
the surface morphology of rat nasal mucosa: A scanning electron microscopy study , 
Pharm. Res. , 7 , 468 – 475 . 

REFERENCES 639 
23. Hardy , J. G. , Lee , S. W. , and Wilson , C. G. ( 1985 ), Intranasal drug delivery by spray and 
drops , J. Pharm. Pharmacol. , 37 , 294 – 297 . 
24. Proctor , D. F. ( 1985 ), Nasal Physiology in intranasal drug administrations , in Chien , 
Y. W. , Ed., Transnasal Systemic Medications , Elsevier , Amsterdam , pp. 101 – 106 . 
25. Greiff , L. , Venge , P. , Andersson , M. , Enander , I. , Linden , M. , Myint , S. , and Persson , 
C. G. ( 2002 ), Effects of rhinovirus - induced common colds on granulocyte activity in 
allergic rhinitis , J. Infect. , 45 , 227 – 232 . 
26. Larsen , C. , Niebuhr , J. M. , Tommerup , B. , Mygind , N. , Dagrosa , E. E. , Grigoleit , H. G. , 
and Malerczyk , V. ( 1987 ), Infl uence of experimental rhinitis on the gonadotropin 
response to intranasal administration of buserelin , Eur. J. Clin. Pharmacol. , 33 , 
155 – 159 . 
27. Kublik , H. , and Vidgren , M. T. ( 1998 ), Nasal delivery systems and their effect on deposition 
and absorption , Adv. Drug Deliv. Rev. , 29 , 157 – 177 . 
28. Hussain , A. A. , Bawarshi - Nassar , R. , and Huang , C. H. ( 1985 ), Physicochemical considerations 
in intranasal drug administration , in Chien , Y. W. , Ed., Transnasal Systemic 
Medication , Elsevier , Amsterdam , pp. 121 – 138 . 
29. Corbo , D. C. , Huang , Y. C. , and Chien , Y. W. ( 1989 ), Nasal delivery of progestational 
steroids in ovarectomized rabbits. II. Effect of penetrant hydrophilicity , Int. J. Pharm. , 
50 , 253 – 260 . 
30. Kimura , R. , Miwa , M. , Kato , Y. , Sato , M. , and Yamada , S. ( 1991 ), Relationship between 
nasal absorption and physicochemical properties of quaternary ammonium compounds , 
Arch. Int. Pharmacodyn. Ther. , 310 , 13 – 21 . 
31. Tonndorf , J. , Chinn , H. I. , and Lett , J. E. ( 1953 ), Absorption from nasal mucous membrane: 
Systemic effect of hyoscine following intranasal administration , Ann. Otol. Rhinol. 
Laryngol. , 62 , 630 – 634 . 
32. Rogerson , A. , and Parr , G. D. ( 1990 ), Nasal drug delivery , in Florence , A. T. , and Salole , 
E. G. , Eds., Topics in Pharmacy, Routes of Drug Administration , Northants , England , 
pp. 1 – 29 . 
33. McMartin , C. , Hutchinson , E. F. , Hyde , R. , and Peters , G. E. ( 1987 ), Analysis of structural 
requirements for the absorption of drugs and macromolecules from the nasal cavity , 
J. Pharm. Sci. , 76 , 535 – 540 . 
34. Donovan , M. D. , Flynn , G. , and Amidon , G. ( 1990 ), Absorption of polyglycols 600 
through 2000: The molecular weight dependence of gastrointestinal and nasal absorption 
, Pharm. Res. , 7 , 863 – 868 . 
35. Fisher , A. , Brown , K. , Davis , S. , Parr , G. , and Smith, D. A. (1987), The effect of molecular 
size on the nasal absorption of water - soluble compounds in the albino rat , J. Pharm. 
Pharmacol. , 39 , 357 – 362 . 
36. Fisher , A. , Illum , L. , Davis , S. , and Schacht , E. ( 1992 ), Di - iodo - l - tyrosine labeled dextrans 
as molecular size markers for nasal absorption in the rat , J. Pharm. Pharmacol. , 44 , 
550 – 554 . 
37. Maitani , Y. , Machida , Y. , and Nagai , T. ( 1989 ), Infl uence of molecular weight and charge 
on nasal absorption of water soluble compounds in the albino rat , Int. J. Pharm. , 40 , 
23 – 27 . 
38. D ’ Souza , R. , Mutalik , S. , Venkatesh , M. , Vidyasagar , S. , and Udupa , N. ( 2005 ), Nasal 
insulin gel as an alternative to parenteral insulin: Formulation, preclinical and clinical 
studies , AAPS PharmSciTech. , 6 , E184 – E189 . 
39. Bommer , R. , Kern , J. , Hennes , K. , and Zwisler , W. ( 2005 ), Preservative - free nasal drug 
delivery systems , Drug Deliv. Technol. , 5 . 

640 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
40. Giroux , M. ( 2005 ), Controlled Particle Dispersion ™ : Effective nasal delivery from a 
versatile, fl exible technology platform , in Nasal Drug Delivery: Rapid Onset via Convenient 
Route , ONdrugdelivery , pp. 13 – 15 . 
41. Hwang , P. , Woo , E. , and Fong , K. , Intranasal deposition of nebulized saline: A radionuclide 
distribution study, paper presented at the 50th annual meeting of the American 
Rhinologic Society, New York, NY, Sept. 2004 . 
42. Giroux , M. , Hwang , P. , and Prasad , A. ( 2005 ), Controlled Particle Dispersion ™ : Applying 
vortical fl ow to optimize nasal drug deposition , Drug Deliv. Technol. , 5 , 44 – 49 . 
43. Djupesland , P. G. , and Watts , J. ( 2005 ), Breath - actuated bidirectional delivery sets the 
nasal market on a new course, ONdrugdelivery , pp. 20 – 23 . 
44. Djupesland , P. G. , Skretting , A. , Winderen , M. , and Holand , T. ( 2004 ), Bi - directional nasal 
delivery of aerosols can prevent lung deposition , J. Aerosol. Med. , 17 , 249 – 259 . 
45. Keldmann , T. ( 2005 ), Advanced simplifi cation of nasal delivery technology: Anatomy + 
innovative device = added value opportunity, ONdrugdelivery , pp. 4 – 7 . 
46. Hirai , S. , Yashiki , T. , and Matsuzawa , T. , Nasal absorption of drugs. Effect of surfactants 
on the nasal absorption of insulin in rats, paper presented at the 98th annual meeting 
of the Pharmaceutical Society of Japan, Apr. 1978 . 
47. Hussain , A. , Hirai , S. , and Bawarshi , R. ( 1980 ), Nasal absorption of propranolol from 
different dosage forms by rats and dogs , J. Pharm. Sci. , 69 , 1411 – 1413 . 
48. Pillion , D. J. , Ahsan , F. , Arnold , J. J. , Balusubramanian , B. M. , Piraner , O. , and Meezan , 
E. ( 2002 ), Synthetic long - chain alkyl maltosides and alkyl sucrose esters as enhancers 
of nasal insulin absorption , J. Pharm. Sci. , 91 , 1456 – 1462 . 
49. Ahsan , F. , Arnold , J. J. , Meezan , E. , and Pillion , D. J. ( 2001 ), Enhanced bioavailability of 
calcitonin formulated with alkylglycosides following nasal and ocular administration in 
rats , Pharm. Res. , 18 , 1742 – 1746 . 
50. Merkus , F. W. H. M. , Verhoef , J. C. , Marttin , E. , Romeijn , S. G. , van der Kuy , P. H. M. , 
Hermens , W. A. J. J. , and Schipper , N. G. M. ( 1999 ), Cyclodextrins in nasal drug delivery , 
Adv. Drug Deliv. Rev. , 6 , 41 – 57 . 
51. Gizurarson , S. ( 1990 ), Animal models for intranasal drug delivery , Acta. Pharm. Nord. , 
2 , 105 – 122 . 
52. Behl , C. R. , Pimplaskar , H. K. , Sileno , A. P. , Xia , W. J. , Gries , W. J. , de Meireles , J. C. , and 
Romeo , V. D. ( 1998 ), Optimization of systemic nasal drug delivery with pharmaceutical 
ecipients , Adv. Drug Deliv. Rev. , 29 , 117 – 133 . 
53. Illum , L. ( 2002 ), Nasal drug delivery: New developments and strategies , Drug Discovery 
Today , 7 , 1184 – 1189 . 
54. Hinchcliffe , M. , and Illum , L. ( 1999 ), Intranasal insulin delivery and therapy , Adv. Drug 
Deliv. Rev. , 35 , 199 – 234 . 
55. Illum , L. , and Fisher , A. N. ( 1997 ), Intranasal delivery of peptides and proteins , in Adjei , 
A. L. , and Gupta , P. K. , Eds., Inhalation Delivery of Therapeutic Peptides and Proteins , 
Marcel Dekker , New York , p. 135 . 
56. Lansley , A. B. , and Martin , G. P. ( 2001 ), Nasal drug delivery , in Hillery , A. M. , Lloyd , 
A. W. , and Swarbrick , J. , Eds., Drug Delivery and Targeting , 2nd ed. , Taylor & Francis , 
New York , p. 237 . 
57. Triplitt , C. L. , Reasner , C. A. , and Isley , W. ( 2005 ), Diabetes mellitus , in Dipiro , J. T. , 
Talbert , R. L. , Yee , G. C. , Matzke , G. R. , Wells , B. G. , and Posey , L. M. Eds., Pharmacotherapy: 
A Pathophysiological Approach , 6th ed. , McGraw - Hill , New York , p. 1333 . 
58. Bliss , M. (1933), The history of insulin, Diabetes Care , 16 , 4 – 7 . 
59. Woodyatt, R. T. (1922), The clinical use of insulin , J. Metab. Res. , 2 , 793 – 801 . 

REFERENCES 641 
60. Collens , W. S. , and Goldzeiher , M. A. ( 1932 ), Absorption of insulin by nasal mucous 
membranes , Proc. Soc. Exp. Biol. Med. , 29 , 756 . 
61. Major , R. H. (1935), The intranasal application of insulin, J. Lab. Clin. Med. , 21 , 278 . 
62. Major , R. H. (1936), The intranasal application of insulin, Am. J. Med. Sci. , 192 , 257 . 
63. Gordon , G. S. , Moses , A. C. , Silver , R. D. , Flier , J. S. , and Carey , M. C. ( 1985 ), Nasal 
absorption of insulin: Enhancement by hydrophobic bile salts , Proc. Natl. Acad. Sci. USA , 
82 , 7419 – 7423 . 
64. Morimoto , K. , Morisaka , K. , and Kamada , A. ( 1985 ), Enhancement of nasal absorption 
of insulin and calcitonin using polyacrylic acid gel , J. Pharm. Pharmacol. , 37 , 134 – 136 . 
65. Pillion , D. J. , Atchison , J. A. , Gargiulo , C. , Wang , R. X. , Wang , P. , and Meezan , E. ( 1994 ), 
Insulin delivery in nosedrops: New formulations containing alkylglycosides , Endocrinology 
, 135 , 2386 – 2391 . 
66. Pillion , D. J. , Ahsan , F. , Arnold , J. J. , Balusubramanian , B. M. , Piraner , O. , and Meezan , 
E. ( 2002 ), Synthetic long - chain alkyl maltosides and alkyl sucrose esters as enhancers 
of nasal insulin absorption , J. Pharm. Sci. , 91 , 1456 – 1462 . 
67. Ahsan , F. , Arnold , J. J. , Meezan , E. , and Pillion , D. J. ( 2003 ), Sucrose cocoate, a component 
of cosmetic preparations, enhances nasal and ocular peptide absorption , Int. J. 
Pharm. , 251 , 195 – 203 . 
68. Illum , L. , Jabbal - Gill , I. , Hinchcliffe , M. , Fisher , A. N. , and Davis , S. S. ( 2001 ), Chitosan 
as a novel nasal delivery system for vaccines , Adv. Drug Deliv. Rev. , 51 , 81 – 96 . 
69. Illum , L. ( 1992 ), Nasal delivery of peptides, factors affecting nasal absorption , in Topics 
in Pharmaceutical Sciences , Medpharm Scientifi c , Stuttgart , p. 71 . 
70. Illum , L. , Farraj , N. F. , and Davis , S. S. ( 1994 ), Chitosan as a novel nasal delivery system 
for peptide drugs , Pharm. Res. , 11 , 1186 – 1189 . 
71. Fern a ndez - Urrusuno , R. , Calvo , P. , Remu n a n - L o pez , C. , Vila - Jato , J. L. , and Alonso , 
M. J. ( 1999 ), Enhancement of nasal absorption of insulin using chitosan nanoparticles , 
Pharm. Res. , 16 , 1576 – 1581 . 
72. Dyer , A. M. , Hinchcliffe , M. , Watts , P. , Castile , J. , Jabbal - Gill , I. , Nankervis , R. , Smith , A. , 
and Illum , L. ( 2002 ), Nasal delivery of insulin using novel chitosan based formulations: 
A comparative study in two animal models between simple chitosan formulations and 
chitosan nanoparticles , Pharm. Res. , 19 , 998 – 1008 . 
73. Yu , S. , Zhao , Y. , Wu , F. , Zhang , X. , L u , W. , Zhang , H. , and Zhang , Q. ( 2004 ), Nasal 
insulin delivery in the chitosan solution: In vitro and in vivo studies , Int. J. Pharm. , 281 , 
11 – 23 . 
74. Varshosaz , J. , Sadrai , H. , and Alingari, R. (2004), Nasal delivery of insulin using chitosan 
microspheres , J. Microencapsul. , 21 , 761 – 774 . 
75. Varshosaz , J. , Sadrai , H. , and Heidari , A. ( 2006 ), Nasal delivery of insulin using bioadhesive 
chitosan gels , Drug Deliv. , 13 , 31 – 38 . 
76. Pontiroli , A. E. , Alberetto , M. , and Pozza , G. ( 1985 ), Intranasal calcitonin and plasma 
calcium concentrations in normal subjects , Br. Med. J. , 290 , 1390 – 1391 . 
77. Morimoto , K. , Morisaka , K. , and Kamada , A. ( 1985 ), Enhancement of nasal absorption 
of insulin and calcitonin using polyacrylic acid gel , J. Pharm. Pharmacol. , 37 , 134 – 
136 . 
78. Morimoto , K. , Akatsuchi , H. , Aikawa , R. , Morishita , M. , and Morisaka , K. ( 1984 ), 
Enhanced rectal absorption of [Asu1, 7] - eel calcitonin in rats using polyacrylic acid 
aqueous gel base , J. Pharm Sci. , 73 , 1366 – 1368 . 
79. Lee , K. C. , Tak , K. K. , Park , M. O. , Lee , J. T. , Woo , B. H. , Yoo , S. D. , Lee , H. S. , and DeLuca , 
P. P. ( 1999 ), Preparation and characterization of polyethylene - glycol - modifi ed salmon 
calcitonins , Pharm. Dev. Tech. , 4 , 269 – 275 . 

642 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
80. Miron , T. , and Wilcheck , M. ( 1993 ), A simplifi ed method for the preparation of succinimidyl 
carbonate polyethylene glycol for coupling to proteins , Biconj. Chem. , 4 , 
568 – 569 . 
81. Shin , B. S. , Jung , J. H. , Lee , K. C. , and Yoo , S. D. ( 2004 ), Nasal absorption and pharmacokinetic 
disposition of salmon calcitonin modifi ed with low molecular weight polyethylene 
glycol , Chem. Pharm. Bull. , 52 , 957 – 960 . 
82. Na , D. H. , Youn , Y. S. , Park , E. J. , Lee , J. M. , Cho , O. R. , Lee , K. R. , Lee , S. D. , Yoo , S. D. , 
Deluca , P. P. , and Lee , K. C. ( 2004 ), Stability of PEGylated salmon calcitonin in nasal 
mucosa , J. Pharm. Sci. , 93 , 256 – 261 . 
83. Arnold , J. J. , Ahsan , F. , Meezan , E. , and Pillion , D. J. ( 2002 ), Nasal administration of low 
molecular weight heparin , J. Pharm. Sci. , 91 , 1707 – 1714 . 
84. Yang , T. , Mustafa , F. , and Ahsan , F. ( 2004 ), Alkanoylsucroses in nasal delivery of low 
molecular weight heparins: In - vivo absorption and reversibility studies in rats , J. Pharm. 
Pharmacol. , 56 , 53 – 60 . 
85. Mustafa , F. , Yang , T. , Khan , M. A. , and Ahsan , F. ( 2004 ), Chain length - dependent effects 
of alkylmaltosides on nasal absorption of enoxaparin , J. Pharm. Sci. , 93 , 675 – 683 . 
86. Yang , T. , Hussain , A. , Paulson , J. , Abbruscato , T. J. , and Ahsan , F. ( 2004 ), Cyclodextrins 
in nasal delivery of low - molecular - weight heparins: In vivo and in vitro studies , Pharm. 
Res. , 21 , 1127 – 1136 . 
87. Yang , T. , Hussain , A. , Bai , S. , Khalil , I. A. , Harashima , H. , and Ahsan , F. ( 2006 ), Positively 
charged polyethylenimines enhance nasal absorption of the negatively charged drug, 
low molecular weight heparin , J. Controlled Release , 115 , 289 – 297 . 
88. Mason , G. A. , Garbutt , J. C. , and Prange Jr ., A. J. ( 2000 ), Thyrotropin releasing 
hormone: Focus On neurobiology, available: http://www.acnp.org/g4/GN401000048/ 
CH048.html . 
89. Yamamoto , M. , and Shimizu , M. ( 1987 ), Effects of new TRH analogue, YM - 14673 on 
the central nervous system , Naunyn. Schmiedebergs. Arch. Pharmacol. , 336 , 561 – 565 . 
90. Kagatani , S. , Inaba , N. , Fukui , M. , and Sonobe , T. ( 1998 ), Nasal absorption kinetic behavior 
of Azetirelin and its enhancement by acylcarnitines in rats , Pharm. Res. , 15 , 77 – 81 . 
91. Mitsuma , T. , and Nogimori , T. ( 1984 ), Changes in plasma thyrotrophin - releasing hormone, 
thyrotrophin, prolactin and thyroid hormone levels after intravenous, intranasal or rectal 
administration of synthetic thyrotrophin - releasing hormone in man , Acta Endocrinol. , 
107 , 207 – 212 . 
92. Laursen , T. , Grandjean , T. , Jorgensen , J. O. , and Christiansen , J. S. ( 1996 ), Bioavailability 
and bioactivity of three different doses of nasal growth hormone (GH) administered to 
GH - defi cient patients: Comparison with intravenous and subcutaneous administration , 
Eur. J. Endocrinol. , 135 , 309 – 315 . 
93. Leitner , V. M. , Guggi , D. , Krauland A. H. , and Bernkop - Schnurch , A. ( 2004 ), Nasal 
delivery of human growth hormone: In vitro and in vivo evaluation of a thiomer/glutathione 
microparticulate delivery system , J. Controlled. Release , 100 , 87 – 95 . 
94. Hedin , L. , Olsson , B. , Diczfalusy , M. , Flyg , C. , Petersson , A. S. , Rosberg , S. , and Albertsson 
- Wikland , K. ( 1993 ), Intranasal administration of human growth hormone (hGH) in 
combination with a membrane permeation enhancer in patients with GH defi ciency: A 
pharmacokinetic study , J. Clin. Endocrinol. Metab. , 76 , 962 – 967 . 
95. Albertsson - Wikland , K. , and Rosberg , S. ( 1988 ), Analyses of 24 - hour growth hormone 
(GH) profi les in children , J. Clin. Endocrinol. Metab. , 67 , 493 – 500 . 
96. Clark , R. G. , Jansson , J. O. , Isaksson , O. , and Robinson , I. C. A. F. ( 1985 ), Intravenous 
growth hormone: Growth responses to patterned infusions in hypophysectomized rats , 
J. Endocrinol. , 104 , 53 – 61 . 

REFERENCES 643 
97. Eden , S. ( 1979 ), Age and sex related differences in episodic growth hormone secretion 
in the rat , Endocrinology , 105 , 555 – 560 . 
98. Vermehren , C. , Hansen , H. S. , and Thomsen , M. K. ( 1996 ), Time dependent effects of 
two absorption enhancers on the nasal absorption of growth hormone in rabbits , Int. J. 
Pharm. , 128 , 239 – 250 . 
99. Clausen , A. E. , and Schn u rch - Bernkop , A. ( 2000 ), In vitro evaluation of permeation 
enhancing effect of thiolated polycarbophil , J. Pharm. Sci. , 89 , 1253 – 1261 . 
100. Schn u rch - Bernkop , A. , and Steininger , S. ( 2000 ), Synthesis and characterization of 
mucoadhesive thiolated polymers , Int. J. Pharm. , 194 , 239 – 247 . 
101. Schn u rch - Bernkop , A. , Zarti , H. , and Walker , G. F. ( 2001 ), Thiolation of polycarbophil 
enhances its inhibition of soluble and intestinal brush border membrane bound aminopeptidase 
N , J. Pharm. Sci. , 90 , 1907 – 1914 . 
102. Clausen , A. E. , Kast , C. E. , and Schn u rch - Bernkop , A. ( 2002 ), The role of glutathione in 
the permeation enhancing effect of thiolated polymers , Pharm. Res. , 19 , 602 – 608 . 
103. Schn u rch - Bernkop , A. , Guggi , D. , and Pinter , Y. ( 2004 ), Thiolated chitosans: Development 
and in vitro evaluation of a mucoadhesive, permeation enhancing oral drug delivery 
system , J. Controlled Release , 94 , 177 – 186 . 
104. Illum, L. (1996), Animal models for nasal delivery , J. Drug Target. , 3 , 717 – 724 . 
105. Illum , L. , Watts , P. , Fisher , A. N. , Hinchcliffe , M. , Norbury , H. , Jabbal - Gill , I. , Nankervis , 
R. , and Davis , S. S. ( 2002 ), Intranasal delivery of morphine , J. Pharmcol. Exp. Ther. , 301 , 
391 – 400 . 
106. Lindhardt , K. , O lafsson , D. R. , Gizurarson , S. , and Bechgaard , E. ( 2002 ), Intranasal bioavailability 
of diazepam in sheep correlated to rabbit and man , Int. J. Pharm. , 231 , 
67 – 72 . 
107. Kyrkou , M. , Harbord , M. , Kyrkou , N. , Kay , D. , and Coulthard , K. ( 2006 ), Community use 
of intranasal midazolam for managing prolonged seizures , J. Intellect. Dev. Disabil. , 31 , 
131 – 138 . 
108. Lui , C. Y. , Amidon , G. L. , and Goldberg , A. ( 1991 ), Intranasal absorption of fl urazepam, 
midazolam and triazolam in dogs , J. Pharm. Sci. , 80 , 1125 – 1129 . 
109. Olivier , J. C. , Djilani , M. , Fahmy , S. , and Couet , W. ( 2001 ), In situ nasal absorption of 
midazolam in rats , Int. J. Pharm. , 213 , 187 – 192 . 
110. Burstein , A. H. , Modica , R. , Hatton , M. , Forest , A. , and Gengo , F. M. ( 1997 ), Pharmcokinetics 
and pharmacodynamics of midazolam after intranasal administration , J. Clin. 
Pharmacol. , 37 , 711 – 718 . 
111. Knoester , P. D. , Jonker , D. M. , van der Hoeven , R. T. M. , Vermeij , A. C. , Edelbroek , 
P. M. , Brekelmans , G. J. , and de Haan , G. J. ( 2002 ), Pharmacokinetics and pharmacodynamics 
of midazolam administered as a concentrated intranasal spray. A study in healthy 
volunteers , Br. J. Clin. Pharmcol. , 53 , 501 – 507 . 
112. Allonen , H. , Ziegler , G. , and Klotz , U. ( 1981 ), Midazolam kinetics , Clin. Pharmacol. Ther. , 
30 , 653 – 661 . 
113. Lindhardt , K. , Gizurarson , S. , Stefansson , S. , Olafsson , D. R. , and Bechgaard , E. ( 2001 ), 
Electroencephalographic effects and blood levels after administration of diazepam to 
humans , Br. J. Clin. Pharmcol. , 52 , 1 – 12 . 
114. Eriksen , J. , Jensen , N. H. , Kamp - Jensen , M. , Bjarno , H. , Friis , P. , and Brewster , D. ( 1989 ), 
The systemic availability of buprenorphine administered by nasal spray, J. Pharm. Pharmacol., 
41, 803 – 805 . 
115. Shyu , W. C. , Mayol , R. F. , Pfeffer , M. , Pittman , K. A. , and Barbhiya , R. H. ( 1993 ), Biopharmaceutical 
evaluation of transnasal, sublingual and buccal disk dosage forms of 
butorphanol , Biopharm. Drug Dispos. , 14 , 371 – 379 . 

644 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
116. Kaneo , Y. ( 1983 ), Absorption from the nasal mucous membrane: I. Nasal absorption of 
hydralazine in rats , Acta. Pharm. Suec. , 20 , 379 – 388 . 
117. Hirai , S. , Yashiki , T. , Matsuzawa , T. , and Mima , H. ( 1981 ), Absorption of drugs from the 
nasal mucosa of rats , Int. J. Pharm. , 7 , 317 . 
118. Landau , A. J. , Eberhardt , R. T. , and Frishman , W. H. ( 1994 ), Intranasal delivery of cardiovascular 
agents: An innovative approach to cardiovascular pharmacotherapy , Am. 
Heart J. , 127 , 1594 – 1599 . 
119. Hill , A. B. , Bowley , C. J. , Nahrwold , M. L. , Knight , P. R. , Krish , M. M. , and Denlinger , 
J. K. ( 1981 ), Intranasal administration of nitroglycerin , Anethesiology , 54 , 346 – 348 . 
120. Krishnamoorthy , R. , and Mitra , A. K. ( 1998 ), Prodrugs for nasal drug delivery , Adv. Drug 
Deliv. Rev. , 29 , 135 – 146 . 
121. Fossoulaki , A. , and Kaniaris , P. ( 1983 ), Intranasal administration of nitroglycerin attenuates 
the pressor response to laryngoscopy and intubation of the trachea , Br. J. Anaesth. , 
55 , 49 – 51 . 
122. Grover , V. K. , Sharma , S. , Mahajan , R. P. , and Singh , H. ( 1987 ), Intranasal nitroglycerin 
attenuates pressor response to tracheal intubation in beta - blocker treated hypertensive 
patients , Anesthesia , 42 , 884 – 887 . 
123. Hussain , A. , Hirai , S. , and Bawarshi , R. ( 1979 ), Nasal absorption of propranolol in rats , 
J. Pharm. Sci. , 68 , 1196 . 
124. Hussain , A. A. , Foster , T. , Hirai , S. , Kashihara , T. , Batenhorst , R. , and Jones , M. ( 1980 ), 
Nasal absorption of propranolol in humans , J. Pharm. Sci. , 69 , 1240 . 
125. Landau , A. J. , Frishman , W. H. , Alturk , N. , Adjei - Poku , M. , Fornasier - Bongo , M. , and 
Furia , S. ( 1993 ), Immediate beta - adrenergic blockade and improved exercise tolerance 
with intranasal propranolol in patients with angina pectoris , Am. J. Cardiol. , 72 , 995 – 
998 . 
126. Duchateau , G. S. M. J. E. , Zuidema , J. , Albers , W. M. , and Merkus , F. W. H. M. ( 1986 ), 
Nasal absorption of alprenolol and metoprolol , Int. J. Pharm. , 34 , 131 – 136 . 
127. Duchateau , G. S. M. J. E. , Zuidema , J. , and Merkus , F. W. H. M. ( 1986 ), Bioavailability of 
propranolol after oral, sublingual and intranasal administration , Pharm. Res. , 3 , 
108 – 111 . 
128. Wattanakumtornkul , S. , Pinto , A. B. , and Williams , D. B. ( 2003 ), Intranasal hormone 
replacement therapy , Menopause. , 10 , 88 – 98 . 
129. Hermens , W. A. J. J. , Belder , C. W. , Merkus , J. M. , Hooymans , P. M. , Verhoef , J. , and 
Merkus , F. W. ( 1991 ), Intranasal estradiol administration to oophorectomized women , 
Eur. J. Obstet. Gynecol. Reprod. Biol. , 40 , 35 – 41 . 
130. Hermens , W. A. , Belder , C. W. , Merkus , J. M. , Hooymans , P. M. , Verhoef , J. , and Merkus , 
F. W. ( 1992 ), Intranasal administration of estradiol in combination with progesterone to 
oophorectomized women , Eur. J. Obstet. Gynecol. Reprod. Biol. , 43 , 65 – 70 . 
131. Garnero , P. , Tsouderos , Y. , Marton , I. , Pelissier , C. , Varin , C. , and Delmas , P. D. 
( 1999 ), Effects of intranasal 17 . - estradiol on bone marrow turnover and serum insulin - 
like growth factor I in postmenopausal women , J. Clin. Endocrinol. Metab. , 84 , 2390 – 
2397 . 
132. Lievertz , R. W. ( 1987 ), Pharmacology and pharmacokinetics of estrogens , Am. J. Obstet. 
Gynecol. , 156 , 1289 – 1293 . 
133. Studd , J. , Pornel , B. , Marton , I. , Bringer , J. , Varin , C. , Tsouderos , Y. , and Christansen , C. 
(1999), Effi cacy and acceptability of intranasal 17 . - estradiol on menopausal symptoms: 
A randomized dose - response study , Lancet. , 353 , 1574 – 1578 . 
134. Devissaguet , J. P. , Brion , N. , Lhote , O. , and Deloffre , P. ( 1999 ), Pulsed estrogen therapy: 
Pharmacokinetics of intranasal 17 . - estradiol (S21400) in postmenopausal women and 

REFERENCES 645 
comparison with oral and transdermal formulations , Eur. J. Drug Metab. Pharmacokinet. , 
24 , 265 – 271 . 
135. Wilkund , I. , Karlberg , J. , and Mattson , L. A. ( 1993 ), Quality of life of postmenopausal 
women on a regimen of transdermal estradiol therapy: A double blind placebo - 
controlled study , Am. J. Obstet. Gynecol. , 168 , 824 – 830 . 
136. Wilkund , I. , Holst , J. , Karlberg , J. , et al. ( 1992 ), A new methodological approach to the 
evaluation of quality of life in postmenopausal women , Maturitas , 14 , 211 – 224 . 
137. Kupperman , H. S. , Blatt , M. H. G. , Weisbader , H. , and Filler , W. ( 1953 ), Comparative 
clinical evaluation of estrogenic preparations by the menopausal and amenorrheal 
indices , Endocrinology , 13 , 688 – 703 . 
138. Hussain , A. A. , Al - bayati , A. A. , Dakkuri , A. , Okochi , K. , and Hussain , M. A. ( 2002 ), 
Testosterone 17 . - N,N - dimethylglycinate hydrochloride: A prodrug with a potential for 
nasal delivery of testosterone , J. Pharm. Sci. , 91 , 785 – 789 . 
139. Hussain , A. A. , Kimura , R. , and Huang , C. H. ( 1984 ), Nasal absorption of testosterone 
in rats , J. Pharm. Sci. , 73 , 1300 – 1301 . 
140. Illum , L. ( 2000 ), Transport of drugs from the nasal cavity to the central nervous system , 
Eur. J. Pharm. Sci. , 11 , 1 – 18 . 
141. Chow , H. S. , Chen , Z. , and Matsuura , G. T. ( 1999 ), Direct transport of cocaine from the 
nasal cavity to the brain following intranasal cocaine administration in rats , J. Pharm. 
Sci. , 88 , 754 – 758 . 
142. Landsteiner , K. , and Levaditi , C. ( 1910 ), Etude experimentale de la poliomyelite aigue 
(Maladiede Heine Medin) , Ann. Inst. Pasteur. , 24 , 833 – 878 . 
143. Leiner , C. , and von Weisner , R. ( 1910 ), Experimentale untersuchungen uber poliomyelitis 
acuta , Wien. Med. Wchnschr. , 60 , 2482 . 
144. Flexner , S. ( 1912 ), The mode of action of infection epidemic poliomyelitis , JAMA , 59 , 
1371 . 
145. Flexner , S. , and Clark , P. F. ( 1912 ), A note on the mode of infection in epidemic poliomyelitis 
, Proc. Soc. Exp. Biol. Med. , 10 , 1 . 
146. Clark , P. F. , Fraser , F. R. , and Amoss , H. L. ( 1917 ), The relation to the blood of the virus 
of epidemic poliomyelitis , J. Exp. Med. , 19 , 223 – 233 . 
147. Fairbrother , R. W. , and Hurst , E. W. ( 1930 ), The pathogenesis of the propagation of the 
virus in experimental poliomyelitis , J. Path. Bact. , 33 , 17 – 45 . 
148. Faber , H. K. , and Gebhardt , L. D. ( 1938 ), Localization of poliomyelitic virus during 
incubation period after intranasal instillation in monkeys , Proc. Soc. Exper. Biol. Med. , 
30 , 879 – 880 . 
149. Faber , H. K. , and Gebhardt , L. D. ( 1938 ), Localizations of the virus of poliomyelitis. In 
the central nervous system during the pre - paralytic period, after intranasal inoculation , 
J. Exp. Med. , 57 , 933 – 954 . 
150. Sabin , A. B. , and Olitsky , P. K. ( 1936 ), Infl uence of pathway of infection on pathology 
of olfactory bulbs in experimental poliomyelitis , Proc. Soc. Exp. Biol. Med. , 35 , 
300 – 301 . 
151. Bodian , D. , Morgan , I. , and Schwerdt , C. E. ( 1950 ), Virus and host factors infl uencing 
the titer of Lansing poliomyelitis virus in monkeys, cotton rats and mice , Am. J. Hyg. , 
51 , 126 – 133 . 
152. Gopinath , P. G. , Gopinath , G. , and Kumar , T. C. A. ( 1978 ), Target site of intranasally 
sprayed substances and their transport across the nasal mucosa. A new insight into the 
intranasal route of drug delivery , Curr. Ther. Res. , 23 , 596 – 607 . 
153. Hastings , L. ( 1990 ), Sensory neurotoxicology: Use of the olfactory system in the assessment 
of toxicity , Neurotoxicol. Teratol. , 12 , 455 – 459 . 

646 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
154. Hastings , L. , and Evans , J. E. ( 1991 ), Olfactory primary neuron as a route of entry for 
toxic agents into the CNS , Neurotoxicology , 12 , 707 – 714 . 
155. Illum , L. ( 2004 ), Is nose - to - brain transport of drugs in man a reality? J. Pharm. Pharmacol. 
, 56 , 3 – 17 . 
156. Mathison , S. , Nagilla , R. , and Kompella , U. B. ( 1998 ), Nasal route for direct delivery of 
solutes to the central nervous system: Fact or fi ction? J. Drug Target. , 5 , 415 – 441 . 
157. Thorne , R. G. , and Frey , W. H. ( 2001 ), Delivery of neurotropic factors to the central 
nervous system , Brain. Res. , 692 , 278 – 282 . 
158. Pietrowsky , R. , Struben , C. , Molle , M. , Fehm , H. L. , and Born , J. ( 1966 ), Brain potential 
changes after intranasal vs. intravenous administration of vasopressin: Evidence for a 
direct nose - to - brain pathway for peptide effects in humans , Biol. Psychiatry , 39 , 
332 – 340 . 
159. Derad , I. , Willeke , K. , Pietrowsky , R. , Born , J. , and Fehm , H. L. ( 1998 ), Intranasal angiotensin 
II directly infl uences central nervous regulation of blood pressure , Am. J. Hypertens. 
, 11 , 971 – 977 . 
160. Pietrowsky , R. , Thiemann , A. , Kern , W. , Fehm , H. L. , and Born , J. ( 1966 ), A nose - to - brain 
pathway for psychotropic peptides: Evidence from a brain evoked potential study with 
cholecystokinin , Psychoneuroendocrinology , 21 , 559 – 572 . 
161. Lindhardt , K. , Gizurarson , S. , Stefansson , S. B. , Olafsson , D. R. , and Bechgaard , E. ( 2001 ), 
Electroencephalographic effects and serum concentration after intranasal and intravenous 
administration of diazepam to healthy volunteers , Br. J. Clin. Pharmacol. , 52 , 
521 – 527 . 
162. Kern , W. , Peters , A. , Fruehwald - Schultes , B. , Deininger , E. , Born , J. , and Fehm , H. L. 
( 2001 ), Improving infl uence of insulin on cognitive functions in humans , Neuroendocrinology 
, 74 , 270 – 280 . 
163. Okuyama , S. ( 1997 ), The fi rst attempt at radioisotopic evaluation of the integrity of the 
nose - brain barrier , Life Sci. , 60 , 1881 – 1884 . 
164. Quay , S. C. ( 2001 ), Successful delivery of apomorphine to the brain following intranasal 
administration demonstrated in clinical study, PRNewswire, July 18. 
165. Merkus , P. ( 2003 ), Transport of non - peptide drugs from the nose to the CSF, paper presented 
at the Nasal Drug Delivery Meeting, Management Forum, London, Mar. 24 – 25. 
166. Born , J. , Lange , T. , Kern , W. , McGregor , G. P. , Bickel , U. , and Fehm , H. L. ( 2002 ), 
Sniffi ng neuropeptides: A transnasal approach to the human brain , Nat. Neurosci. , 5 , 
514 – 516 . 
167. Seki , T. , Sato , N. , Hasegawa , T. , Kawaguchi , T. , and Juni , K. ( 1994 ), Nasal absorption of 
zidovudine and its transport to cerebrospinal fl uid in rats , Biol. Pharm. Bull. , 17 , 
1135 – 1137 . 
168. Char , H. , Kumar , S. , Patel , S. , Piemontese , D. , Iqbal , K. , Waseem Malick , A. , Salvador , 
R. A. , and Behl , C. R. ( 1992 ), Nasal delivery of [14C] dextromethorphan hydrochloride 
in rats: Levels in plasma and brain , J. Pharm. Sci. , 81 , 750 – 752 . 
169. Sakane , T. , Akizuki , M. , Yoshida , M. , Yamashita , S. , Nadai , T. , Hashida , M. , and Sezaki , 
H. ( 1991 ), Transport of cephalexin to the cerebrospinal fl uid directly from the nasal 
cavity , J. Pharm. Pharmacol. , 43 , 449 – 451 . 
170. Sakane , T. , Akizuki , M. , Yamashita , S. , Sezaki , H. , and Nadai , T. ( 1994 ), Direct drug 
transport from the nasal cavity to the cerebrospinal fl uid: The relation to the dissociation 
of the drug , J. Pharm. Pharmacol. , 46 , 378 – 379 . 
171. Sakane , T. , Akizuki , M. , Yamashita , S. , Nadai , T. , Hashida , M. , and Sezaki , H. ( 1991 ), The 
transport of a drug to the cerebrospinal fl uid directly from the nasal cavity: The relation 
to the lipophilicity of the drug , Chem. Pharm. Bull. , 39 , 2456 – 2458 . 

REFERENCES 647 
172. Sakane , T. , Akizuki , M. , Taki , Y. , Yamashita , S. , Sezaki , H. , and Nadai , T. ( 1995 ), Direct 
drug transport from the nasal cavity to the cerebrospinal fl uid: The relation to the 
molecular weight of the drugs , J. Pharm. Pharmacol. , 47 , 379 – 381 . 
173. Dahlin , M. , Jansson , B. , and Bjork , E. ( 2001 ), Levels of dopamine in the blood and brain 
following nasal administration to rats , Eur. J. Pharm. Sci. , 14 , 75 – 80 . 
174. Wang , Y. , Aun , R. , and Tse , F. L. S. ( 1998 ), Brain uptake of dihydroergotamine 
after intravenous and nasal administration in the rat , Biopharm. Drug Dispos. , 19 , 
571 – 575 . 
175. Gizurarson , S. , Trovaldsson , T. , Sigurdsson , P. , and Gunnarsson , E. ( 1996 ), Selective 
delivery of insulin into the brain: Introlfatory absorption , Int. J. Pharm. , 140 , 77 – 83 . 
176. Wang , F. , Jiang , X. , and Lu , W. ( 2003 ), Profi les of methotrexate in blood and CSF following 
intranasal and intravenous administration to rats , Int. J. Pharm. , 263 , 1 – 7 . 
177. Frey , W. H. , Liu , J. , Thorne , R. G. , and Rahman , Y. E. ( 1995 ), Intranasal delivery of 
125 I - labelled nerve growth factor to the brain via the olfactory route , in Iqbal , K. , Mortimer 
, J. A. , Winbald , B. , and Wisniewski , H. M. , Eds., Research Advances in Alzheimer 
’ s Disease and Related Disorders , Wiley , New York , pp. 329 – 335 . 
178. Thorne , R. G. , Emroy , C. R. , Ala , T. A. , and Frey , W. H. ( 1995 ), Quantitative analysis 
of the olfactory pathway for drug delivery to the brain , Brain. Res. , 692 , 278 – 282 . 
179. Vyas , T. K. , Babbar , A. K. , Sharma , R. K. , and Misra , A. ( 2005 ), Intranasal mucoadhesive 
microemulsions of zolmitriptan: Preliminary studies on brain - targeting , J. Drug 
Target. , 13 , 317 – 324 . 
180. Fliedner , S. , Schulz , C. , and Lehnert , H. ( 2006 ), Brain uptake of intranasally applied 
radioiodinated leptin in wistar rats , Endocrinology , 147 , 2088 – 2094 . 
181. Westin , U. E. , Bostr o m , E. , Gr a sj o , J. , Hammarlund - Udenaes , M. , and Bj o rk , E. ( 2006 ), 
Direct nose - to - brain transfer of morphine after nasal administration to rats , Pharm. 
Res. , 23 , 565 – 572 . 
182. Zhang , Q. Z. , Zha , L. S. , Zhang , Y. , Jiang , W. M. , Lu , W. , Shi , Z. Q. , Jhang , X. G. , and 
Fu , S. K. ( 2006 ), The brain targeting effi ciency following nasally applied MPEG - PLA 
nanoparticles in rats , J. Drug Target. , 14 , 281 – 290 . 
183. Shi , Z. , Zhang , Q. , and Jiang , X. ( 2005 ), Pharmacokinetic behavior in plasma, cerebrospinal 
fl uid and cerebral cortex after intranasal administration of hydrochloride meptazinol 
, Life Sci. , 77 , 2574 – 2583 . 
184. Wang , X. , Haibing , H. , Leng , W. , and Tang , X. ( 2006 ), Evaluation of brain - targeting 
for the nasal delivery of estradiol by the microdialysis method , Int. J. Pharm. , 317 , 
40 – 46 . 
185. Jones , N. ( 2001 ), The nose and paranasal sinuses physiology and anatomy , Adv. Drug 
Deliv. Rev. , 51 , 5 – 19 . 
186. Graff , C. L. , and Pollack , G. M. ( 2005 ), Nasal drug administration: Potential for targeted 
central nervous system delivery , J. Pharm. Sci. , 94 , 1187 – 1195 . 
187. Sherwood , L. ( 1989 ), Human Physiology. From Cells to Systems , West Publishing , 
St. Paul, MN , Chapter 5. 
188. Stanley , P. A. , and Walter , O. A. (2004), in Plotkin, S. A. , Orenstein, W. A. , and Offi t, P. A. , 
Eds., A Short History of Vaccination. Vaccines , 4th ed., Saunders , Philadelphia, pp. 1 – 15 . 
189. Ogra , P. L. , Faden , H. , and Welliver , R. C. ( 2001 , Apr.), Vaccination strategies for 
mucosal immune responses , Clin. Microbiol. Rev. , 14 ( 2 ), 430 – 445 . 
190. Cui , Z. R. , and Mumper , R. J. ( 2002 ), Intranasal administration of plasmid DNA - coated 
nanoparticles results in enhanced immune responses , J. Pharm. Pharmacol. , 54 , 1195 – 
1203 . 
191. Beverley , L. C. P. ( 2002 ), Immunology of vaccination , Br. Med. Bull. , 62 , 15 – 28 . 

648 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
192. Illum , L. , and Davis , S. S. ( 2001 ), Nasal vaccination: A non - invasive vaccine delivery 
method that holds great promise for the future , Adv. Drug Deliv. Rev. , 51 , 1 – 3 . 
193. Davis , S. S. ( 2001 ), Nasal vaccines , Adv. Drug Deliv. Rev. , 51 , 21 – 42 . 
194. Partidos , C. D. ( 2000 ), Intranasal vaccines: Forthcoming challenges , PSTT , 3 , 273 – 281 . 
195. Mestecky , J. , and McGhee , J. R. ( 1987 ), Immunoglobulin A: Molecular and cellular 
interactions in IgA biosynthesis and immune response , Adv. Immunol. , 40 , 153 – 245 . 
196. Gill , J. I. , Lin , W. , Kistner , O. , Davis , S. S. , and Illum , L. ( 2001 ), Polymeric lamellar substrate 
particles for intranasal vaccination , Adv. Drug Deliv. Rev. , 51 , 97 – 111 . 
197. Biesterveld , N. ( 2002 ), Nasal vaccine delivery, vaccine delivery adjuvants, Merck & 
Company, West Point, PA, a Literature review submitted to the Department of 
Chemistry, Lehigh University, Aug. 2002. 
198. Gander , B. ( 2005 ), Trends in particulate antigen and DNA delivery systems for vaccines , 
Adv. Drug Deliv. Rev. , 57 , 321 – 323 . 
199. Friede , M. , and Aguado , T. M. ( 2005 ), Need for new vaccine formulation and potential 
of particulate antigen and DNA delivery system , Adv. Drug Deliv. Rev. , 57 , 325 – 331 . 
200. Illum , L. ( 2003 ), Nasal drug delivery – possibilities, problems and solutions , J. Controlled 
Release , 87 , 187 – 198 . 
201. Alpar , O. H. , Somavarapu , S. , Atuah , N. K. , and Bramwell , W. V. ( 2005 ), Biodegradable 
mucoadhesive particulates for nasal and pulmonary antigen and DNA delivery . Adv. 
Drug Deliv. Rev. , 57 , 411 – 430 . 
202. Mackay , M. , Williamson , I. , and Hastewell , J. ( 1991 ), Cell biology of Epithelia , Adv. Drug 
Deliv. Rev. , 7 , 313 – 338 . 
203. Storni , T. , Kundig , M. T. , Senti , G. , and Johansen , P. ( 2005 ), Immunity in response to 
particulate antigen - delivery systems , Adv. Drug Deliv. Rev. , 57 , 333 – 355 . 
204. O ’ Hagan , T. D. , and Rappuoli , R. ( 2004 ), Novel approaches to vaccine delivery , Pharm. 
Res. , 21 ( 9 ), 1519 – 1530 . 
205. McNeela , A. E. , and Mills , G. H. K. ( 2001 ), Manipulating the immune response: Humoral, 
versus, cell, mediated, immunity , Adv. Drug Deliv. Rev. , 51 , 43 – 54 . 
206. Edelman , R. ( 2002 ), The development and use of vaccine adjuvants , Mol. Biotechnol. , 
21 , 129 – 148 . 
207. Singh , M. , and O ’ Hagan , D. ( 1999 ), Advances in vaccine adjuvants , Nat. Biotechnol. , 17 , 
1075 – 1081 . 
208. McNeela , E. A. , O ’ Connor , D. , Gill , J. I. , Illum , L. , Davis , S. S. , Pizza , M. , Peppoloni , S. , 
Rappuoli , R. , and Mills , G. H. K. ( 2000 ), A mucosal vaccine against diphtheria: Formulation 
of cross reacting material (CRM 197 ) of diphtheria toxin with chitosan enhances local 
and systemic antibodies and Th2 responses following nasal delivery , Vaccine. , 19 , 
1188 – 1198 . 
209. Illum , L. , Gill , J. I. , Hinchcliffe , M. , Fisher , N. A. , and Davis , S. S. ( 2001 ), Chitosan as a 
novel nasal delivery system for vaccines , Adv. Drug Deliv. Rev. , 51 , 81 – 96 . 
210. Jabbal - Gill , I. , Fisher , A. N. , Rappuoli , R. , Davis , S. S. , and Illum , L. ( 1998 ), Stimulation 
of mucosal and systemic antibody responses against Bordetella pertussis fi lamentous 
haemagglutinin and recombinant pertussis toxin after nasal administration with chitosan 
in mice , Vaccine , 16 , 2039 – 2046 . 
211. Boyaka , P. N. , Marinaro , M. , Jackson , R. J. , Menon , S. , Kiyona , H. , Jirillo , E. , and McGhee , 
J. R. ( 1999 ), IL - 12 is an effective adjuvant for induction of mucosal immunity , J. Immunol. , 
162 , 122 – 128 . 
212. Arulanandan , B. P. , O ’ Toole , M. , and Metzger , D. W. ( 1999 ), Intranasal interleukin - 12 is 
a powerful adjuvant for protective mucosal immunity , J. Infect. Dis. , 180 , 940 – 949 . 

REFERENCES 649 
213. McCluskie , M. J. , and Davis , H. L. (2001), Oral, intrarectal and intranasal immunizations 
using CpG and non - CpG oligodeoxynucleotides as adjuvants , Vaccine. , 19 , 413 – 422 . 
214. Moldoveanu , Z. , Love - Homan , L. , Huang , W. Q. , and Krieg , A. M. ( 1998 ), CpG DNA, a 
novel immune enhancer for systemic and mucosal immunization with infl uenza virus , 
Vaccine , 16 , 1216 – 1224 . 
215. de Haan , A. , Geerligs , H. J. , Huchshorn, J. P. , van Scharrenburg , G. J. , Palache , A. M. , and 
Wilschut , J. ( 1995 ), Mucosal immunoadjuvant activity of liposomes: Induction of systemic 
IgG and secretory IgA responses in mice by intranasal immunization with an 
infl uenza subunit vaccine and coadministered liposomes , Vaccine , 13 , 155 – 162 . 
216. de Haan , A. , Tomee , J. F. C. , Huchshorn , J. P. , and Wilschut , J. ( 1995 ), Liposomes as an 
immunoadjuvant system for stimulation of mucosal and systemic antibody response 
against inactivated measles virus administered intranasally to mice , Vaccine , 13 , 1320 – 
1324 . 
217. Aramaki , Y. , Fujii , Y. , Yachi , K. , Kikuchi , H. , and Tsuchiya , S. ( 1994 ), Activation of systemic 
and mucosal immune response following nasal administration of liposomes , 
Vaccine , 12 , 1241 – 1245 . 
218. Childers , N. K. , Tong , G. , Mitchell , S. , Kirk , K. , Russel , M. W. , and Michalek , S. M. ( 1999 ), 
A controlled clinical study of the effect of nasal immunization with a Streptococcus 
mutans antigen alone or incorporated into liposomes on induction of immune responses , 
Infect. Immun. , 67 , 618 – 623 . 
219. Gluck , U. , Gebbers , J. O. , and Gluck , R. ( 1999 ), Phase I evaluation of intranasal virosomal 
infl uenza vaccine with and without Escherichia coli heat - labile toxin in adult volunteers 
, J. Virol. , 73 , 7780 – 7786 . 
220. Gl u ck , R. ( 1999 ), Adjuvant activity of immunopotentiating reconstituted infl uenza virosomes 
(IRIVs) , Vaccine , 17 , 1782 – 1787 . 
221. Baca - Estrada , M. E. , Foldvari , M. , Snider , M. , Harding , K. , Kournikakis , B. , Babiuk , 
L. A. , and Griebel , P. ( 2000 ), Intranasal immunization with liposome - formulated Yersinia 
pestis vaccine enhances mucosal immune responses , Vaccine , 18 , 2203 – 2211 . 
222. L o vgren , K. , Kaberg , H. , and Morein , B. ( 1990 ), An experimental infl uenza subunit 
vaccine (iscom): Induction of protective immunity to challenge infection in mice after 
intranasal or subcutaneous administration , Clin. Exp. Immunol. , 82 , 435 – 439 . 
223. Hsu , S. C. , Schadeck , E. B. , Delmas , A. , Shaw , M. , and Steward , M. W. ( 1996 ), Linkage 
of a fusion peptide to a CTL epitope from the nucleoprotein of measles virus enables 
incorporation into ISCOMS and induction of CTL responses following intranasal immunization 
, Vaccine , 14 , 1159 – 1166 . 
224. Carol, H. , Nieto , A. , Villacres-Eriksson, M. , and Morein, B. (1997), Intranasal immunization 
of mice with Echinococcus granulosus surface antigens iscoms evokes a strong 
immune response, biased towards glucidic epitopes , Parasite Immunol. , 19 , 197 – 205 . 
225. Hu , K. F. , Elvander , M. , Merza , M. , Akerblom , L. , BrAndenburg , A. , and Morein , B. 
( 1998 ), The immunostimulatory complex (ISCOM) is an effi cient mucosal delivery 
system for respiratory syncytial virus (RSV) envelope antigens inducing high local and 
systemic antibody responses , Clin. Exp. Immunol. , 113 , 235 – 243 . 
226. Jaganathan , K. S. , and Vyas , S. P. ( 2006 ), Strong systemic and mucosal immune responses 
to surface - modifi ed PLGA microspheres containing recombinant hepatitis B antigen 
administered intranasally , Vaccine , 24 , 4201 – 4211 . 
227. Debin , A. , Kravtzoff , R. , Vaz Santiago , J. , Cazales , L. , Speranido , S. , Melber , K. , Janowicz , 
Z. , Betbeder , D. , and Moynier , M. ( 2002 ), Intranasal immunization with recombinant 
antigens associated with new cationic particles induces strong mucosal as well as systemic 
antibody and CTL responses , Vaccine , 20 , 2752 – 2763 . 

650 NASAL DELIVERY OF PEPTIDE AND NONPEPTIDE DRUGS 
228. Aguilar , J. C. , Lobaina , Y. , Muzio , V. , Garcia , D. , Penton , E. , Iglesias , E. , Pichardo , D. , 
Urquiza , D. , Rodeiguez , D. , Silva , D. , Petrovsky , N. , and Guillen , G. ( 2004 ), Development 
of a nasal vaccine for chronic hepatitis B infection that uses the ability of hepatitis B 
core antigen to stimulate a strong Th1 response against hepatitis B surface antigen , 
Immnol. Cell. Biol. , 82 , 539 – 546 . 
229. Hordness , K. , Tynning , T. , Brown , T. A. , Haneberg , B. , and Jonsson , R. ( 1997 ), Nasal 
immunization with group B streptococci can induce high levels of specifi c IgA antibodies 
in cervicovaginal secretions of mice , Vaccine , 15 , 1244 – 1251 . 
230. Kurono , Y. , Yamamoto , M. , Fujihashi , K. , Kodama , S. , Suzuki , M. , Mogi , G. , McGhee , 
J. R. , and Kiyono , H. ( 1991 ), Nasal immunization induces Haemophilis infl uenzae - 
specifi c Th1 and Th2 responses with mucosal IgA and systemic IgG antibodies for protective 
immunity , J. Infect. Dis. , 180 , 122 – 132 . 
231. Hagiwara , Y. , Komase , K. , Chen , Z. , Matsuo , K. , Suzuki , Y. , Aizawa , C. , Kurata , T. , and 
Tamura , S. ( 1999 ), Mutants of cholera toxin as an effective and safe adjuvant for nasal 
infl uenza vaccine , Vaccine , 17 , 2918 – 2926 . 
232. Bastein , N. , Trudel , M. , and Simard , C. ( 1999 ), Complete protection of mice from respiratory 
syncytial virus infection following mucosal delivery of synthetic peptide vaccines , 
Vaccine , 17 , 832 – 836 . 

651 
5.7 
NASAL POWDER DRUG DELIVERY 
Jelena Filipovi - Gr i and Anita Hafner 
University of Zagreb, Zagreb, Croatia 
Contents 
5.7.1 Introduction 
5.7.2 Nasal Dry Powder Formulations 
5.7.2.1 Benefi ts Associated with Nasal Powder Drug Delivery 
5.7.2.2 Drug Powder or Drug/Polymer Powder Formulation for Nasal Drug 
Delivery? 
5.7.2.3 Powder Properties Affecting Nasal Deposition and Drug Delivery 
5.7.3 Polymers in Nasal Powder Delivery System 
5.7.4 Microspheres as Nasal Drug Delivery Devices 
5.7.4.1 Preparation Methods 
5.7.4.2 Microsphere Characterization 
5.7.4.3 Chitosan - Formulated Spray - Dried Microspheres 
5.7.5 Toxicological Considerations 
References 
5.7.1 INTRODUCTION 
Intranasal drug administration has been practiced since ancient times. In Tibet 
extracts of sandalwood and aloewood were inhaled to treat emesis. Egyptians 
treated epistaxis and rhinitis using intranasal medication. North American Indians 
relieved headaches inhaling crushed leaves of Ranunculus acris [1] . Due to the rich 
vasculature and high permeability of nasal mucosa, the absorption rate and pharmacokinetics 
of nasally administrated drug are comparable to that obtained by 
intravenous drug delivery, while noninvasive nasal drug administration is more 
convenient to patients. As nasally administrated drugs avoid fi rst - pass hepatic 
metabolism, improved bioavailability can be expected. However, rapid mucociliary 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc. 
c c c

652 NASAL POWDER DRUG DELIVERY 
clearance reduces the residence time of nasal drug delivery system at the site of 
absorption. Dry powders have been shown to delay mucociliary clearance, thus 
prolonging the contact time between the drug delivery system and mucosa compared 
to liquid formulations. Most of the dry powder investigations are based on 
mucoadhesive swellable polymers as they can additionally improve drug absorption 
and bioavailability. Dry powder delivery systems such as microspheres are of special 
interest, offering the possibility of predictable and controlled drug release from the 
polymeric device [2, 3] . 
5.7.2 NASAL DRY POWDER FORMULATIONS 
Liquid preparations are most frequently used nasal dosage forms at present. 
However, such preparations are characterized with short residence time in the nasal 
cavity, low drug concentration at the site of absorption, and problems linked to the 
chemical stability of the drug and the stability of the preparation. In the case of 
liquid formulations, drugs must be administered in small volumes. The maximum 
volume of a single dose in one nostril is about 200 . L. The volume of therapeutic 
dose should not exceed the capacity of the nasal cavity, as it would drain out of the 
nose. Thus, only low - dose or highly soluble drugs can be administered nasally in the 
form of a simple liquid formulation [4] . Dry powder formulations have been 
recognized as effi cient nasal delivery systems offering numerous advantages over 
liquid formulations, such as avoidance of preservatives, improved formulation 
stability, and prolonged contact with the mucosa. For a powder formulation, the 
maximum quantity is approximately 50 mg, depending upon the bulk density of the 
material [5] . 
5.7.2.1 Benefi ts Associated with Nasal Powder Drug Delivery 
A powder form was found to be more effective than liquid formulations in a number 
of investigations described in the literature [6 – 11] . Dry powders are characterized 
by prolonged residence time and higher drug concentration at the site of deposition 
as well as improved formulation stability with no requirement for preservatives [12] . 
Prolonged residence time of the powder delivery systems at the absorption site 
results in enhanced systemic bioavailability, compared to the liquid formulations. In 
the case of powders, higher drug concentration at the site of absorption causes rapid 
transmucosal diffusion and faster onset of action [13] . Most of the dry powder formulations 
are based on mucoadhesive swellable polymers (e.g., starch, dextran, 
chitosan) as they can additionally improve drug bioavailability, prolonging the residence 
time in the nasal cavity or even promoting drug absorption. Powder formulation 
with a water - insoluble and nonswellable drug carrier may also improve nasal 
bioavailability of the polar drugs. Ishikawa et al. [14] found that a nasal powder 
delivery system of elcatonin based on CaCO 3 signifi cantly increased the systemic 
elcatonin bioavailability in rats and rabbits compared to the liquid formulation. 
Enhanced bioavailability has been primarily ascribed to the retardation of the clearance 
of the drug powder delivery system from the nasal cavity. 
The use of dry powder formulations in nasal vaccine delivery has been extensively 
reviewed elsewhere [15 – 18] . The association of vaccines to some of the par

ticulate systems has been proved to enhance the systemic and mucosal immune 
responses against the antigens [19 – 21] . Dry powder formulations for nasal vaccine 
delivery may also provide signifi cant advantages with respect to stability shortcomings 
compared to conventional liquid intranasal and intramuscular formulations, 
which require frozen storage or refrigeration [22] . 
5.7.2.2 Drug Powder or Drug/Polymer Powder Formulation for 
Nasal Drug Delivery? 
The drug candidate for nasal administration should possess a number of attributes, 
such as appropriate aqueous solubility and nasal absorption characteristics, minimal 
nasal irritation, low dose, no offensive odor or aroma, and suitable stability characteristics 
[23] . In the case of drug powder formulations it is possible to hide or alter 
the unfavorable characteristics of a drug using suitable polymers as drug carriers. 
Thus, improvement of the dissolution behavior of drugs of low aqueous solubility 
after incorporation in polymeric powder devices such as microspheres has been 
reported in the literature [24, 25] . The improvement of the drug dissolution rate 
from the microspheres has been ascribed to several factors, such as high microsphere 
surface – volume ratio, the hydrophilic nature of the polymer, and drug amorphization 
due to drug – polymer interaction and/or the microsphere preparation method 
[25, 26] . 
Nasally administrated polymer – drug powders were also characterized by 
improved drug absorption compared to pure drug powders [25, 27] . Teshima et al. 
[27] monitored changes in the plasma glucagons and glucose concentrations after 
nasal administration of the powder form of glucagon alone and glucagon mixed with 
the carrier, microcrystalline cellulose (MCC). Glucagon and glucose plasma concentrations 
remained unchanged after nasal administration of the powder form of glucagon 
alone while it increased after glucagon – MCC administration in an MCC 
content – dependent manner. Results of in vivo nasal administration of carbamazepine 
- loaded chitosan microspheres revealed an increase in carbamazepine concentration 
in serum compared to the pure carbamazepine powder [25] . Such an increase 
in drug absorption has been ascribed to both improved dissolution of carbamazepine 
and adhesion of the chitosan microspheres to the mucosal surface. 
The infl uence of polymers on the drug stability in the powder formulation has 
also been reported [8] . Green coloration of polyacrylic acid powder dosage forms 
loaded with 60% apomorphine due to atmospheric drug oxidation upon storage has 
been observed. Dosage forms with lower drug loadings (and higher polymer content) 
showed no coloration, indicating the protective role of polymers against drug oxidation 
in powder formulations. 
Powders intended for nasal administration have to be optimized in terms of particle 
size and morphology as these properties are related to potential irritation in 
the nasal cavity [23] . Certain procedures (e.g., spray drying process) can modify the 
particle size of the drug powder raw material, but in order to optimize the morphology 
and fl owability properties of some pure drug powders, excipients need to be 
used. Sacchetti et al. [28] reported that the use of mannitol as a fi ller and hydroxypropylmethyl 
cellulose (HPMC) as a shaper of spray - dried caffeine microparticles 
modifi ed the typical needle shape of spray - dried caffeine to a more convenient 
roundish shape. Further addition of polyethylene glycol (PEG) resulted in increased 
NASAL DRY POWDER FORMULATIONS 653

654 NASAL POWDER DRUG DELIVERY 
cohesiveness between particles, producing agglomerates considered as benefi cial for 
nasal deposition. 
5.7.2.3 Powder Properties Affecting Nasal Deposition and Drug Delivery 
Powder formulation effi ciency is strongly dependent on two technological aspects 
of delivery: fi rst, the complete release of the dose through the powder delivery 
device, that is, the quantitative aspect, and, second, the distribution of the powder 
on the nasal mucosa, that is, the qualitative aspect. For example, rhinitis treatment 
requires drug deposition over the entire mucosal area, while brain targeting requires 
deposition strictly at the roof of the nose. The qualitative aspect is defi ned by the 
aerodynamic properties of the powder cloud produced (puff) and is assessed by a 
photographic technique providing information on the spray pattern and cloud 
geometry. The parameters involved are shape, height, area, density, particle size, and 
velocity. For nasal impacting the powder cloud should remain as compact as possible 
to achieve an effi cient shot of powder to the nasal mucosa, whereas for sedimentation 
a larger cloud would be preferred [12] . 
The quantitative aspect of nasal powder delivery is largely dependent on the type 
of delivery device. Devices for powder dosage forms, including insuffl ators, monodose 
and multidose powder inhalers, and pressurized metered - dose inhalers, are 
extensively reviewed elsewhere [29, 30] . Particles intended for nasal delivery should 
have good fl ow properties to be reproducibly fi lled in the dose reservoir and easily 
insuffl ated to obtain appropriate nasal deposition [28] . Since the particle size of the 
applied powder formulation has a major impact on its nasal deposition, the characterization 
of this parameter is very important. Only particles over 5 . m are deposited 
in the nostrils while smaller particles can be inhaled into the lower parts of the 
respiratory system [23] . For that reason, it is necessary to determine particle sizes 
not only prior to fi lling the nasal delivery device but also after its actuation, since 
the particle size of the formulation leaving the device depends on the device - disaggregating 
properties [29] . Furthermore, it is known that powder deposition in the 
anterior part of the nasal epithelium contributes to nasal absorption more than 
powder deposition in the posterior part, since ciliated cells cover mainly the posterior 
part of the nasal epithelium, providing a faster clearance of particles [31, 32] . 
Thus, deposition studies must include both the amount of deposited particles and 
the location of the particle deposition [29] . Distribution of the formulation in the 
nasal cavity should be evaluated considering two different deposition types: the 
initial deposition immediately after application and the secondary deposition, owing 
to translocation by mucociliary clearance [29] . Cornaz et al. [33] investigated the 
ability of microsphere delivery, studying the shape and area of the clouds of microspheres 
emitted from a nasal device. The puff areas of loaded particles were smaller 
compared to unloaded particles due to different particle porosities. Pringels et al. 
[31] investigated the infl uence of the deposition pattern and spray characteristics of 
nasal powder formulations on insulin bioavailability. It has been shown that posterior 
deposition of the powder formulation in the nasal cavity lowered insulin bioavailability. 
To study the spray characteristics, the shape and cross section of the 
emitted powder cloud (puff) were analyzed. It has been concluded that the powder 
bulk density of the formulation infl uenced the spray pattern. Comparing two 
powders with different bulk densities and particle sizes, it has been revealed that 

the powder with higher bulk density and smaller particle size was more compact 
and was characterized by higher resistance to airfl ow, resulting in a slower spray 
time and larger spray pattern [31] . No infl uence of the powder bulk density and 
spray pattern on insulin bioavailability has been observed. 
5.7.3 POLYMERS IN NASAL POWDER DELIVERY SYSTEM 
It has been demonstrated that low absorption of drugs can be improved by using 
absorption enhancers or prolonging contact between drug and absorptive sites in 
the nasal cavity by delaying mucociliary clearance of the formulation. Some mucoadhesive 
polymers can serve both functions. They are typically high - molecular - 
weight polymers with fl exible chains which can interact with mucin through hydrogen 
bonding, electrostatic, hydrophobic or van der Waals interactions [18, 34, 35] . 
The mucoadhesive polymers are often hydrophilic and swellable, containing 
numerous hydrogen bond – forming groups such as hydroxyl, carboxyl, or amine, 
which favors adhesion. When used in a dry form they attract water from the mucosal 
surface and swell, leading to polymer – mucus interaction, increased viscosity of 
polymer – mucus mixture, and reduced mucociliary clearance [34] . Beside the type 
of polymer functional groups, the mucoadhesive force of a polymer material is 
dependent on the polymer molecular weight, concentration, fl exibility of the polymer 
chain, spatial conformation, contact time, environmental pH, and physiological 
factors such as mucin turnover and disease state [3] . There is a critical polymer 
molecular weight for each polymer type below or above which there is reduced 
adhesive power [34] . The mucoadhesive properties can also be affected by the 
degree of cross - linking of the polymer since mucoadhesion requires an adequate 
free chain length for interpenetration to occur. Hence, the more cross - linked the 
polymer, the less strong the mucoadhesive interaction [36] . Hydration and swelling 
present both polymer - and environment - related factors. Overhydration causes 
extended swelling, resulting in slippery mucilage formation [30] . The polymer concentration 
that is required for optimum mucoadhesion is different between gels and 
solid mucoadhesives. In the liquid state, an optimum concentration exists for each 
polymer for which best adhesion can occur, while with solid dosage forms, increased 
polymer concentration leads to increased mucoadhesive power [35] . Studies have 
shown that polymers with charge density can serve as good mucoadhesive agents, 
although their mucoadhesive properties are affected by the pH of the surrounding 
media. The presence of metal ions, which can interact with charged polymers, may 
also affect the adhesion process [35, 36] . It has also been reported that polyanion 
polymers are more effective bioadhesives than polycation polymers or nonionic 
polymers [37] . 
Research on nasal powder drug delivery has employed polymers such as starch, 
dextrans, polyacrylic acid derivatives (e.g., carbopol, polycarbophil), cellulose derivatives 
(microcrystalline cellulose, semicrystalline cellulose, hydroxypropylmethyl 
cellulose, hydroxypropyl cellulose, carboxymethyl cellulose), chitosan, sodium alginate, 
hyaluronans, and polyanhydrides such as poly(methyl vinyl ether - co - maleic 
anhydride) (PVM/MA). Many of these polymers have already been used as excipients 
in pharmaceutical formulations and are often referred to as fi rst - generation 
bioadhesives [38 – 45] . In nasal dry powder a single bioadhesive polymer or a 
POLYMERS IN NASAL POWDER DELIVERY SYSTEM 655

656 NASAL POWDER DRUG DELIVERY 
combination of two or more polymers has been formulated as freeze - dried or spray - 
dried particles or micropheres. 
Crystalline cellulose, hydroxypropyl cellulose, and Carbopol 934 have been 
studied in combination with lyophilized insulin as bioadhesive powder dosage forms 
for nasal delivery. Each formulation tested resulted in an decrease in plasma glucose 
level after nasal administration in dog and rabbit models. The most effective formulation, 
crystalline cellulose blended with insulin, decreased the plasma glucose level 
to 49% of the control value. In ternary systems the lyophilized Carbopol 934 and 
insulin blend with crystalline cellulose powder has been the most effective, leading 
to a hypoglycemia on the order of one - third of the effect obtained after intravenous 
injection of the same dose of insulin. The plasma glucose levels obtained in the 
volunteers after administration of the insulin – Carbopol – crystalline cellulose powder 
formulation were quite variable [38] . 
The various powder formulations were prepared by dry blending of octreotide 
with microcrystalline cellulose, semicrystalline cellulose, hydroxyethyl starch, cross - 
linked dextran, microcrystalline chitosan, pectin, and alginic acid [40] . Their potential 
to enhance the nasal absorption of the somatostatin analogue peptide octreotide 
was studied in vivo in the rat model. The powder mixtures were also characterized 
in vitro regarding calcium binding, water uptake, and drug release. The bioavailabilities 
obtained for all of the powder formulations were low, with the highest 
values for alginic acid and cross - linked dextran powder formulations (4.1 and 5.56%, 
respectively). 
Callens and Remon [46] have shown improved nasal absorption of insulin in 
rabbits by using a bioadhesive powder formulation containing drum - dried waxy 
maize starch (mainly amylopectin) and Carbopol 974P. The bioavailability of 14% 
has been obtained. They have shown [47] that the initial advantage of a longer residence 
time of the powder formulation in the nasal cavity might turn into a disadvantage 
after multiple administration and impact bioavailability. They investigated 
the infl uence of eight daily administrations of two powder formulations to rabbits 
on the bioavailability and therapeutic effect of the insulin [47] . The fi rst powder 
formulation consisted of a co - spray - dried mixture of Amioca starch and Carbopol 
974P and the second one has been a physical mixture of drum - dried waxy maize 
starch and Carbopol 974P. By a single nasal administration to rabbits, absolute bioavailabilities 
of 17.8 and 13.4% have been obtained, respectively. The lower insulin 
bioavailabilities (4.4 and 3.6%, respectively) after multiple administrations were 
observed with both formulations, mainly due to the high viscosity of the bioadhesive 
powders in the nasal mucus, causing a physical barrier toward absorption and a 
strongly decelerated mucociliary clearance. Long residence times of the powder 
formulations were also reported by Ugwoke et al. [42, 48] , who noticed nasal residence 
times of more than 24 h using powder formulations containing Carbopol 971P 
and carboxymethyl cellulose. 
Rhinocort is a commercially available mucoadhesive transnasal powder preparation 
of beclomethasone dipropionate with hydroxypropyl cellulose (HPC) as a gel - 
forming drug carrier developed by Suzuki and Makino [49] . The HPC has been 
shown to promote the absorption of low - molecular - weight drugs, but it was not that 
effective with a peptide drug salmon calcitonin. Microcrystalline cellulose has been 
shown to be effective for the promotion of absorption of calcitonin in humans, 
producing about 10% bioavailability with rapid absorption onset [49] . In the study 

of the effect of an HPC and MCC combination on the development of nasal powder 
preparations for peptide delivery, signifi cant absorption enhancement of leuprolide, 
calcitonin, and fl uorescein isothiocyanate (FITC) – dextran in rabbits has been 
obtained by the addition of 10 – 20% HPC to MCC. It has been suggested that MCC 
works as an absorption enhancer by causing a locally high concentration of drugs 
in the vicinity of the mucosa surface while HPC works to increase retention of drugs 
on the nasal mucosa due to its gel - forming property. In a comparative study [50] of 
a series of MCC nasal sprays and lyophilized powder formulations of ketorolac, the 
spray formulations have been shown to be better absorbed than powder formulations. 
The absolute bioavailability of ketorolac from a powder formulation has been 
38%, and no signifi cant differences in absorption between different powder formulations 
have been observed. 
Lim et al. [51, 52] compared a number of mucoadhesive microspheres prepared 
by solvent evaporation composed of hyaluronic acid (HA), chitosan glutamate 
(CH), and a combination of the two with microcapsules of HA and gelatin prepared 
by complex coacervation. Some other polymers — such as alginates [53, 54] , a natural 
polymer of low toxicity, irritability, and immunogenicity; epichlorohydrine cross - 
linked starch (Spherex) [55 – 61] and dextran (Sephadex) [62 – 65] ; poly(lactide - co - 
glycolide) (PLGA) [66] ; and the biocompatible and biodegradable copolymer of 
lactic and glycolic acids, which have also been approved by the Food and Drug 
Administration (FDA) [67] — have mainly been used in microspheres for nasal dry 
powder delivery and are referred to in more detail in the next section. 
Recently, thiolated polymers or thiomers, a new generation of permeation - 
enhancing agents, have been introduced in the pharmaceutical literature. Thiomers 
are characterized by covalent immobilization of sulfhydryl groups on their polymeric 
backbone, which are responsible for improved permeation - enhancing properties 
combined with mucoadhesive and enzyme - inhibitory properties [68] . A further 
improvement of the permeation - enhancing effect of thiomers has been achieved by 
the addition of the permeation mediator glutathione [69] . The improvement of 
human growth hormone (hGH) bioavailability (8.11%) by intranasal administration 
of the microparticulate formulation composed of thiomer polycarbophil - cysteine 
(PCP - Cys) and permeation mediator glutathione has been shown. Evaluation of the 
effect of PCP - Cys on the ciliary beat frequency (CBF) of human nasal epithelial 
cells in vitro has shown no ciliotoxic effect [70] . 
Chitosan is a hydrophilic, biocompatible, and biodegradable polymer of low 
toxicity, and it has been extensively investigated for pharmaceutical and medical 
purposes. It has been included in the European Pharmacopoeia since 2002. Chitosan 
is a polysaccharide composed of N - acetyl - d - glucosamine (approximately 20%) and 
glucosamine (approximately 80%). It is derived by deacetylation of chitin, which 
after cellulose is the most abundant polymer found in nature. It is a polycation at 
acidic pH values where most of the amino groups are protonated and has an apparent 
p K a of 5.5 [71 – 74] . In the context of drug delivery, chitosan has been used for 
the preparation of microcapsules and microspheres with encapsulated small polar 
molecules, proteins, enzymes, DNA, and cells, as a nasal delivery system for insulin 
[75] , as a system for oral vaccination, and as a stabilizing constituent of liposomes. 
Several studies have highlighted the potential use of chitosan as an absorption - 
enhancing agent due to its mucoadhesive properties and ability to open the tight 
junctions in the mucosal cell membrane [72] . 
POLYMERS IN NASAL POWDER DELIVERY SYSTEM 657

658 NASAL POWDER DRUG DELIVERY 
Its biodegradability and low toxicity in humans have aided the recent increased 
interest in chitosan as an immunopotentiating agent. In vivo studies have demonstrated 
that chitosan powder and solution formulations are able to enhance 
the systemic and mucosal immune responses after nasal vaccine delivery [19, 22, 
76] . 
The nasal absorption of insulin after administration in chitosan powder was the 
most effective formulation for nasal delivery of insulin in sheep compared to chitosan 
nanoparticles and chitosan solution [11] . Similarly, chitosan powder formulations 
have been shown to enable an effi cient nasal absorption of goserelin in a sheep 
model where bioavailabilities of 20 – 40% were obtained depending on the nature 
of the formulation [9] . 
There has been a report on chitosan utility in improving the intranasal absorption 
of high - molecular - weight ( > 10 - kDa) therapeutic protein. Chitosan glutamate 
powder blend or granules with recombinant hGH have been evaluated for intranasal 
administration in sheep. Relative to subcutaneous injection the nasal formulations 
produced bioavailabilities of 14 and 15%, respectively [77] . 
Various chitosan derivatives of enhanced solubility, mucoadhesive, and permeation 
properties were developed. N - Trimethyl chitosan chloride (TMC) is a quaternized 
derivative of chitosan with superior aqueous solubility over a broader pH 
range and penetration - enhancing properties under physiological conditions [78] . 
Carboxymethylated chitosan (CMChi) is a polyampholytic polymer able to form 
viscoelastic gels in aqueous environments. CMChi appears to be less potent compared 
with the quaternized derivative. Neither TMC nor CMChi have been found 
to provoke damage of the cell membrane, and therefore, they should not alter the 
viability of nasal epithelial cells [79] . 
Thiolated chitosans, chitosan thioglycolic acid conjugates, chitosan – cysteine conjugates, 
and chitosan - 4 - thio - butyl - amidine conjugates are new - generation polymers 
that are pH sensitive. The pH range at which the gelation and mucoadhesion of 
these polymers are optimal is within the physiological range (pH 5 – 6.5) of the nasal 
mucosa, but these polymers have been primarily investigated for oral drug delivery 
[80] . 
5.7.4 MICROSPHERES AS NASAL DRUG DELIVERY DEVICES 
Developing an appropriate drug delivery system for a given drug can completely 
alter the drug ’ s unfavorable properties, such as improve its effectiveness or reduce 
its side effects. Dry powder delivery systems such as microspheres are of special 
interest. In the last two decades they have been extensively studied with respect to 
nasal delivery and a considerable number of studies have been reported on that 
subject [3, 23] . 
In general, microspheres as specialized drug delivery systems represent spherical 
polymeric devices that are small in size (from 1 to 1000 . m), are characterized by 
high surface - to - volume ratio, and are able to provide targeted and predictable controlled 
release of the drug [3] . In the scope of nasal delivery, except for controlled 
drug release rate, microspheres are benefi cial due to their broad surface area, which 
can provide extensive interaction with the mucin layer and protection of incorporated 
drug from enzymatic degradation in the nasal cavity [10, 41] . 

Microspheres prepared with bioadhesive polymers have some additional advantages; 
they assure much more intimate and prolonged contact with the mucous layer 
and improved drug absorption due to additional delay in mucociliary clearance. 
Bioadhesive microspheres can signifi cantly improve patient compliance as all 
the advantages described lead to reduction in the frequency of drug administration 
[3, 74] . 
Bioadhesive microspheres that have been extensively studied for nasal drug 
delivery are water insoluble but they swell in contact with the mucosa. Swollen 
microspheres form a gellike system that adheres onto the mucus, retaining drug at 
the absorption site for prolonged periods [2, 81] . Swelling of the microspheres causes 
mucosal dehydration and reversible shrinkage of the cells, resulting in the temporary 
widening of the tight junctions and increased permeability of hydrophilic 
compounds, or more precisely, paracellular absorption of the drug [62] . Oechslein 
et al. [40] suggested that the opening of tight junctions could be related to the local 
decrease in Ca 2+ concentration as well, since the absorption - promoting effect of 
investigated particulate drug delivery systems correlated directly with their capability 
to bind Ca 2+ . In order to additionally improve nasal drug absorption, bioadhesive 
particulate systems have been combined with biological absorption enhancers 
[55 – 57] . 
A number of studies of the nasal mucociliary clearance rate of microspheres 
confi rmed their potential to retain drug at the absorption site longer than liquid 
formulations [58, 67, 81, 82, 83] . Methods to measure formulation clearance rates 
from the nasal cavity can be divided into three groups, differing in the detected 
substance [58] . The most exact method is gamma scintigraphy, which monitors the 
deposition and clearance of radiolabeled drug delivery systems. The second method 
involves mixing of a fl uorescent dye with the formulation and monitoring the cumulative 
tracer amount in the pharynx. The third method is the saccharin test, in which 
saccharin is mixed with the formulation and the clearance rate is determined by the 
fi rst perception of sweet taste [84] . The fi rst study of the mucociliary clearance of 
microspheres using gamma scintigraphy was reported by Illum et al. [81] . They 
evaluated clearance rates of starch, dextran, and albumin microspheres: Three hours 
after nasal administration about 50% of albumin and starch microspheres and 60% 
of dextran microspheres were still detected at the site of deposition. Nasal clearance 
study of melatonin starch microspheres and the melatonin solution applied revealed 
that more than 80% of the starch microspheres remained in the nasal mucosa 2 h 
after administration, compared to only 30% for the melatonin solution [58] . The 
study of the clearance rate of alginate, PLGA, and Sephadex microspheres revealed 
that alginate and PLGA were suitable for nasal delivery as they had the best mucoadhesive 
properties [67] . It has also been shown that the limiting step of the mucociliary 
clearance of nasally administrated microspheres was their clearance from the 
initial deposition site. The same conclusion has been drawn for the Carbopol 971P 
and carboxymethyl cellulose microspheres [82] . Soane et al. [83] evaluated the clearance 
rate of chitosan microspheres and chitosan solutions, compared to the control 
solution from the nasal cavity in sheep, by gamma scintigraphy. They found that 
both chitosan systems had higher retention times compared to the control. Also, 
chitosan microspheres were cleared at a slower rate than the chitosan solution, with 
half times of clearance of 115 and 43 min, respectively. The nasal clearance rates 
found in the sheep model were similar to the clearance rates found in their previous 
MICROSPHERES AS NASAL DRUG DELIVERY DEVICES 659

660 NASAL POWDER DRUG DELIVERY 
study carried out on human subjects [85] , indicating that the sheep could be a suitable 
model for in vivo nasal clearance studies. 
Starch Microspheres Bioadhesive starch microspheres in the context of the nasal 
delivery system were fi rst introduced by Illum et al. [81] in a study that examined 
human nasal mucociliary clearance. Since then starch microspheres have been shown 
to promote the nasal absorption of a number of drugs. Animal studies using the sheep 
model showed greatly improved absorption of gentamicin [59] and human growth 
hormone [56] when administered in combination with starch microspheres in a 
freeze - dried formulation. Similar fi ndings have been reported for insulin [55, 60] and 
desmopressin [61] loaded starch microspheres compared to simple drug solutions 
administered to animal models. Biodegradable starch microspheres have also been 
investigated for nasal delivery of metoclopramide whereas enhanced bioavailability 
was achieved compared to nasal spray [86] . The bioadhesive starch microspheres 
were shown to act synergistically with the absorption enhancers improving the transport 
of insulin across the nasal membrane [57] . Recently, the potential of starch 
microspheres for the nasal delivery of melatonin was investigated [58] . An in vitro 
release study revealed a sustained drug release profi le. Melatonin bioavailability 
after nasal administration of starch microspheres was high, 84%. A good correlation 
between the in vitro release profi le and in vivo absorption has been observed. 
The use of degradable starch microspheres has proved to be well tolerated in 
both experimental animals and humans. No alterations of nasal mucosa were 
detected after eight weeks of nasal administration of starch microspheres to rabbits. 
Additionally, a preliminary test on healthy volunteers also showed good acceptability 
[62] . Another study of healthy volunteers revealed no changes in mucociliary 
clearance or in the geometry of the nasal cavities after eight days of nasal administration 
of dry starch microspheres [87] . 
Dextran Microspheres Similar to starch microspheres, Illum et al. [81] introduced 
dextran microspheres as a bioadhesive drug delivery system able to prolong the 
residence time in the nasal cavity. However, Ryd e n and Edman [65] reported dextran 
microspheres were not shown to improve nasal absorption of insulin in rats as 
insulin was too strongly bound to the Diethylaminoethyl (DEAE) groups to be 
released by a solution with an ionic strength corresponding to physiological conditions. 
In a later study it has been shown that the localization of insulin infl uenced 
the in vivo behavior of dextran microspheres [63] . The distribution of insulin at the 
surface or inside the dextran microspheres after the lyophilization loading process 
was determined by the cut - off limit of the microspheres. Microspheres with insulin 
left at the surface showed higher insulin absorption – enhancing effect than the 
microspheres with insulin inside the dextran matrix. Dextran microspheres have 
also been evaluated in vivo as a delivery system for octreotide [40] and in vitro for 
nicotine [33] . Ciliotoxicity studies performed in vitro on explants from rat trachea 
showed that dextran microspheres had no effect on the ciliary beat frequency [64] . 
The immediate recovery of the ciliary movement after dextran microspheres washing 
off indicated that the cilia were not damaged by dextran microspheres. 
Gelatin Microspheres Several studies characterizing gelatin microspheres as a 
nasal drug delivery system have been reported. Gelatin microspheres were shown 

to swell readily in contact with nasal mucosa and to have good bioadhesive properties 
[65] . An in vitro release study using a Franz diffusion cell on levodopa - loaded 
gelatin microspheres showed prolonged drug release as compared to drug alone 
[88] . Negatively and positively charged gelatin microspheres intended for nasal and 
intramuscular delivery of salmon calcitonin were prepared by Morimoto et al. [10] . 
Both types of microspheres enhanced nasal absorption of salmon calcitonin compared 
to the solution. Positively charged gelatin microspheres seamed to exhibit 
greater enhancing effect on nasal absorption than negatively charged gelatin microspheres. 
Recently gelatin and gelatin – poly(acrylic acid) microspheres were studied 
with respect to oral and nasal delivery of oxprenolol [89] . Combining the gelatin 
with poly(acrylic acid) resulted in microspheres with improved bioadhesive properties. 
Also, nasal administration of gelatin – poly(acrylic acid) microspheres resulted 
in improved bioavailability of the drug compared to nasal administration of the drug 
solution. 
Polyacrylate Microspheres Cross - linked polyacrylate microspheres as nasal 
powder delivery systems have been investigated in several studies [39, 41, 90] . Microspheres 
were produced by spray drying and emulsifi cation methods and their nasal 
drug delivery potential has been evaluated only in vitro. Carbopol 934P microspheres 
were shown to have the best bioadhesive properties compared to other 
hydrophilic microspheres prepared with polyvinyl alchohol, chitosan, and hydroxypropylmethyl 
cellulose [41] . Improved permeation - enhancing effect of polycarbophil 
microparticles was obtained when microparticles were prepared with the 
thiolated polycarbophil and the permeation mediator glutathione [68] . 
Chitosan Microspheres As a specifi c chitosan - based delivery system, chitosan 
microspheres have been extensively studied and number of reports has verifi ed their 
potential regarding nasal drug delivery [9, 21, 25, 41, 53, 73, 91, 92] . Chitosan microspheres 
have been prepared by the emulsifi cation solvent evaporation method [51, 
52, 93] , emulsifi cation cross - linking process [91] , spray drying method [25, 53] , and 
ionic gelation process [94] . They have been shown to signifi cantly reduce mucociliary 
clearance from the nasal cavity of sheep and humans compared to solutions [83, 
85] . The bioadhesive properties of chitosan microspheres were shown to be inversely 
proportional to particle size: Among chitosan microspheres in the size class between 
50 and 200 . m, smaller microspheres appeared to swell faster than large microspheres, 
providing a more powerful mucoadhesive system [41] . 
A modulated release rate of drug from the swellable chitosan microspheres has 
been achieved with cross - linking agents such as glutaraldehyde [95] , citric acid [92] , 
ascorbic acid, or ascorbyl palmitate [91] that reacted with chitosan forming covalent 
bonds with chitosan amino groups. However, to maintain the bioadhesive properties 
of cross - linked chitosan microspheres, the amount of cross - linking agent should be 
optimized [95] . 
Chitosan molecular weight has also been reported to infl uence drug release. Jiang 
et al. [94] studied Bordetella bronchiseptica dermonecrotoxin (BBD) release from 
chitosan microspheres prepared by tripolyphosphate ionic gelation. It has been 
shown that the BBD release rate increased with chitosan molecular weight decrease. 
It has been explained by the weaker BBD interaction with chitosan of lower molecular 
weight and lower content of free amine groups, responsible for their interaction. 
MICROSPHERES AS NASAL DRUG DELIVERY DEVICES 661

662 NASAL POWDER DRUG DELIVERY 
Chitosan microspheres were shown to enhance nasal bioavailability of several 
peptide drugs such as insulin and goserelin. A simple chitosan – insulin powder formulation 
provided about 20% of absolute insulin bioavailability in sheep [96] . 
Improved bioavailability (of 44%, in rats) was obtained when insulin was loaded 
into chitosan microspheres prepared with ascorbyl palmitate as cross - linking agent 
[91] . Chitosan microspheres have also been shown to improve nasal goserelin 
absorption providing about 40% bioavailability relative to goserelin intravenous 
application [9] . 
Krauland et al. [93] prepared the microparticles with thiolated chitosan (chitosan - 
TBA; chitosan – 4 - thiobutylamidine conjugate) intended for nasal peptide delivery. 
During the preparation process microparticles were stabilized by the formation of 
inter - and intramolecular cross - linking via disulfi de bonds. Chitosan – TBA microparticles 
were characterized by improved swelling ability and displayed 3.5 - fold higher 
insulin bioavailability compared to unmodifi ed chitosan microparticles. 
Besides the polymer derivatization, combining the polymers in microsphere 
preparations can result in improved drug delivery and absorption characteristics. 
Hyaluronic acid – chitosan microspheres appeared to improve the absorption of 
incorporated gentamicin compared to the individual polymers, assembling the 
mucoadhesive potential of both polymers and the penetration - enhancing effect of 
chitosan [51, 52] . 
Chitosan microparticulate systems have also been investigated for vaccine nasal 
delivery and have proven to induce strong systemic and mucosal immune responses 
[18, 21, 76] . 
5.7.4.1 Preparation Methods 
The design of bioadhesive microspheres includes selection of the most suitable 
preparation method, considering the nature of the drugs and polymers used as well 
as the route of administration. A number of methods for the preparation of microspheres 
have been described in the literature [3, 97] . In the scope of nasal delivery, 
the fi rst microspheres in use were starch and dextran microspheres, prepared by an 
emulsion polymerization technique employing epichlorohydrine as a cross - linking 
agent [55, 56, 59, 60, 62, 81] . Currently techniques based on solvent removal, such 
as solvent evaporation [41, 51, 66, 93, 98] and solvent extraction [88, 99] , are most 
frequently in use. There are tree processes involved in such microensapsulation 
procedures: the preparation of emulsion, solvent removal, and separation of the 
particles obtained. Selection of the type of (oil - and - water) emulsion system (O/W, 
W/O, W/O/W, W/O/O, etc.) depends on the physicochemical properties of the drug 
and polymer used. After the preparation of stable emulsion, solvent is removed from 
the system at high or low temperature, at low pressure, or by addition of another 
solvent that enables the extraction of polymer solvent to the continuous phase. 
Hardened microspheres are then washed, centrifuged, and lyophilized. 
Emulsion techniques are suitable for the preparation of microspheres intended 
for nasal delivery since they allow controlling the size of the particles. Freiberg and 
Zhu [97] reviewed solvent evaporation process parameters (e.g., polymer concentration, 
viscosity, stirring rate, temperature and percentage of emulsifying agent) affecting 
microsphere size. It can be assumed that the particle size is directly proportional 
to polymer concentration and inversely proportional to stirring rate and percentage 
of emulsifying agent [41, 88] , while there is a nonlinear correlation between particle 

size and process temperature. Yang et al. [100] reported that larger microspheres 
were produced at lower temperatures due to the higher viscosity of solution and at 
higher temperatures due to the higher solvent fl ow pressure moving more material 
from the microsphere center outward. In the same work encapsulation effi ciency 
was also correlated with the temperature of solvent evaporation in the process of 
microsphere preparation. It was found that the highest encapsulation effi ciencies 
occurred at the lowest and highest temperatures tested. 
Recently, the spray drying method has been extensively used for the preparation 
of microspheres intended for nasal delivery [3, 25, 39, 44, 45, 53, 54, 101, 102] . Spray 
drying is a single - step procedure transforming liquid into dry particulate form (e.g., 
microparticles, microspheres, microcapsules) applicable to drugs and polymers of 
various solubility characteristics. It is a fast, simple, and reproducible technique that 
is easy to scale up [3] . Spray drying can be described as follows: The liquid is fed to the 
nozzle with a peristaltic pump, atomized by the force of the compressed air, and blown 
together with hot air into the chamber, where the solvent in the droplets is evaporated. 
The dry product is then collected in a collection bottle. Spray - dried microspheres 
are reported to have relatively low production yields, rarely higher than 50%. 
The loss of material during spay drying has been explained by the powder adhering 
to the cyclone walls, small amounts of materials processed in each batch, and loss of 
the smallest and lightest particles through the exhaust of the spray dryer apparatus, 
which lacks a trap to recover the lighter and smaller particles [25, 53, 103, 104] . 
Spray drying offers the possibility to control the particle size of the product. 
Microparticles of desirable size can be obtained by optimizing the spray drying 
process parameters, such as size of the nozzle, feeding pump rate, inlet temperature, 
and compressed airfl ow rate. In accordance with this, He et al. [105] reported that 
larger particles were formed at a larger size of nozzle and faster feeding pump rate, 
while smaller particles were formed at a greater volume of air input [105] . The size 
and other properties of spray - dried microspheres (e.g., morphology, density, shape, 
porosity, and fl owability) can also be affected by the qualitative and quantitative 
composition of the liquid feed [28] . Thus, feed concentration has been reported to 
infl uence the particle size distribution, as spray drying of more concentrated liquid 
feeds resulted in the formation of larger particles [104, 106, 107] . 
5.7.4.2 Microsphere Characterization 
Microspheres intended for nasal administration need to be well characterized in 
terms of particle size distribution, since intranasal deposition of powder delivery 
systems is mostly determined by their aerodynamic properties and particle sizes. 
Commonly used methods for particle size determinations described in the literature 
are sieving methods [108] , light microscopy [58] , photon correlation spectroscopy 
[66] , and laser diffractometry [25, 41, 53, 93] . The morphology of the microparticles 
(shape and surface) has been evaluated by optical, scanning, and transmission electron 
microscopy [66, 95] . 
Determination of the zeta potential is an important part of microsphere characterization, 
as the zeta potential has a substantial infl uence on the adhesion of drug delivery 
systems onto biological surfaces [109] . For example, Jaganathan and Vyas [110] 
reported the reduction in the nasal clearance rate of PLGA microspheres modifi ed 
with chitosan compared to unmodifi ed PLGA microspheres due to the change in zeta 
potential from negative for PLGA microspheres to positive for surface - modifi ed 
MICROSPHERES AS NASAL DRUG DELIVERY DEVICES 663

664 NASAL POWDER DRUG DELIVERY 
PLGA microspheres. Methods to measure the zeta potential of microspheres are 
laser doppler anemometry [105] and photon correlation spectroscopy [110] . 
The physical state of the drug incorporated in a powder drug delivery system 
(e.g., degree of crystallinity and possible interactions with the polymer) is assessed 
by differential scanning calorimetry (DSC) or Fourier transform infrared (FTIR) 
spectroscopy. These observations can clarify the results of other parameter investigations, 
especially the results of in vitro drug release studies. 
To predict microsphere performance in vivo, the swelling properties of nasal 
powder delivery systems need to be evaluated. Methods described in the literature 
are mostly based on the weight difference measurements between the dry and 
swollen powder [40] . Swelling properties of nasal powders such as water - absorbing 
capacity can be evaluated using a Franz diffusion cell [43, 107] . The swelling capacity 
may also be expressed as the volume expansion of the microspheres that is determined 
at equilibrium after placing the microspheres in water using a graduated 
cylinder [108] . Gavini et al. [53] determined the swelling properties of microspheres 
in vitro by laser diffractometry. That method allows us to evaluate the variation of 
particle size versus time. 
In vitro evaluation of mucoadhesive properties is essential in the development of 
a nasal drug powder delivery system, since mucoadhesion is of great importance for 
the in vivo performance of formulation. A large number of in vitro and in vivo 
methods used to assess mucoadhesive properties of microspheres have been extensively 
described in the literature [3, 111, 112] . Many in vitro methods are based on 
the interaction of microspheres with mucin. Evaluation of that interaction can be 
performed using scanning and transmission electron microscopy [95] or photon correlation 
spectroscopy [45] . Scanning electron microscopy (SEM) provides the information 
on morphological changes on the microsphere surface in contact with mucin, 
while transmission electron microscopy confi rms SEM results and reveals the ultrastructural 
features of the surface interactions between microspheres and mucin 
chains [95] . He et al. [102] evaluated the mucoadhesive properties of chitosan microspheres 
by measuring the amount of mucin adsorbed on the microspheres. Gavini et 
al. [53] evaluated the mucoadhesive properties of metoclopramide - loaded microspheres 
by determining the amount of microspheres that stuck to a fi lter paper saturated 
with mucin after exposure to the air stream. Vidgren et al. [39] used a tensiometer 
to measure the force required for the separation of two fi lter paper discs saturated 
with mucin and with the examined microspheres placed between them. 
In the work reported by Witschi and Mrsny [54] mucoadhesion of dry powder 
microparticles was investigated using Callu - 3 cells as a surrogate for human nasal 
epithelia: Microparticles were applied to the apical surface of cell sheets and at certain 
time points were washed with phosphate - buffered saline (PBS) to remove poorly 
adhering microparticles. Rango Rao and Buri [113] developed an in situ method to 
evaluate the bioadhesive properties of polymers and microparticles, based on washing 
off a mucous membrane covered with the formulation to be tested by simulated biological 
fl ow. The mucoadhesion of gelatin microspheres [10] was measured by an in 
situ nasal perfusion experiment. In the work reported by Lim et al. [51] the mucoadhesive 
properties of microspheres were evaluated by determining the mucociliary 
transport rate of the microparticles across an isolated frog palate. The boiadhesive 
properties of microspheres can be evaluated by the everted sac technique using a 
section of everted intestinal tissue or the CAHN dynamic contact angle analyzer [3] . 

Santos et al. [112] correlated these two methods and concluded that each method 
could be used alone as the relevant indicator of microsphere bioadhesion. 
In vitro drug release experiments can be performed in order to characterize the 
release behavior of microparticles in general. For that purpose microparticles can 
be dispersed directly in the dissolution medium [51, 91] or a dynamic dialysis technique 
can be employed [58] . However, to obtain results comparable with the in vivo 
situation of nasal administration, it is necessary to provide experimental conditions 
similar to those encountered in the nasal cavity as nasally administrated powders 
are not being dispersed directly in the large quantity of liquid [8] . Such in vitro drug 
release experiments can be performed by a modifi ed U.S. Pharmacopeia (USP) 
XXII rotating basket [8] . The drug - loaded powder formulation is weighed on a 
membrane fi lter placed between the fi lter holder and the cup and then immersed 
in the released medium. Thus, the membrane fi lter separates the donor and acceptor 
compartment but at the same time allows the powders to hydrate and to form a gel. 
Drug is released to the release medium after diffusion through the swollen gel of 
known surface area. Cornaz et al. [33] developed a special diffusion chamber that 
simulated the hydration conditions of the nasal mucosa to study the in vitro release 
of nicotine from dextran microspheres. A number of authors have used Franz diffusion 
cells for in vitro release experiments since that model provides conditions 
similar to those encountered in the nasal cavity and slow hydration of the microspheres 
[41, 43 – 45, 107] . 
5.7.4.3 Chitosan - Formulated Spray - Dried Microspheres 
Chitosan, a biocompatible and biodegradable polycationic polymer with low toxicity, 
is known for its swelling ability and permeation - enhancing properties and represents 
a polymer of choice for the preparation of microspheres intended for nasal 
administration [74] . 
Spray drying has proved to be a suitable and simple technique for the preparation 
of chitosan microspheres with preserved chitosan properties, offering numerous 
advantages over other microencapsulation methods. It has been successfully used 
to entrap both hydrophilic and lipophilic drugs into the chitosan matrix, since a 
variety of colloidal systems (e.g., polymer solutions, emulsions, dispersions, suspensions) 
can be subjected to spray drying. Chitosan - based spray - dried microspheres 
have been prepared with chitosan alone (resulting in conventional microspheres) 
or in combination with another polymer (resulting in composed microspheres). 
Combining the polymers has been reported to result in microspheres with improved 
properties regarding surface characteristics, entrapment effi ciency, or control over 
the drug release rate. Recently, several drugs, such as carbamazepine [25] , propranolol 
hydrochloride [45] , metoclopramide hydrochloride [53] , and loratadine [103, 
107] , have been incorporated into chitosan - based nasal powder formulations produced 
by the spray drying - method. They were characterized in terms of encapsulation 
effi ciency, morphology, size distribution, zeta potential, physical state of the 
drug, in vitro drug release behavior, and swelling and bioadhesive properties. 
Chitosan – ethyl cellulose composed microspheres improved loratadine entrapment 
compared to conventional chitosan microspheres, which infl uenced directly the 
microsphere surface characteristics: Loratadine was less present at the surface of 
the microspheres and consequently had less infl uence on their bioadhesive proper- 
MICROSPHERES AS NASAL DRUG DELIVERY DEVICES 665

666 NASAL POWDER DRUG DELIVERY 
ties. Thus, although showing moderate swelling ability, loratadine - loaded composed 
microspheres were more bioadhesive than conventional chitosan microspheres. 
Composed microspheres showed good loratadine - sustained release potential in 
vitro, depending on the polymeric weight ratio and concentration of the spray - dried 
system [103, 107] . Gavini et al. [53] produced metoclopramide - loaded alginate and/ 
or chitosan microspheres by the spray drying method. The results obtained revealed 
that complexation of chitosan with alginate in the microsphere preparation provided 
improved control of the drug release in vitro compared to chitosan alone. 
Despite the chitosan complexation, composed microspheres showed good bioadhesive 
properties. Ex vivo drug permeation tests carried out using sheep nasal mucosa 
showed higher drug permeation from chitosan - based microspheres than from alginate 
microspheres, confi rming the well - known chitosan permeation - enhancing 
properties. Cerchiara et al. [45] developed spray - dried chitosan – poly(methyl vinyl 
ether - co - maleic anhydride) microparticles for nasal delivery of propranolol hydrochloride. 
Chitosan was combined with polyanhydride, able to enhance the formation 
of hydrogen bonds between the polymers and mucosal components through carboxylic 
acid groups generated after polyanhydride hydrolytical degradation. The 
swelling and bioadhesive properties of chitosan – polyanhydride microparticles 
increased in a pH - dependent manner. Both chitosan and chitosan – polyanhydride 
microparticles provided sustained propranolol hydrochloride release. 
Microparticulate spray - dried delivery systems have shown great potential for 
nasal delivery of drugs characterized by poor water solubility. According to DSC 
analysis, the spray drying method together with the carriers seemed to promote the 
amorphization of loratadine [107] and carbamazepine [25] . Carbamazepine incorporated 
into chitosan microspheres was characterized by increased dissolution rate 
compared to carbamazepine raw material. It was explained not only by the promoted 
drug amorphization but also by the chitosan well - known dissolution rate 
enhancer properties and by the small size of microspheres (or high surface - to - 
volume ratio). Results of in vivo nasal administration of carbamazepine - loaded 
chitosan microspheres revealed a remarkable increase in carbamazepine concentration 
in serum compared to the pure carbamazepine powder [25] . Such an increase 
in drug absorption has been ascribed to both improved dissolution of carbamazepine 
and adhesion of the chitosan microspheres to the mucosal surface. 
The mucoadhesive function of chitosan has also been employed in vaccine dry 
powder delivery. Alpar et al. [18] produced bovine serum albumin (BSA) – loaded 
chitosan microspheres using the spray drying method. It has been shown that the 
stability of encapsulated BSA was preserved in the microspheres prepared, indicating 
that spray drying was appropriate even for the preparation of antigen - loaded 
microspheres. BSA - loaded chitosan microspheres generated higher immune response 
than the free BSA, thus proving to be a suitable system for nasal antigen delivery. 
5.7.5 TOXICOLOGICAL CONSIDERATIONS 
The possible toxicological effects of dry powder formulation on the nasal mucosa, 
including local irritation, effect on mucociliary clearance, and epithelial damage and 
recovery rate [114] , should be investigated in an early stage of its development. 
There were some attempts to defi ne the categories of toxic effects as well of the 
constituents of nasal formulations according to their toxic potential. Thus, Hvidberg 

et al. [115] introduced a scale of irritation in the nose (0, no irritation; 1, slight irritation; 
2, acceptable; 3, unwilling to accept the treatment again) that was later used to 
evaluate the degree of nasal powder irritation after administration in human volunteers 
[27] . Ugwoke et al. [82] classifi ed the degree of ciliary beat frequency change 
caused by nasally applied liquid formulation as follows: no effect (less than 10%), 
mild (10 – 20%), moderate (20 – 50%), and severe (more than 50%). Soon after, that 
classifi cation was applied to a powder ciliotoxicity study [116] . Reversibility of ciliotoxic 
effect after washing out the tested compound was classifi ed as well, resulting 
in three categories: reversible, partially reversible, and irreversible effects [82] . 
Merkus et al. [117] proposed the three categories of constituents of nasal formulations 
based on the recovery of ciliary beat frequency after the tested compound was 
washed out . The fi rst category is cilio friendly, with ciliary beat frequency recovery 
of 75% or more; the second is cilio inhibiting, with recovery between 25 and 75%; 
and the third category is ciliostatic, with recovery of 25% or less. 
Methods to evaluate the possible toxicological effect of formulations on the nasal 
mucosa described in the literature mainly refer to histopathological evaluation as a 
standard method for cytotoxicity evaluation [64] and a study on the release of marker 
enzymes [118] . In the work reported by Callens et al. [118] the possible toxicological 
effects of multiple starch and carbopol powder nasal administration were evaluated 
using rabbits by measuring the proteins and lactate dehyrogenase (LDH) release 
from the nasal mucosa. Contrary to the invasive in situ perfusion method, performable 
with anaesthetized animals [119] , this method has been shown to be noninvasive, 
applicable to nonanaesthetized and nonsedated animals, and suitable for repeated 
measurement of the marker protein release on the same animal during a long - term 
administration study. A histopathological study has also been performed. In agreement 
with attempting to replace the use of vertebrates in scientifi c experiments with 
lower organisms such as invertebrates, plants, and microorganisms, Adriaens and 
Remon [120] developed a new mucosal toxicity screening method using the slug 
Arion lusitanicus as the model organism. The body wall of the slug resembles the nasal 
mucosa since it consists of a single - layered epithelium containing both ciliated and 
mucus - secreting cells. Callens et al. [118] successfully used that method for screening 
the mucosal irritation potential of bioadhesive starch and carbopol powder formulations. 
The possible toxicological effects of the powder formulations were evaluated 
by measuring the proteins LDH and alkaline phosphatase (ALP) release from the 
body wall of the slugs as well as the amount of mucus produced. 
Among various nasal toxicity studies, ciliotoxicity studies are of special interest 
due to the importance of maintaining optimal ciliary beating to protect the lower 
respiratory system from infections. If the drug formulation inhibits the ciliary beating, 
such inhibition needs to be completely reversible upon formulation removal [116] . 
Methods to determine ciliary beat frequency and mucociliary transport in vitro 
and in vivo have been extensively reviewed elsewhere [84, 121] . In general, in vivo 
methods are more reliable for ciliotoxicity studies than in vitro methods and are 
essential to confi rm the safety of nasal drug formulations. However, in vitro methods 
are more suitable for the large number of screening studies required during formulation 
development [122] . 
In vitro methods to measure ciliary beat frequency can be performed on explants 
of ciliated mucosa [64] or on different types of ciliated cells, such as ciliated chicken 
embryo trachea cells [123] , nasal cell lines derived from carcinomas of epithelial 
origin (RPMI 2650, BT, NAS 2BL), or human lung adenocarcinoma cell line Calu - 3 
TOXICOLOGICAL CONSIDERATIONS 667

668 NASAL POWDER DRUG DELIVERY 
[124] . In vitro ciliotoxicity of dextran microspheres was evaluated on explants from 
rat trachea [64] . 
Human nasal epithelial cell cultures can serve as a relevant screening tool for 
prediction of nasal formulation toxicity in humans as long as the cells maintain differentiated 
morphological and biochemical characteristics of the original tissue 
[124] . Epithelial cells intended for initiation of primary cell culture should be taken 
from the regions of the nasal cavity where formulations are supposed to be deposited 
[125] . Considering toxicological investigations, human nasal epithelial cells 
cultured in an air – liquid interface system are the most promising ones at the moment, 
as in these culturing conditions cell differentiation closely resembles cell differentiation 
in vivo [124] . Monolayer immersion feeding and air – liquid interface cultures 
have already been used for ciliary beat frequency measurements. However, unstable 
ciliary activity and their short life span in the culture have proved to be the main 
shortcomings of these systems. Jorissen et al. [126] developed a suspension culture 
system of human nasal epithelium with actively beating cilia for several months. 
That system has been validated for ciliotoxicity investigations [122] and later applied 
in other studies [116, 127] . At this time it is the most successful model of human 
nasal epithelium culture useful for ciliary beat frequency determination [124] . 
In vivo methods to determine the toxicological effects of drug formulations on 
the nasal mucosa mainly refer to mucociliary clearance rate studies. Methods 
described in the literature are mostly based on the gamma scintigraphic technique, 
in which cleared formulations are labeled with the radiotracer [82] . In vivo scintigraphic 
evaluation of the nasal clearance of drug delivery systems requires a nondiffusible 
and stable radiotracer to prevent its absorption and decomposition. Another 
method described in the literature is the saccharin test [87] , in which saccharin is 
mixed with the formulation and the clearance rate is determined by the fi rst perception 
of sweet taste [84] . Gamma scintigraphy [31] is more relevant for the ciliary 
function monitoring than the saccharin test, since it investigates the whole mucosal 
surface, while the saccharin test investigates only the fastest fl ow rate [82] . 
A number of ciliotoxicity studies have pointed out a low correlation between the 
results obtained using different in vitro and in vivo methods [121] . The effects of 
nasal formulations on the ciliary beat frequency in vitro are usually more expressed 
than in vivo, since in vivo, cilia are partially protected by the mucous layer and 
investigated formulation is eventually cleared from the nasal cavity due to the 
mucociliary clearance mechanism. Also, toxic effects of the formulations on the cilia 
in vivo may be reversible due to the constant nasal mucosal cell turnover [121] . 
In conclusion, in order to make predictions regarding the safety of the nasal formulation 
on mucociliary clearance, both in vitro and in vivo studies have to be performed. 
It is also essential to determine long - term use effects in animals and 
in humans if the nasal formulation is intended for subchronic or chronic 
administration. 
Dry powder formulations for nasal delivery of peptides and proteins have been 
investigated for the fi rst time by Nagai and others [38] . Since then, much research 
work has been done on dry powders containing bioadhesive polymers for nasal drug 
administration. The bioavailability and duration of action of drugs administered by 
the nasal route are increased by the use of the principle of mucoadhesion and dry 
powder formulations. Research work on dry powder formulation containing bioadhesive 
polymers is summarized in Table 1 . 

TABLE 1 Summary of Research Work on Nasal Dry Powder Formulations 
Powder 
Formulation Preparation Method Drug Polymer Studies Comments Reference 
Microparticles Spray drying BSA Starch, 
alginate, 
chitosan, carbopol 
In vitro Chitosan microparticles 
provided most desirable 
characteristics for protein 
delivery 
54 
Microparticles 
Budesonide Polymethacrylic 
acid – polyethylene 
glycol 
[P(MAA – PEG)] 
In rabbits Continuous drug release for 
at least 8 h 
High bioavailability 
133 
Microparticles Solvent evaporation 
Gentamicin Hyaluronic 
acid – chitosan 
In rabbits Polymers improved 
gentamicin absorption 
synergistically 
52 
Microparticles Lyophilization hGH Polycarbophil – 
cysteine 
In rats Improved bioavailability 
68 
Microparticles W/O emulsifi cation 
solvent 
evaporation 
Insulin Chitosan – TBA In rats Improved bioavailability 
93 
Microparticles Spray drying Propranolol 
hydrochloride 
CH - PVM/MA In vitro Sustained drug release 45 
Microparticles, 
nanoparticles 
W/O/W solvent 
evaporation 
Model protein, 
tetanus toxoid 
PLA - PEG In rats Size - dependent mucosal 
uptake 
66 
Microspheres Ionic gelation with 
tripolyphosphate 
Bordetella, 
bronchiseptica, 
dermonecrotoxin 
Chitosan In mice Systemic and mucosal 
immune responses induced 
21 
Microspheres Ionic gelation 
process with 
tripolyphosphate 
Bordetella, 
bronchiseptica, 
dermonecrotoxin 
Chitosan In vitro Chitosan molecular weight – 
related drug release profi le 
94 
Microspheres Spray drying Carbamazepine Chitosan 
hydrochloride, 
chitosan 
glutamate 
In sheep Increased drug dissolution 
rate and absorption 
25 
669

Powder 
Formulation Preparation Method Drug Polymer Studies Comments Reference 
Microspheres W/O/O emulsion 
solvent 
evaporation 
. - Cobrotoxin PLGA/P(CPP: 
CEFB) 
In rats Increased strength and 
duration of antinociceptive 
effect 
98 
Microspheres Emulsion 
polymerization 
Desmopressin Starch In rats, 
in 
sheep 
Increased bioavailability with 
LPC 
61 
Microspheres W/O emulsifi cation 
solvent 
evaporation 
FITC – dextran Carbopol 394P 
Chitosan 
HPMC 
PVA 
In vitro Initial release at a constant 
rate 
Chitosan microspheres 
exhibited size - dependent 
release effect 
41 
Microspheres 
FITC – dextran Chitosan, 
Carbopol 
934P 
In rabbits Improved bioavailability 131 
Microspheres Cross - linking with 
epichlorohydrin 
Gabexate mesylate Starch cyclodextrin In vitro Fast release rate 
108 
Microspheres Solvent evaporation Gentamicin Chitosan, 
hyaluronan, 
gelatine 
In vitro Prolonged release, 
improved 
mucoadhesive properties 
51 
Microspheres Emulsion 
polymerization 
Gentamicin Starch In sheep, 
in 
rats 
Improved bioavailability with 
microsphere/enhancer 
(LPC) system 
59 
Microspheres Spray drying Gentamicin sulfate HPMC In vitro Modifi 
ed drug release 
44 
Microspheres Solvent evaporation Heparin Poly(lactic acid) 
In rats Sustained - release effect 134 
Microspheres Emulsion 
polymerization 
hGH Starch In sheep Enhanced nasal absorption 
with microspheres, rapid 
and higher absorption with 
microsphere/enhancer 
(LPC) system 
56 
Microspheres Emulsion 
polymerization 
Insulin Starch, 
dextran In rats Rapid absorption 
62 
Microspheres Emulsion 
polymerization 
Insulin Starch In sheep Improved bioavailability with 
microsphere/enhancer 
(LPC) system 
55 
TABLE 1 Continued 670

Powder 
Formulation Preparation Method Drug Polymer Studies Comments Reference 
Microspheres Emulsion 
polymerization 
Insulin Starch In rats Improved nasal absorption 
60 
Microspheres Emulsion 
polymerization 
Insulin Dextran In rats Promoted absorption 63, 
65 
Microspheres Emulsifi 
cation – cross - 
linking 
Insulin Chitosan In rats Promising absolute 
bioavailability of 44% 
91 
Microspheres Emulsifi cation – 
solvent 
evaporation 
Insulin Hyaluronic acid 
ester 
In sheep Increased nasal absorption 
128 
Microspheres W/O emulsifi cation 
solvent extraction 
Levodopa Gelatin In vitro Initial fast release rate, 
followed by a second 
slower release rate 
88 
Microspheres Spray drying Loratadine Chitosan, 
chitosan/ 
ethylcellulose 
In vitro Moderate swelling behavior, 
sustained drug release 
107 
Microspheres Emulsifi 
cation – cross - 
linking 
Melatonin Starch In rabbits Increased residence time, 
rapid absorption rate, high 
absolute bioavailability 
58 
Microspheres Spray drying Metoclopramide Sodium alginate, 
chitosan, sodium 
alginate/chitosan 
In vitro Controlled drug release, 
promising properties as 
nasal drug carriers 
53 
Microspheres Emulsifi 
cation – cross - 
linking 
Metoprolol tartrate 
Alginate In rabbits Improved therapeutic 
effi cacy 
132 
Microspheres W/O emulsifi 
cation Morphine Chitosan, 
starch In sheep High bioavailability 
73 
Microspheres Emulsion 
polymerization 
Nicotine Dextran In vitro Rapid release, 
good 
dispersion ability 
33 
Microspheres W/O emulsifi cation 
solvent extraction 
Oxprenolol Gelatin – poly(acrylic 
acid) 
In rats Slow - release drug delivery 
system with good adhesive 
characteristics 
89 
Microspheres W/O emulsion 
cross - linking 
Pentazocine Chitosan In rabbits Matrix diffusion controlled 
delivery, improved 
bioavailability, in vitro/in 
vivo correlation 
129 
Microspheres Emulsion solvent 
evaporation 
Salbutamol Chitosan In rabbits Prolonged and controlled 
release 
92 
671

Powder 
Formulation Preparation Method Drug Polymer Studies Comments Reference 
Microspheres W/O emulsion 
cross - linking 
Salmon calcitonin Gelatin In rats Enhanced nasal absorption 
10 
Microsphers Emulsion 
polymerization 
Insulin Starch In sheep Microspheres and absorption 
enhancers (LPC, GDC, 
and STDHF) acted 
synergistically to enhance 
absorption 
57 
Powder Lyophilization Apomorphine Carbopol 971P, 
polycarbophil 
In rabbits Sustained release, 
improved 
bioavailability 
8 
Powder Lyophilization Apomorphine Carboxymethyl 
cellulose 
In rabbits Sustained plasma level 42 
Powder Lyophilization Apomorphine Carbopol 971P, 
carboxymethyl 
cellulose 
In rabbits Increased residence time 
48 
Powder Lyophilization Apomorphine HCl Carbopol 971P, 
Carbopol 974P, 
polycarbophil 
In rabbits In vitro release but not in 
vivo absorption has been 
infl uenced by drug loading 
130 
Powder Spray drying Cyanocobalamin Microcrystalline 
cellulose, dextran, 
crospovidone 
In rabbits Improved bioavailability 43 
Powder Press - on force 
method 
Glucagon Microcrystalline 
cellulose 
In human 
volunteers 
Increased formulation 
stability, decreased 
irritability 
27 
Powder — Goserelin Chitosan In sheep Improved bioavailability 
9 
Powder Material mixing with 
a pestle 
Insulin Chitosan In rats, 
in 
sheep 
Improved bioavailability 
11 
Powder Blending/ 
lyophilization 
Insulin Microcrystalline 
cellulose, 
hydroxypropyl 
cellulose, 
Carbopo 934 
In dogs Decreased plasma glucose 
level 
38 
TABLE 1 Continued 672

Powder 
Formulation Preparation Method Drug Polymer Studies Comments Reference 
Powder Lyophilization Insulin Starch – Carbopol 
974P, 
maltodextrin – 
Carbopol 974P 
In rabbits The highest absolute 
bioavailability obtained 
was 14.4% 
46 
Powder Lyophilization Ketorolac Microcrystalline 
cellulose 
In rabbits Signifi cantly lower 
bioavailability of drug 
from powders compared 
to spray formulation 
50 
Powder — Leuprolide, 
calcitonin, 
FITC – dextran 
Hydroxypropyl 
cellulose, 
microcrystalline 
cellulose 
In rabbits Enhanced absorption 49 
Powder Manual blending 
using mortar and 
pestle 
Morphine Chitosan In human 
volunteers 
Rapid onset of pain relief, 
formulations well tolerated 
by patients 
73 
Powder Dry blending Octreotide Dextran, 
microcrystalline 
cellulose, 
semicrystalline 
cellulose, 
hydoxyethyl 
starch, 
microcrystalline 
chitosan, pectin, 
alginic acid 
In rats Correlation between carrier 
calcium binding properties 
and their potential as nasal 
absorption enhancers for 
peptides 
40 
Powder, 
granules 
Powder: 
manual 
mixing using 
mortar and pestle 
Recombinant hGH Chitosan In sheep Relative bioavailability of 
hGH from powder and 
granules have been 14 and 
15%, respectively 
77 
Abbreviations: 
PLGA: 
poly(lactide - co - glycolide); 
P(CPP:CEFB): 
poly[1,3 - bis(p - carboxy - phenoxy) propane - co - p - (carboxyethylformamido) benzoic anhydride]; 
chitosan – TBA: chitosan – 4 - thiobutylamidine conjugate; LPC: lysophosphatidyl choline; hGH: human growth hormone; GDC: glycodeoxycholate sodium; STDHF: 
sodium taurodihydroxyfusidate 
673

674 NASAL POWDER DRUG DELIVERY 
REFERENCES 
1. Quraishi , M. S. , Jones , N. S. , and Tomason , J. D. T. ( 1997 ), The nasal delivery of drugs , 
Clin. Otolaryngol. , 22 , 289 – 301 . 
2. Pereswetoff - Morath , L. ( 1998 ), Microspheres as nasal drug delivery systems , Adv. Drug 
Deliv. Rev. , 29 , 185 – 194 . 
3. Vasir , J. K. , Tambwekar , K. , and Garg , S. ( 2003 ), Bioadhesive microspheres as a controlled 
drug delivery system , Int. J. Pharm. , 255 , 13 – 32 . 
4. Tirucherai , G. S. , Pezron , I. , and Mitra , A. K. ( 2002 ), Novel approaches to nasal delivery 
of peptides and proteins , S. T. P. Pharm. Sci. , 12 , 3 – 12 . 
5. Davis , S. S. ( 1999 ), Delivery of peptide and non - peptide drugs through the respiratory 
tract , Pharm. Sci. Technol. Today , 2 , 450 – 456 . 
6. Resta , O. , Barbaro , M. , and Carnimeo , N. ( 1992 ), A comparison of sodium cromoglycate 
nasal solution and powder in the treatment of allergic rhinitis , Br. J. Clin. Practice , 46 , 
94 – 98 . 
7. Schipper , N. , Romeijn , S. , Verhoef , J. , and Merkus , F. ( 1993 ), Nasal insulin delivery with 
dimethyl beta cyclodextrin as an absorption enhancer in rabbits: Powder more effective 
than liquid formulations , Pharm. Res. , 10 , 682 – 686 . 
8. Ugwoke , M. I. , Exaud , S. , Van Den Mooter , G. , Verbeke , N. , and Kinget , R. ( 1999 ), Bioavailability 
of apomorphine following intranasal administration of mucoadhesive drug 
delivery systems in rabbits , Eur. J. Pharm. Sci. , 9 , 213 – 219 . 
9. Illum , L. , Watts , P. , Fisher , A. N. , Jabbal - Gill , I. , and Davis , S. S. ( 2000 ), Novel chitosan - 
based delivery systems for the nasal administration of a LHRH - analogue , S. T. P. Pharm. 
Sci. , 10 , 89 – 94 . 
10. Morimoto , K. , Katsumata , H. , Yabut , T. , Iwanaga , K. , Kakemi , M. , Tabata , Y. , and Ikada , 
Y. ( 2001 ), Evaluation of gelatin microspheres for nasal and intramuscular administrations 
of salmon calcitonin , Eur. J. Pharm. Sci. , 13 , 179 – 185 . 
11. Dyer , A. M. , Hinchcliffe , M. , Watts , P. , Castile , J. , Jabbal - Gill , I. , Nankervis , R. , Smith , A. , 
and Illum , L. ( 2002 ), Nasal delivery of insulin using novel chitosan based formulations: 
A comparative study in two animal models between simple chitosan formulations and 
chitosan nanoparticles , Pharm. Res. , 19 , 998 – 1008 . 
12. Colombo , P. ( 1999 ), Mucosal drug delivery, nasal , in Mathiowitz , E. , Ed., Encyclopedia 
of Controlled Drug Delivery , J Wiley , New York , pp. 593 – 605 . 
13. Ishikawa , F. , Murano , M. , Hiraishi , M. , Yamaguchi , T. , Tamai , I. , and Tsuji , A. ( 2002 ), 
Insoluble powder formulation as an effective nasal drug delivery system , Pharm. Res. , 
19 , 1097 – 1104 . 
14. Ishikawa , F. , Katsura , M. , Tamai , I. , and Tsuji , A. ( 2001 ), Improved nasal bioavailability 
of elcatonin by insoluble powder formulation , Int. J. Pharm. , 224 , 105 – 114 . 
15. Alpar , H. O. , Eyles , J. E. , Williamson , E. D. , and Somavarapu , S. ( 2001 ), Intranasal vaccination 
against plaque, tetanus and diphteria , Adv. Drug Deliv. Rev. , 51 , 173 – 201 . 
16. Vajdy , M. , and O ’ Hagan , D. T. ( 2001 ), Microparticles for intranasal immunisation , Adv. 
Drug Deliv. Rev. , 51 , 127 – 141 . 
17. Davis , S. S. (2005), The use of soluble polymers and polymer microparticles to provide 
improved vaccine responses after parenteral and mucosal delivery, Vaccine , 24 , Suppl. 
2, S7 – S10 . 
18. Alpar , H. O. , Somavarapu , S. , Atuah , K. N. , and Bramwell , V. W. ( 2005 ), Biodegradable 
mucoadhesive particulates for nasal and pulmonary antigen and DNA delivery , Adv. 
Drug Deliv. Rev. , 57 , 411 – 430 . 

REFERENCES 675 
19. Van der Lubben , I. M. , Verhoef , J. C. , Borchard , G. , and Junginger , H. E. ( 2001 ), Chitosan 
and its derivatives in mucosal drug and vaccine delivery , Eur. J. Pharm. Sci. , 14 , 201 – 
207 . 
20. Van der Lubben , I. M. , Kersten , G. , Fretz , M. M. , Beuvery , C. , Verhoef , J. C. , and 
Junginger , H. E. ( 2003 ), Chitosan microparticles for mucosal vaccination against diphteria: 
Oral and nasal effi cacy studies in mice , Vaccine , 21 , 1400 – 1408 . 
21. Kang , M. L. , Kang , S. G. , Jiang , H. - L. , Shin , S. W. , Lee , D. Y. , Ahn , J. - M. , Rayamahji , N. , 
Park , I. - K. , Shin , S. J. , Cho , C. - S. , and Yoo , H. S. ( 2006 ), In vivo induction of mucosal 
immune responses by intranasal administration of chitosan microspheres containing 
Bordetella bronchiseptica DNT . Eur. J. Pharm. Biopharm. , 63 , 215 – 220 . 
22. Huang , J. , Garmise , R. J. , Crowder , T. M. , Mar , K. , Hwang , C. R. , Hickey , A. J. , Mikszta , 
J. A. , and Sullivan , V. J. ( 2004 ), A novel dry powder infl uenza vaccine and intranasal 
delivery technology: Induction of systemic and mucosal immune responses in rats , 
Vaccine , 23 , 794 – 801 . 
23. Behl , C. R. , Pimplaskar , H. K. , Sileno , A. P. , de Meireles , J. , and Romeo , V. D. ( 1998 ), 
Effects of physicochemical properties and other factors on systemic nasal drug delivery , 
Adv. Drug Deliv. Rev. , 29 , 89 – 116 . 
24. Genta , I. , Pavanetto , F. , Conti , B. , Giunchedi , P. , and Conte , U. ( 1995 ), Improvement of 
dexamethasone dissolution rate from spray - dried chitosan microspheres , S. T. P. Pharm. 
Sci. , 5 , 202 – 207 . 
25. Gavini , E. , Hegge , A. B. , Rassu , G. , Sanna , V. , Testa , C. , Pirisino , G. , Karlsen , J. , and 
Giunchedi , P. ( 2006 ), Nasal administration of carbamazepine using chitosan microspheres: 
In vitro/in vivo studies , Int. J. Pharm. , 307 , 9 – 15 . 
26. Filipovi - Gr i , J. , Perissutti , B. , Moneghini , M. , Voinovich , D. , Martinac , A. , and Jal s enjak , 
I. ( 2003 ), Spray - dried carbamazepine - loaded chitosan and HPMC microspheres: Preparation 
and characterisation , J. Pharm. Pharmacol. , 55 , 921 – 931 . 
27. Teshima , D. , Yamauchi , A. , Makino , K. , Kataoka , Y. , Arita , Y. , Nawata , H. , and Oishi , R. 
( 2002 ), Nasal glucagon delivery using microcrystalline cellulose in healthy volunteers , 
Int. J. Pharm. , 233 , 61 – 66 . 
28. Sacchetti , C. , Artusi , M. , Santi , P. , and Colombo , P. ( 2002 ), Caffeine microparticles for 
nasal administration obtained by spray drying , Int. J. Pharm. , 242 , 335 – 339 . 
29. Kublik , H. , and Vidgren , M. T. ( 1998 ), Nasal delivery systems and their effect on deposition 
and absorption , Adv. Drug Deliv. Rev. , 29 , 157 – 177 . 
30. Ugwoke , M. I. , Verbeke , N. , and Kinget , R. ( 2001 ), The biopharmaceutical aspects of 
nasal mucoadhesive drug delivery , J. Pharm. Pharmacol. , 53 , 3 – 22 . 
31. Pringels , E. , Callens , C. , Vervaet , C. , Dumont , F. , Slegers , G. , Foreman , P. , and Remon , 
J. P. ( 2006 ), Infl uence of deposition and spray pattern of nasal powders on insulin bioavailability 
, Int. J. Pharm. , 310 , 1 – 7 . 
32. Mygind , N. , and Dahl , R. ( 1998 ), Anatomy, physiology and function of the nasal cavities 
in health and disease , Adv. Drug Deliv. Rev. , 29 , 3 – 12 . 
33. Cornaz , A. - L. , De Ascentis , A. , Colombo , P. , and Buri , P. ( 1996 ), In vitro characteristics 
of nicotine microspheres for transnasal delivery , Int. J. Pharm. , 129 , 175 – 183 . 
34. Ugwoke , M. I. , Agu , R. U. , Verbeke , N. , and Kinget , R. ( 2005 ), Nasal mucoadhesive drug 
delivery: Background, applications, trends and future perspectives , Adv. Drug Deliv. Rev. , 
57 , 1640 – 1665 . 
35. Smart , J. D. ( 2005 ), The basics and underlying mechanisms of mucosdhesion , Adv. Drug 
Deliv. Rev. , 57 , 1556 – 1568 . 
36. Mathiowitz , E. , and Chickering , D. E. ( 1999 ), Defi nitions, mechanisms and theories of 
bioadhesion , in Mathiowitz , E. , Chickering , D. E. , and Lehr , C. - M. , Eds., Bioadhesive 
c c c

676 NASAL POWDER DRUG DELIVERY 
Drug Delivery Systems: Fundamentals, Novel Approaches and Development , Marcel 
Decker , New York , pp. 1 – 10 . 
37. Lee , J. W. , Park , J. H. , and Robinson , J. R. ( 2000 ), Bioadhesive - based dosage forms: The 
next generation , J. Pharm. Sci. , 89 , 850 – 866 . 
38. Nagai , T. , Nishimoto , Y. , Nambu , N. , Suzuki , Y. , and Sekine , K. ( 1984 ), Powder dosage 
form of insulin for nasal administration , J. Controlled Release , 1 , 15 – 22 . 
39. Vidgren , P. , Vidgren , M. , Arppe , J. , Hakuli , T. , Laine , E. , and Paronen , P. ( 1992 ), In vitro 
evaluation of spray - dried mucoadhesive microspheres for nasal administration , Drug 
Dev. Ind. Pharm. , 18 , 581 – 597 . 
40. Oechslein , C. R. , Fricker , G. , and Kissel , T. ( 1996 ), Nasal delivery of octreotide: Absorption 
enhancement by particulate carrier systems , Int. J. Pharm. , 139 , 25 – 32 . 
41. Abd El - Hameed , M. D. , and Kellaway , I. W. ( 1997 ), Preparation and in vitro characterisation 
of mucoadhesive polymeric microspheres as intra - nasal delivery systems , Eur. J. 
Pharm. Biopharm. , 44 , 53 – 60 . 
42. Ugwoke , M. I. , Kaufmann , G. , Verbeke , N. , and Kinget , R. ( 2000 ), Intranasal bioavailability 
of apomorphine from carboxymethylcellulose - based drug delivery systems , Int. 
J. Pharm. , 202 , 125 – 131 . 
43. Garc i a - Arieta , A. , Torrado - Santiago , S. , Goya , L. , and Torrado , J. J. ( 2001 ), Spray - dried 
powders as nasal absorption enhancers of cyanocobalamin , Biol. Pharm. Bull. , 24 , 
1411 – 1416 . 
44. Has c i c ek , C. , G o n u l , N. , and Erk , N. ( 2003 ), Mucoadhesive microspheres containing 
gentamicin sulfate for nasal administration: Preparation and in vitro characterisation , 
Il Farmaco , 58 , 11 – 16 . 
45. Cerchiara , T. , Luppi , B. , Chidichimo , G. , Bigucci , F. , and Zecchi , V. ( 2005 ), Chitosan and 
poly(methyl vynil ether - co - maleic anhydride) microparticles as nasal sustained delivery 
systems , Eur. J. Pharm. Biopharm. , 61 , 195 – 200 . 
46. Callens , C. , and Remon , J. P. ( 2000 ), Evaluation of starch - maltodextrin - Carbopol 
974 P mixtures for the nasal delivery of insulin in rabbits , J. Controlled Release , 66 , 
215 – 220 . 
47. Callens , C. , Pringels , E. , and Remon , J. P. ( 2003 ), Infl uence of multiple nasal administrations 
of bioadhesive powders on the insulin bioavailability , Int. J. Pharm. , 250 , 415 – 
422 . 
48. Ugwoke , M. I. , Agu , R. U. , Vanbilloen , H. , Baetens , J. , Augustijns , P. , Verbeke , N. , 
Mortelmans , L. , Verbruggen , A. , Kinget , R. , and Bormans , G. ( 2000 ), Scintigraphic evaluation 
in rabbits of nasal drug delivery systems based on carbopol 971p and carboxymethylcellulose 
, J. Controlled Release , 68 , 207 – 214 . 
49. Suzuki , Y. , and Makino , Y. ( 1999 ), Mucosal drug delivery using cellulose derivatives as 
a functional polymer , J. Controlled Release , 62 , 101 – 107 . 
50. Quadir , M. , Zia , H. , and Needham , T. E. ( 2000 ), Development and evaluation of nasal 
formulations of ketorolac , Drug Deliv. , 7 , 223 – 229 . 
51. Lim , S. T. , Martin , G. P. , Berry , D. J. , and Brown , M. B. ( 2000 ), Preparation and evaluation 
of the in vitro drug release properties and mucoadhesion of novel microspheres of 
hyaluronic acid and chitosan , J. Controlled Release , 66 , 281 – 292 . 
52. Lim , S. T. , Forbes , B. , Berry , D. J. , Martin , G. P. , and Brown , M. B. ( 2002 ), In vivo evaluation 
of novel hyaluronan/chitosan microparticulate delivery systems for the nasal delivery 
of gentamicin in rabbits , Int. J. Pharm. , 231 , 73 – 82 . 
53. Gavini , E. , Rassu , G. , Sanna , V. , Cossu , M. , and Giunchedi , P. ( 2005 ), Mucoadhesive 
microspheres for nasal administration of an antiemetic drug, metoclopramide: In - vitro/ 
ex - vivo studies , J. Pharm. Pharmacol. , 57 , 287 – 294 . 

REFERENCES 677 
54. Witschi , C. , and Mrsny , R. J. ( 1999 ), In vitro evaluation of microparticles and polymer 
gels for use as nasal platforms for protein delivery . Pharm. Res. , 16 , 382 – 390 . 
55. Farraj , N. F. , Johansen , B. R. , Davis , S. S. , and Illum , L. ( 1990 ), Nasal administration of 
insulin using bioadhesive microspheres as a delivery system , J. Controlled Release , 13 , 
253 – 261 . 
56. Illum , L. , Faraj , N. , Davis , S. , Johansen , B. , and O ’ Hagan , D. ( 1990 ), Investigation of the 
nasal absorption of biosynthetic human growth hormone in sheep: Use of a bioadhesive 
microsphere delivery system , Int. J. Pharm. , 63 , 207 – 211 . 
57. Illum , L. , Fisher , A. N. , Jabbal - Gill , I. , and Davis , S. S. ( 2001 ), Bioadhesive starch microspheres 
and absorption enhancing agents act synergistically to enhance the nasal absorption 
of polypeptides , Int. J. Pharm. , 222 , 109 – 119 . 
58. Mao , S. , Chen , J. , Wei , Z. , Liu , H. , and Bi , D. ( 2004 ), Intranasal administration of melatonin 
starch microspheres , Int. J. Pharm. , 272 , 37 – 43 . 
59. Illum , L. , Farraj , N. F. , Critchley , H. , and Davis , S. S. ( 1988 ), Nasal administration of gentamicin 
using a novel microsphere delivery system , Int. J. Pharm. , 46 , 261 – 265 . 
60. Bj o rk , E. , and Edman , P. ( 1988 ), Degradable starch microspheres as a nasal delivery 
system for insulin , Int. J. Pharm. , 47 , 233 – 238 . 
61. Critchley , H. , Davis , S. S. , Farraj , N. F. , and Illum , L. ( 1994 ), Nasal absorption of desmopressin 
in rats and sheep. Effect of a bioadhesive microsphere delivery system , J. Pharm. 
Pharmacol. , 46 , 651 – 656 . 
62. Edman , P. , Bj o rk , E. , and Ryd e n , L. ( 1992 ), Microspheres as a nasal delivery system for 
peptide drugs , J. Controlled Release , 21 , 165 – 172 . 
63. Pereswetoff - Morath , L. , and Edman , P. ( 1995 ), Dextran microspheres as a potential nasal 
drug delivery system for insulin — in vitro and in vivo properties , Int. J. Pharm. , 124 , 
37 – 44 . 
64. Pereswetoff - Morath , L. , Bjurstr o m , S. , Khan , R. , Dahlin , M. , and Edman , P. ( 1995 ), 
Toxicological aspects of the use of dextran microspheres and thermogelling 
ethyl(hydroxyethyl) cellulose (EHEC) as nasal drug delivery systems , Int. J. Pharm. , 128 , 
9 – 21 . 
65. Ryd e n , L. , and Edman , P. ( 1992 ), Effect of polymers and microspheres on the nasal 
absorption of insulin in rats , Int. J. Pharm. , 83 , 1 – 10 . 
66. Vila , A. , S a nchez , A. , E vora , C. , Soriano , I. , McCallion , O. , and Alonso , M. J. ( 2005 ), 
PLA - PEG particles as nasal protein carriers: The infl uence of the particle size , Int. J. 
Pharm. , 292 , 43 – 52 . 
67. Tafaghodi , M. , Tabassi , S. A. S. , Jaafari , M. R. , Zakavi , S. R. , and Momen - nejad , M. ( 2004 ), 
Evaluation of the clearance characteristics of various microspheres in the human nose 
by gamma - scintigraphy , Int. J. Pharm. , 280 , 125 – 135 . 
68. Leitner , V. M. , Guggi , D. , Krauland , A. H. , and Bernkop - Schn u rch , A. ( 2004 ), Nasal 
delivery of human growth hormone: In vitro and in vivo evaluation of a thiomer/ 
glutathione microparticulate delivery system , J. Controlled Release , 100 , 87 – 95 . 
69. Bernkop - Schn u rch , A. , Guggi , D. , and Pinter , Y. ( 2004 ), Thiolated chitosans: Development 
and in vivo evaluation of a mucoadhesive permeation enhancing oral drug delivery 
system , J. Controlled Release , 94 , 177 – 186 . 
70. Greimel , A. , Dorly del Curto , M. , D ’ Antonio , M. , Palmberger , T. , Sprinzl , G. M. , and 
Bernkop - Schn u rch , A. ( 2006 ), In vitro evaluation of thiomer microparticles for nasal 
drug delivery , J. Drug Deliv. Sci. Technol. , 16 , 103 – 108 . 
71. Dodane , V. , Khan , M. A. , and Merwin , J. R. ( 1999 ), Effect of chitosan on epithelial permeability 
and structure , Int. J. Pharm. , 182 , 21 – 32 . 

678 NASAL POWDER DRUG DELIVERY 
72. Singla , A. K. , and Chawla , M. ( 2001 ), Chitosan: Some pharmaceutical and biological 
aspects — An update , J. Pharm. Pharmacol. , 53 , 1047 – 1067 . 
73. Illum , L. , Watts , P. , Fisher , A. N. , Hinchcliffe , M. , Norbury , H. , Jabbal - Gill , I. , Nankervis , 
R. , and Davis , S. S. ( 2002 ), Intranasal delivery of morphine , J. Pharm. Exp. Ther. , 301 , 
391 – 400 . 
74. Illum , L. ( 2003 ), Nasal drug delivery — Possibilities, problems and solutions , J. Controlled 
Release , 87 , 187 – 198 . 
75. Illum , L. , Farraj , N. F. , and Davis , S. S. ( 1994 ), Chitosan as a novel nasal delivery system 
for peptide drugs , Pharm. Res. , 8 , 1186 – 1189 . 
76. Illum , L. , Jabbal - Gill , I. , Hinchcliffe , M. , Fisher , A. N. , and Davis , S. S. ( 2001 ), Chitosan 
as a novel nasal delivery system for vaccines , Adv. Drug Deliv. Rev. , 51 , 81 – 96 . 
77. Cheng , Y. - H. , Dyer , A. M. , Jabbal - Gill , I. , Hinchcliffe , M. , Nankervis , R. , Smith , A. , and 
Watts , P. ( 2005 ), Intranasal delivery of recombinant human growth hormone (somatropin) 
in sheep using chitosan - based powder formulations , Eur. J. Pharm. Sci. , 26 , 9 – 15 . 
78. Thanou , M. M. , Verhoef , J. C. , Romeijn , S. G. , Nagelkerke , J. F. , Merkus , F. W. H. M. , and 
Junginger , H. E. ( 1999 ), Effects of N - trimethyl chitosan chloride, a novel absorption 
enhancer, on Caco - 2 intestinal epithelia and the ciliary beat frequency of chicken embryo 
trachea , Int. J. Pharm. , 185 , 73 – 82 . 
79. Kotze , A. F. , Lue . en , H. L. , de Boer , A. G. , Verhoef , J. C. , and Junginger , H. E. ( 1998 ), 
Chitosan for enhanced intestinal permeability: Prospects for derivatives soluble in 
neutral and basic environments , Eur. J. Pharm. Sci. , 7 , 145 – 151 . 
80. Bernkop - Schn u rch , A. , Hornof , M. , and Guggi , D. ( 2004 ), Thiolated chitosans , Eur. J. 
Pharm. Biopharm. , 57 , 9 – 17 . 
81. Illum , L. , Jorgensen , H. , Bisgaard , H. , Krogsgaard , O. , and Rossing , N. ( 1987 ), Bioadhesive 
microspheres as a potential nasal drug delivery system , Int. J. Pharm. , 39 , 189 – 199 . 
82. Ugwoke , M. I. , Agu , R. U. , Jorissen , M. , Augustijns , P. , Sciot , R. , Verbeke , N. , and Kinget , 
R. ( 2000 ), Nasal toxicological investigations of Carbopol 971P formulation of apomorphine: 
Effects on ciliary beat frequency of human nasal primary cell culture and in vivo 
on rabbit nasal mucosa , Eur. J. Pharm. Sci. , 9 , 387 – 396 . 
83. Soane , R. J. , Hinchcliffe , M. , Davis , S. S. , and Illum , L. ( 2001 ), Clearence characteristics 
of chitosan based formulations in the sheep nasal cavity , Int. J. Pharm. , 217 , 183 – 191 . 
84. Jones , N. ( 2001 ), The nose and paranasal sinuses physiology and anatomy , Adv. Drug 
Deliv. Rev. , 51 , 5 – 19 . 
85. Soane , R. J. , Frier , M. , Perkins , A. C. , Jones , N. S. , Davis , S. S. , and Illum, L. (1999), Evaluation 
of the clearence characteristics of bioadhesive systems in humans , Int. J. Pharm. , 
178 , 55 – 65 . 
86. Vivien , N. , Buri , P. , Balant , L. , and Lacroix , S. ( 1994 ), Nasal absorption of metoclopramide 
administrated to man , Eur. J. Pharm. Biopharm. , 40 , 228 – 231 . 
87. Holmberg , K. , Bj o rk , E. , Bake , B. , and Edman , P. ( 1994 ), Infl uence of degradable starch 
microspheres on human nasal mucosa , Rhinology , 32 , 74 – 77 . 
88. Brime , B. , Ballesteros , M. P. , and Frutos , P. ( 2000 ), Preparation and in vitro characterization 
of gelatin microspheres containing Levodopa for nasal administration , J. Microencapsul. 
, 17 , 777 – 784 . 
89. Preda , M. , and Leucuta , E. ( 2003 ), Oxprenolol - loaded bioadhesive microspheres: Preparation 
and in vitro/in vivo characterization , J. Microencapsul. , 20 , 777 – 789 . 
90. Kriwet , B. , and Kissel , T. ( 1996 ), Poly(acrylic acid) microparticles widen intercellular 
spaces of Caco - 2 cell monolayers: An examination by confocal laser scanning microscopy 
, Eur. J. Pharm. Biopharm. , 42 , 233 – 240 . 

REFERENCES 679 
91. Varshosaz , J. , Sadrai , H. , and Alinagari , R. ( 2004 ), Nasal delivery of insulin using chitosan 
microspheres , J. Microencapsul. , 21 , 761 – 774 . 
92. Jain , S. K. , Chourasia , M. K. , Jain , A. K. , Jain , R. K. , and Shrivastava , A. K. ( 2004 ), Development 
and characterization of mucoadhesive microspheres bearing salbutamol for 
nasal delivery , Drug Deliv. , 11 , 113 – 122 . 
93. Krauland , A. H. , Guggi , D. , and Bernkop - Schn u rch , A. ( 2006 ), Thiolated chitosan microparticles: 
A vehicle for nasal peptide drug delivery , Int. J. Pharm. , 307 , 270 – 227 . 
94. Jiang , H. - L. , Park , I. - K. , Shin , N. - R. , Kang , S. - G. , Yoo , H. - S. , Kim , S. - I. , Suh , S. - B. , Akaike , 
T. , and Cho , C. - S. ( 2004 ), In vitro study of the immune stimulating activity of an athrophic 
rhinitis vaccine associated to chitosan microspheres , Eur. J. Pharm. Biopharm. , 58 , 
471 – 476 . 
95. Genta , I. , Costantini , M. , Asti , A. , Conti , B. , and Montanari , L. ( 1998 ), Infl uence of glutaraldehyde 
on drug release and mucoadhesive properties of chitosan microspheres , 
Carbohydr. Polym. , 36 , 81 – 88 . 
96. Hinchcliffe , M. , and Illum , L. ( 1999 ), Intranasal insulin delivery and therapy , Adv. Drug 
Deliv. Rev. , 35 , 199 – 234 . 
97. Freiberg , S. , and Zhu , X. X. ( 2004 ), Polymer microspheres for controlled drug release , 
Int. J. Pharm. , 282 , 1 – 18 . 
98. Li , Y. , Jiang , H. L. , Zhu , K. J. , Liu , J. H. , and Hao , Y. L. ( 2005 ), Preparation, characterization 
and nasal delivery of a - cobrotoxin - loaded poly(lactide - co - glycolide)/polyanhydride 
microspheres , J. Controlled Release , 108 , 10 – 20 . 
99. Ugwoke , M. I. , Verbeke , N. , and Kinget , R. ( 1997 ), Microencapsulation of apomorphine 
HCI with gelatin , Int. J. Pharm. , 148 , 23 – 32 . 
100. Yang , Y. - Y. , Chung , T. - S. , Bai , X. - L. , and Chan , W. - K. ( 2000 ), Effect of preparation conditions 
on morphology and release profi les of biodegradable polymeric microspheres 
containing protein fabricated by double - emulsion method , Chem. Eng. Sci. , 55 , 
2223 – 2236 . 
101. Conte , U. , Giunchedi , P. , Maggi , L. , and Torre , M. L. ( 1994 ), Spray dried albumin microspheres 
containing nicardipine , Eur. J. Pharm. Biopharm. , 40 , 203 – 208 . 
102. He , P. , Davis , S. S. , and Illum , L. ( 1998 ), In vitro evaluation of the mucoadhesive properties 
of chitosan microspheres , Int. J. Pharm. , 166 , 75 – 68 . 
103. Martinac , A. , Filipovic - Gr ic , J. , Voinovich , D. , Perissutti , B. , and Franceschinis , E. ( 2005 ), 
Development and bioadhesive properties of chitosan - ethylcellulose microspheres for 
nasal delivery , Int. J. Pharm. , 291 , 69 – 77 . 
104. Giunchedi , P. , Juliano , C. , Gavini , E. , Cossu , M. , and Sorrenti , M. ( 2002 ), Formulation 
and in vivo evaluation of chlorhexidine buccal tablets prepared using drug - loaded chitosan 
microspheres , Eur. J. Pharm. Biopharm. , 53 , 233 – 239 . 
105. He , P. , Davis , S. S. , and Illum , L. ( 1999 ), Chitosan microspheres prepared by spray drying , 
Int. J. Pharm. , 187 , 53 – 65 . 
106. Pavanetto , F. , Genta , I. , Giunchedi , P. , Conti , B. , and Conte , U. ( 1994 ), Spray dried 
albumin microspheres for the intra - articular delivery of dexamethazone , J. Microencapsul. 
, 11 , 445 – 454 . 
107. Martinac , A. , Filipovic - Gr ic , J. , Perissutti , B. , Voinovich , D. , and Pavelic , Z . ( 2005 ), 
Spray - dried chitosan/ethylcellulose microspheres for nasal drug delivery: Swelling study 
and evaluation of in vitro drug release properties , J. Microencapsul. , 22 , 549 – 561 . 
108. Fundueanu , G. , Constantin , M. , Dalpiaz , A. , Bortolotti , F. , Cortesi , R. , Ascenzi , P. , and 
Menegatti , E. ( 2004 ), Preparation and characterization of starch/cyclodextrin bioadhesive 
microspheres as platform for nasal administration of Gabexate Mesylate (Foy(r)) 
in allergic rhinitis treatment , Biomaterials , 25 , 159 – 170 . 
c
c

680 NASAL POWDER DRUG DELIVERY 
109. Berthold , A. , Cremer , K. , and Kreuter , J. ( 1996 ), Preparation and characterization of 
chitosan microspheres as drug carrier for prednisolone sodium phosphate as model for 
anti - infl ammatory drugs , J. Controlled Release , 39 , 17 – 25 . 
110. Jaganathan , K. S. , and Vyas , S. P. ( 2006 ), Strong systemic and mucosal immune responses 
to surface - modifi ed PLGA microspheres containing recombinant Hepatitis B antigen 
administrated intranasally , Vaccine , 24 , 4201 – 4211 . 
111. Callens , C. , Ceulemans , J. , Ludwig , A. , Foreman , P. , and Remon , J. P. ( 2003 ), Rheological 
study on mucoadhesivity of some nasal powder formulations . Eur. J. Pharm. Biopharm. , 
55 , 323 – 328 . 
112. Santos , C. A. , Jacob , J. S. , Hertzog , B. A. , Freedman , B. D. , Press , D. L. , Harnpicharnchai , 
P. , and Mathiowitz , E. ( 1999 ), Correlations of two bioadhesion assays: The everted sac 
technique and the CAHN microbalance , J. Controlled Release , 61 , 113 – 122 . 
113. Rango Rao , K. V. , and Buri , P. ( 1989 ), A novel in situ method to test polymers and coated 
microparticles for bioadhesion , Int. J. Pharm. , 52 , 265 – 270 . 
114. Agu , R. U. , Jorissen , M. , Kinget , R. , Verbeke , N. , and Augustijns , P. ( 2002 ), Alternatives 
to in vivo nasal toxicological screening for nasally - administrated drugs , S. T. P. Pharm. 
Sci. , 12 , 13 – 22 . 
115. Hvidberg , A. , Djurup , R. , and Hilsted , J. ( 1994 ), Glucose recovery after intranasal glucagon 
during hypoglycaemia in man , Eur. J. Clin. Pharmacol. , 46 , 15 – 17 . 
116. Ugwoke , M. I. , Agu , R. U. , Jorissen , M. , Augustijns , P. , Sciot , R. , Verbeke , N. , and Kinget , 
R. ( 2000 ), Toxicological investigations of the effects carboxymethylcellulose on ciliary 
beat frequency of human nasal epithelial cells in primary suspension culture and in vivo 
on rabbit nasal mucosa , Int. J. Pharm. , 205 , 43 – 51 . 
117. Merkus , P. , Romeijn , S. G. , Verhoef , J. C. , Merkus , F. W. , and Schouwenburg , P. F. 
( 2001 ), Classifi cation of cilio - inhibiting effects of nasal drugs , Laryngoscope , 111 , 
595 – 602 . 
118. Callens , C. , Adriaens , E. , Dierckens , K. , and Remon , J. P. ( 2001 ), Toxicological evaluation 
of a bioadhesive nasal powder containing a starch and Carbopol 974 P on rabbit nasal 
mucosa and slug mucosa , J. Controlled Release , 76 , 81 – 91 . 
119. Hirai , S. , Yashiki , T. , Matsuzawa , T. , and Mima , H. ( 1981 ), Absorption of drugs from the 
nasal mucosa of rat , Int. J. Pharm. , 7 , 317 – 325 . 
120. Adriaens , E. , and Remon , J. P. ( 1999 ), Gastropods as an evaluation tool for screening 
the irritating potency of absorption enhancers and drugs , Pharm. Res. , 16 , 1239 – 1243 . 
121. Marttin , E. , Schipper , N. G. M. , Verhoef , J. C. , and Merkus , F. W. H. M. ( 1998 ), Nasal 
mucociliary clearance as a factor in nasal drug delivery , Adv. Drug Deliv. Rev. , 29 , 
13 – 38 . 
122. Agu , R. U. , Jorissen , M. , Willems , T. , Van Den Mooter , G. , Kinget , R. , and Augustijns , P. 
( 1999 ), Effects of pharmaceutical compounds on ciliary beating in human nasal epithelial 
cells: A comparative study of cell culture models , Pharm. Res. , 16 , 1380 – 1385 . 
123. Merkus , F. W. H. M. , Schipper , N. G. M. , Hermens , W. A. J. J. , Romeijn , S. G. , and Verhoef, 
J. C. ( 1993 ), Absorption enhancers in nasal drug delivery: Effi cacy and safety , J. Controlled 
Release , 24 , 201 – 208 . 
124. Dimova , S. , Brewster , M. E. , Noppe , M. , Jorissen, M. , and Augustijns , P. (2005), The use 
of human nasal in vitro cell systems during drug discovery and development , Toxicol. in 
Vitro , 19 , 107 – 122 . 
125. Schmidt , M. C. , Peter , H. , Lang , S. R. , Ditzinger , G. , and Merkle , H. P. ( 1998 ), In vitro 
cell models to study nasal mucosal permeability and metabolism , Adv. Drug Deliv. Rev. , 
29 , 51 – 79 . 

REFERENCES 681 
126. Jorissen , M. , Van der Schueren , B. , Tyberghein , J. , Van Der Berghe , H. , and Cassiman , 
J. J. ( 1989 ), Ciliogenesis and coordinated ciliary beating in human nasal epithelial cells 
cultured in vitro , Acta Oto - Rhino - Laryngol. Begica , 43 , 67 – 73 . 
127. Agu , R. U. , Jorissen , M. , Willems , T. , Van Den Mooter , G. , Kinget , R. , Verbeke , N. , and 
Augustijns , P. ( 2000 ), Safety assesment of selected cyclodextrins: Effect on ciliary activity 
using a human cell suspension model exhibiting in vitro ciliogenesis , Int. J. Pharm. , 193 , 
219 – 226 . 
128. Illum , L. , Farraj , N. F. , Fisher , A. N. , Gill , I. , Miglietta , M. , and Benedetti , L. M. ( 1994 ), 
Hyaluronic acid ester microspheres as a nasal delivery system for insulin , J. Controlled 
Release , 29 , 133 – 141 . 
129. Sankar , C. , Rani , M. , Srivastava , A. K. , and Mishra , B. ( 2001 ), Chitosan based pentazocine 
microspheres for intranasal systemic delivery: Development and biopharmaceutical 
evaluation , Pharmazie , 56 , 223 – 226 . 
130. Ugwoke , M. I. , Sam , E. , Van Den Mooter , G. , Verbeke , N. , and Kinget , R. ( 1999 ), Nasal 
mucoadhesive delivery systems of the anti - parkinsonian drug, apomorphine: Infl uence 
of drug - loading on in vitro and in vivo release in rabbits , Int. J. Pharm. , 181 , 125 – 138 . 
131. El - Shafy , M. A. , Kellaway , I. W. , Taylor , G. , and Dickinson , P. A. ( 2000 ), Improved nasal 
bioavailability of FITC - dextran (Mw 4300) from mucoadhesive microspheres in rabbits , 
J. Drug Target. , 7 , 355 – 361 . 
132. Rajinikanth , P. S. , Sankar , C. , and Mishra , B. ( 2003 ), Sodium alginate microspheres of 
metoprolol tartarate for intranasal systemic delivery: Development and evaluation , 
Drug Deliv. , 10 , 21 – 28 . 
133. Nakamura , K. , Maitani , Y. , Lowman , A. M. , Takayama , K. , Peppas , N. A. , and Nagai , T. 
( 1999 ), Uptake and release of budesonide from mucoadhesive, pH - sensitive copolymers 
and their application to nasal delivery , J. Controlled Release , 61 , 329 – 335 . 
134. Yildiz , A. , Okyar , A. , Baktir , G. , Araman , A. , and O zsoy , Y. ( 2005 ), Nasal administration 
of heparin - loaded microspheres based on poly(lactic acid) , Il Farmaco , 60 , 919 – 924 . 


683 
5.8 
AEROSOL DRUG DELIVERY 
Michael Hindle 
Virginia Commonwealth University, Richmond, Virginia 
Contents 
5.8.1 Introduction 
5.8.2 Human Respiratory Tract and Aerosol Particle Deposition 
5.8.2.1 Human Respiratory Tract 
5.8.2.2 Mechanisms of Particle Deposition 
5.8.2.3 Pharmacokinetics 
5.8.3 Therapeutic Indications for Aerosol Delivery 
5.8.3.1 Current Applications 
5.8.3.2 Future Applications 
5.8.4 Aerosol Drug Delivery Devices 
5.8.4.1 Introduction 
5.8.4.2 Characteristics of Ideal Delivery Device 
5.8.5 Metered Dose Inhalers 
5.8.5.1 Introduction 
5.8.5.2 Metered Dose Inhaler and HFA Reformulation 
5.8.5.3 Propellants 
5.8.5.4 Excipients 
5.8.5.5 Valves 
5.8.5.6 Actuators 
5.8.5.7 Canisters 
5.8.5.8 Breath Actuation 
5.8.5.9 Spacers 
5.8.5.10 Dose Counters 
5.8.6 Dry Powder Inhalers 
5.8.6.1 Introduction 
5.8.6.2 Size Reduction and Particle Formation Technologies 
5.8.6.3 Drug – Lactose Formulations 
5.8.6.4 Dry Powder Inhaler Design 
5.8.6.5 Exubera 
5.8.7 Nebulizers 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

684 AEROSOL DRUG DELIVERY 
5.8.8 Emerging Technologies 
5.8.8.1 Soft Mist Aerosols 
5.8.8.2 Respimat 
5.8.8.3 AERx 
5.8.8.4 Mystic 
5.8.8.5 Capillary Aerosol Generator 
5.8.8.6 Staccato 
5.8.9 Conclusions 
References 
5.8.1 INTRODUCTION 
Aerosol drug delivery to the lungs has long been the route of choice for the treatment 
of respiratory diseases, including asthma and chronic obstructive airway 
disease. Metered dose inhalers (MDIs), dry powder inhalers (DPIs), and nebulizers 
have been employed to successfully deliver a wide range of pharmaceuticals principally 
to the lungs for local action. However, with their unique characteristics, the 
lungs have now begun to be targeted as a means of noninvasive delivery of systemically 
acting compounds, including genes, proteins, peptides, antibiotics, and other 
small molecules [1, 2] . The primary function of the respiratory tract is gaseous 
exchange, transferring oxygen from the inspired air to the blood and removing 
carbon dioxide from the circulation. This pulmonary circulation offers rapid absorption 
and systemic distribution of suitable drugs deposited in the airways. Due to its 
anatomical structure, however, an important secondary role is the protection of the 
body from inhalation of foreign particles (including aerosol drug particles). The 
challenge of aerosol drug delivery is to overcome this highly effective barrier and 
accurately and reproducibly deliver aerosol drug particles in suffi cient doses to their 
targeted sites within the lungs for either local action or systemic absorption. Effective 
aerosol drug delivery is tied to the aerosol inhaler that generates and delivers 
the respirable aerosol. This chapter will primarily focus on aerosol drug delivery 
devices, their development, and future prospects for pulmonary administration. 
5.8.2 HUMAN RESPIRATORY TRACT AND 
AEROSOL PARTICLE DEPOSITION 
5.8.2.1 Human Respiratory Tract 
The human respiratory tract can be divided into three main regions: fi rst, the upper 
airways, including the nose, mouth and throat (oropharnyx), and the larynx [3] . The 
conducting airways consist of the regions from the trachea to the respiratory bronchioles 
and have airway diameters between 0.6 and 20 mm. The alveolar region 
consists of respiratory bronchioles and alveolar sacs and has airway diameters 
between 0.2 and 0.6 mm. The lungs are a branching system which commences asymmetrically, 
dividing fi rst at the base of the trachea. The left and right bronchi branch 

dichotomously into the conducting airways. There are approximately 23 generations 
before the respiratory bronchioles give way to the alveoli, the site of gaseous 
exchange [4] . This branching produces a progressive reduction in airway diameter 
and also signifi cantly increases the total surface area of the lower airways [3] . 
Another important characteristic with respect to drug delivery is the extensive vascular 
circulation. The blood vessels supplying the conducting airways are part of the 
systemic circulation. In contrast, the alveolar region is connected to the pulmonary 
circulatory pathway; drugs absorbed into this circulation will avoid fi rst - pass hepatic 
metabolism effects. 
5.8.2.2 Mechanisms of Particle Deposition 
Aerosol particles are deposited in the lungs by three main mechanisms, and the site 
of deposition is dependent upon the predominating mechanism. Inertial impaction 
occurs because a particle traveling in an air stream has its own momentum (the 
product of its mass and velocity). As the direction of the airfl ow changes due to a 
bend or obstacle, the particle will continue in its original direction for a certain distance 
because of its inertia. Particles with a high momentum, due to high velocity 
or large size, are often unable to change direction before they impact on the surface 
in front of them [5] . Impaction of particles entering the mouth with a high velocity 
occurs either at the back of the mouth or at the bend where the pharynx leads to 
the trachea. Only a small fraction of particles greater than 15 . m will reach the 
trachea following mouth breathing. The majority, due to their size, will impact in the 
oropharyngeal region. Deposition by impaction will also occur as the trachea splits 
into the left and right bronchus. As the velocity of the particles decreases, inertial 
impaction becomes a less important mechanism of deposition in the smaller airways. 
Following the removal of larger particles in the upper airways by inertial impaction, 
gravitational sedimentation is the mechanism by which smaller particles (2 – 5 . m) 
are deposited in the respiratory bronchioles and alveoli. These particles settle under 
gravity and accelerate to a steady terminal velocity when the gravitational force is 
balanced by the resistance of the air through which it is traveling [6] . It is a time - 
dependent process which is aided by breath holding [7] . Brownian motion or diffusion 
is a mechanism which signifi cantly affects only particles less than 0.5 . m in 
diameter. These particles are subjected to bombardment by surrounding gas 
molecules causing random movement of the particles. In this situation, the 
diffusivity of a particle is inversely proportional to its diameter. For an extensive 
mechanistic review of the area of particle deposition readers should consult Finlay 
(2001) [8] . 
Aerosol particle size and polydispersity are major determinants of the site and 
mechanism of pulmonary deposition. Fundamental deposition studies using monodisperse 
aerosols together with mathematical models have established the optimum 
aerosol particle size for lung deposition [9 – 12] . Aerosols larger than 10 . m will 
deposit in the oropharyngeal region and will not be inhaled. Particles less than 3 . m 
will be capable of penetrating into the alveolar region. Aerosols in the size range 
3 – 10 . m will be distributed in the central and conducting airways [13] . A polydisperse 
aerosol containing a range of these particle sizes will allow deposition throughout 
the lungs. In theory, lung site deposition targeting should be possible by 
controlling the particle size of the inhalation aerosol [14] . However, a number of 
HUMAN RESPIRATORY TRACT AND AEROSOL PARTICLE DEPOSITION 685

686 AEROSOL DRUG DELIVERY 
other signifi cant variables can affect deposition within the respiratory tract and 
these often confound any efforts at targeting [15] . The patient ’ s respiratory cycle, 
both the rate and depth of breathing, will affect aerosol deposition, and this is also 
the source of large intersubject variability in deposition [16] . Slow and deep inhalations 
are required for deposition in the peripheral airways, and this is the technique 
often recommended for inhalation with the MDI [17] . A different technique may 
be required for DPIs, where the patient ’ s inspiratory effort is often the powder dispersion 
and delivery force. Flow rates greater than 60 L/min are commonly employed 
for powder inhalers [18, 19] . A fi nal respiratory maneuver can be employed to 
promote deposition; breath holding up to 10 s is generally recommended to enhance 
deposition by sedimentation [17] . Other parameters that will affect lung deposition 
are the disease state within the lungs and its effect on airway caliber together with 
the patient ’ s age and airway morphology [20 – 27] . 
5.8.2.3 Pharmacokinetics 
Once deposited on the surface of the airways, the particle is subject to absorption 
and clearance processes depending upon its physical properties and the site of 
deposition [28 – 30] . For example, a lipophilic small molecule deposited in the central 
airways would have a different pharmacokinetic profi le than a 50 - kDa macromolecule 
deposited in the alveolar region. The former may undergo mucociliary clearance 
following deposition on a ciliated epithelial cell. Following dissolution, lipophilic 
drugs may be transported across the epithelium by passive transcytosis, while hydrophilic 
compounds are taken up by other pathways such as via tight junctions and 
endocytosis. Having overcome the barrier of the epithelial layer, the drug is available 
for distribution into the systemic circulation or to its site of action. Finally, the 
drug may also be subject to metabolism within the airways. For the macromolecule 
deposited in the peripheral airways, the absorption rate has been shown to be 
dependent upon molecular size. Larger molecules are subject to active processes 
such as caveolae or vesicular transport across the cell. Diffusion remains the predominant 
mechanism for smaller lipophilic macromolecules. Insoluble molecules 
can be phagocytosed by alveolar macrophages and removed via the lymphatic 
system or the mucociliary escalator. The pharmacokinetics of inhaled drugs is complicated 
by the fact that a signifi cant fraction of the delivered dose is deposited in 
the oropharnyx or removed from the lungs via mucociliary clearance and in both 
cases subsequently swallowed [31] . An often desired goal for a pulmonary formulation 
is prolonged action within the lung. Rapid clearance or metabolism results in 
short duration of action for most inhaled drugs. A number of approaches using formulation 
excipient additives have been investigated to increase the residency or 
prolong release of drug at its site of action within the lungs [32, 33] . Microspheres 
containing nanoparticles have been formulated as dry powders for inhalation offering 
sustained - release properties [34] . In addition, prodrugs which are activated 
locally within the lungs have been used in an alternative approach [35 – 37] . 
The pharmacokinetic process of absorption, distribution, metabolism, and excretion 
within the lungs is an enormous subject area and readers are referred to specifi c 
reviews for further details [38 – 43] . Of particular interest may be the subject of 
absorption enhancer methodologies for lung delivery, which is beyond the scope of 
this chapter [44] . 

5.8.3 THERAPEUTIC INDICATIONS FOR AEROSOL DELIVERY 
5.8.3.1 Current Applications 
Aerosolized drug delivery is currently used to deliver a limited range of therapeutic 
classes of compounds. These are mainly for asthma and chronic obstructive airway 
disease. These classes of compounds include short - and long - acting . - adrenoceptor 
agonist, corticosteroids, mast cell stabilizers, and muscarinic antagonists. Of recent 
note is the popularity of combination products. These have obvious advantages from 
a patient compliance perspective. In addition, certain combinations of drugs have 
shown synergistic therapeutics benefi ts when compared to the drugs given by separate 
inhalers [45] . Long - acting . - adrenoceptor agonists and corticosteroids formulated 
as combination products are available as both MDIs and DPIs [46] . Also 
recently introduced was a MDI formulation, the R enantiomer of albuterol, which 
is believed to be mainly responsible for bronchodilation in the racemic mixture [47] . 
Zanamivir is licensed in the United States as an inhaled antiviral agent for the treatment 
of infl uenza [48] . Recombinant human deoxyribonuclease (rhDNAase) is 
available as a nebulizer product for the treatment of cystic fi brosis, in which it acts 
to liquefy viscous lung secretions [49] . And recently, insulin was approved as an 
inhaled powder for glycemic control in type I and II diabetes (see Section 5.8.6.5 ) 
[50] . 
5.8.3.2 Future Applications 
Research and development are presently underway covering a vast array of novel 
applications. Clark (2004) provides an extensive list of products and their current 
state of development [51] . A signifi cant future advance will be the development 
of inexpensive, noninvasive, stable, single - dose vaccine delivery via the lungs [52] . 
Efforts in this area are being led by the World Health Organization in the Measles 
Aerosol Project, and in a separate project, the Grand Challenges in Global Health 
initiative has funded a program to further develop an inhalation aerosol measles 
vaccine. Delivery of the measles vaccine via the lungs has been demonstrated to 
be both safe and effective [53 – 58] . Now the challenge of each of these projects is 
to produce stable inhalation vaccine formulations to be delivered via inexpensive 
inhalers while maintaining both safety and effi cacy [59] . The use of inhaled 
vaccinations in the event of a bioterrorism attack is also a potential application 
[60, 61] . 
The use of the inhalation route for the delivery of gene therapy is also an area 
of signifi cant interest [60, 62] . Cationic liposomes and polymers together with adenoviral 
vectors containing the reporting genes have been aerosolized using nebulizers 
for the majority of clinical studies. However, there are a signifi cant number of challenges 
that must be overcome before pulmonary gene delivery is deemed completely 
successful, the most important being low gene transfer effi ciency at the 
cellular level. This problem is not unique to inhalation therapy. Inhalation of a 
recombinant adenovirus containing the cystic fi brosis transmembrane regulator 
(Ad2/CFTR) demonstrated the feasibility of this approach for the treatment of 
cystic fi brosis [63, 64] . However, the limited duration of transfection and low cellular 
uptake effi ciency still remain a barrier to full utilization of this route [60, 65] . There 
THERAPEUTIC INDICATIONS FOR AEROSOL DELIVERY 687

688 AEROSOL DRUG DELIVERY 
are a number of reviews that provide updates as to recent developments in this area 
[60, 66 – 70] . 
Given the success of delivering insulin, other peptides and proteins are being 
considered for pulmonary applications [71, 72] . Leuprolide is a nonapeptide which 
has been investigated as both an MDI and DPI formulation for the treatment of 
prostrate cancer [73 – 76] . Other hormones being investigated include calcitonin for 
the treatment of Paget disease and osteoporosis, parathyroid hormone to treat 
osteoporosis, growth hormone releasing factor for the treatment of pituitary dwarfism, 
and vasoactive intestinal peptide (VIP) for the treatment of pulmonary diseases 
[60, 77 – 81] . 
Other potential inhalation applications include drugs for both local and systemic 
delivery. Inhaled tobramycin is being investigated for the treatment of Pseudomonas 
aeruginosa exacerbations in cystic fi brosis [82, 83] . Liposomal ciprofl oxacin is 
being developed as a fi rst - line defense against biowarfare agents (e.g., anthrax) 
[61] . Inhaled cyclosporine has been shown to improve survival rates and extend 
periods of chronic rejection - free survival in lung transplant patients [84] . Apomorphine 
has been proposed as an inhalation formulation for the treatment of erectile 
dysfunction [85] . Aerosol delivery of chemotherapeutic drugs has been advocated 
for the treatment of lung cancer [86] . Morphine and fentanyl have been investigated 
for alternative routes of administering analgesics [87 – 90] . Heparin and low - 
molecular - weight heparins have been aerosolized and advocated for the treatment 
of emphysema and thrombosis [91 – 93] . Iloprost, a stable prostacyclin analog, has 
been aerosolized by nebulization for use in the treatment of pulmonary hypertension 
[94] . This list of potential new treatments approached via the inhalation 
route is not exhaustive; among the other compounds under investigation are .1 - 
antitrypsin, sumatriptan, ergotamine, nicotine as replacement therapy, pentamidine, 
and ribavirin. Readers should be aware that a large number of these examples 
are proof - of - concept studies that may not get beyond in vitro experiments and 
animal studies. 
5.8.4 AEROSOL DRUG DELIVERY DEVICES 
5.8.4.1 Introduction 
As can be seen from the previous section, aerosol drug delivery continues to be an 
area of intensive research and development for the pharmaceutical industry. Not 
only are new applications for the pulmonary route being investigated, but also new 
delivery technologies are under development. The reformulation of MDIs with 
hydrofl uoroalkane (HFA) propellants together with the potential of using the inhalation 
route as a means of systemic administration has led to signifi cant technological 
advances in delivery devices. In parallel to MDI research DPIs have been 
developed from breath - actuated single - dose devices to both multiple - dose inhalers 
and active - dispersion DPIs. There is an extensive literature detailing the fundamental 
mechanisms of powder dispersion aimed at improving pulmonary deposition 
from powder inhalers. In addition, novel particle production technologies have been 
developed that provide alternatives to the traditional micronized powder for formulation 
in both MDIs and DPIs. Nebulizer technology has evolved from previously 

nonportable devices into high - effi ciency, hand - held nebulizers that offer alternatives 
to the MDI and DPI for certain treatment regimes. Finally, novel soft mist inhalers 
that generate aerosols by solution atomization have emerged on the inhaler landscape. 
All this research has focused on improving aerosol deposition effi ciency and 
reproducibility within the lungs, together with targeting the peripheral lungs for 
systemic absorption. The efforts of the last decade culminated in two signifi cant 
events. First, the regulatory approval of Proventil HFA and QVAR, the fi rst suspension 
and solution HFA MDIs, respectively. Second, in 2006, the U.S. and European 
regulatory authorities approved Exubera, an insulin DPI for the systemic treatment 
of type I and II diabetes. Exubera offered a noninvasive alternative to subcutaneous 
injections of insulin. 
5.8.4.2 Characteristics of Ideal Delivery Device 
With these developments, innovation continues toward development of the ideal 
inhaler. A number of authors have compiled lists of desired characteristics for an 
aerosol inhaler [95 – 97] . These can be grouped into patient - desired or industry - 
driven properties. From the patient ’ s perspective the overriding requirement is a 
device that is simple to operate. This is becoming increasing diffi cult to achieve as 
evidenced by the intensive patient education initiative that is being planned for the 
launch of the Exubera insulin inhaler. Poor compliance and adherence to prescribed 
therapy may be related to patients ’ failure to use the inhaler correctly [98] . Inhalers 
should be portable and contain a large number of doses. The device should also give 
some indication to the patient when it is empty. The inhaler should be suitable for 
use by all of the population, especially children and the elderly. Ganderton (1999) 
cited that from a device perspective aerosol generation should be independent of 
the patient ’ s inhalation and should continue for a substantial portion of the inspiratory 
cycle. This would minimize the reliance on coordinating inhalation and actuation 
of the device [99] . Breath - actuated devices have been developed to address this 
issue. In order to achieve lung deposition targeting, the particle size distribution of 
the aerosol should be capable of being altered depending upon the specifi c target 
region. For example, the central airways may be targeted with a 3 – 5 . m aerosol for 
the treatment of acute bronchoconstriction, while a smaller aerosol (1 – 3 . m) might 
be used for deep lung deposition and subsequent systemic absorption [99] . In addition, 
the dose should be delivered reproducibly with minimal oropharyngeal deposition, 
perhaps as a low - velocity aerosol. There should be a minimal number of small 
parts in the inhaler, and it should be robust and reliable when placed “ in use. ” The 
manufacturer has the option of producing a disposable or refi llable unit; however, 
the inhaler should protect the formulation from environment and not affect its 
stability. 
Dolovich et al. (2005) have provided an extensive evidence - based evaluation of 
aerosol drug devices. They concluded that when selecting an inhalation delivery 
system the following should be considered: device and drug combination availability, 
clinical setting, patient age, the ability of the patient to use the device correctly, 
device use with multiple medications, cost and reimbursement, drug administration 
time, convenience in outpatient and inpatient settings, and patient and physician 
preference [100] . Other reviews have compared the benefi ts and disadvantages of 
inhalers from clinical and patients ’ perspectives [101, 102] . 
AEROSOL DRUG DELIVERY DEVICES 689

690 AEROSOL DRUG DELIVERY 
5.8.5 METERED DOSE INHALERS 
5.8.5.1 Introduction 
Since their development, MDIs have been widely used for pulmonary aerosol drug 
delivery [103] . Despite their recognized limitations, they remain the device of choice 
for many physicians around the globe. From a patient ’ s perspective, they are light, 
portable, and robust and contain multiple doses of medication. They are also relatively 
simple to operate (press and fi re); however, signifi cant numbers of patients 
experience diffi culties correctly using the MDI due to coordination problems [104] . 
To maximize lung drug deposition, actuation (pressing the MDI canister) by the 
patient must be coordinated with a slow, deep inhalation. Studies have reported that 
51% of patients fail to operate the MDI correctly [104] . This leads to low lung 
deposition, high oropharyngeal deposition, and ultimately perhaps therapeutic 
failure. From the pharmaceutical industry perspective, the components are relatively 
inexpensive; however, the formulation and manufacturing have become increasingly 
complex. There are numerous studies describing the multifaceted and interactive 
effects of propellant [105 – 110] , excipient [111 – 115] , metering valve [110, 116] , and 
actuator [116 – 119] on the aerosol particle size characteristics of the MDI [120, 121] . 
To date, the success of the MDI has relied in part on the potency and relative safety 
of the bronchodilators and corticosteroids commonly used for the treatment of 
respiratory disorders rather than its delivery effi ciency. The relatively low and often 
variable aerosol deposition effi ciency, only around 10 – 20% of the nominal dose 
being delivered to the lungs, is the challenge that is beginning to be addressed as 
the MDI looks to enter the next 50 years of aerosol drug delivery. 
5.8.5.2 Metered Dose Inhaler and HFA Reformulation 
The basic design and operation of the MDI has changed little over its lifetime. 
Aerosols are generated from a formulation of drug (0.1 – 1% w/w) either suspended 
or in solution in the liquefi ed propellant. The formulation is held under pressure in 
a canister. 
Figure 1 shows the basic components of the MDI, consisting of a canister sealed 
with a metering valve which is inserted into a plastic actuator containing the mouthpiece. 
Aerosol generation takes place when the canister is pressed against the actuation 
sump by the patient. Actuation causes the outlet valve to open and the liquefi ed 
propellant formulation is released through the actuator nozzle and subsequently 
through the mouthpiece to the patient. Metered volumes between 20 and 100 . L 
are dispensed, and as the pressurized propellant is released, it forms small liquid 
droplets traveling at high velocity. These droplets evaporate to leave drug particles 
for inhalation [117] . Purewal and Grant (1998) have assembled a defi nitive reference 
source for issues relating to the design, manufacturing, and performance of 
MDIs [122] . 
The currently marketed MDIs may look similar to the devices that were fi rst 
developed by Riker in 1950. However, due to the replacement of the ozone - 
depleting chlorofl uorocarbon (CFC) propellants with HFA propellants, virtually all 
of the components of the MDI have been altered. In 1987, the Montreal Protocol 
was drawn up, leading to the eventual phase - out of CFC propellants. MDIs contain

ing CFC propellants were granted essential - use exemptions until viable alternatives 
became available. Therefore, with this impending withdrawal, a consortium of pharmaceutical 
companies (IPACT - I and IPACT - II) worked to identify and toxicologically 
test alternative propellants for MDIs. HFA 134a and HFA 227 were identifi ed 
as viable alternatives and the task of reformulation began. At fi rst look, it appeared 
that the most expeditious route to replacing a CFC product would be to produce a 
suspension HFA MDI with exactly the same in vitro characteristics as the CFC 
MDI. This would prove to be a time - consuming route [123, 124] . While some manufacturers 
focused on producing HFA products with identical characteristics to the 
current CFC versions to accelerate the pathway through clinical testing to market. 
Others undertook extensive research and development in the area of HFA formulation 
options, and this has led to the possibility of utilizing the MDI for both local 
and systemic administration. During this reformulation effort, the industry has 
taken the opportunity to address some of the other shortcomings of the MDI [125] . 
Among these issues were poor peripheral lung delivery, variable dose delivery, and 
limitations as to the dose capable of being delivered to the lung (typically about 
1 mg) [126] . 
The replacement of CFC MDIs with inhalers formulated with the HFA propellants 
is now well underway in Europe. Although progress in the United States has 
been slower, with the introduction of suitable alternatives for albuterol inhalers, the 
FDA has ordered that CFC albuterol MDIs be withdrawn from the market by the 
end of 2008 [127] . Examples of reformulated products available in the United States 
include Ventolin HFA, which is a suspension albuterol sulfate formulation using 
HFA 134a alone. ProAir is an alternative albuterol sulfate product manufactured 
by Ivax which contains ethanol and HFA 134a. Xopenex HFA has recently been 
approved for marketing in the United States [128] . This product contains levalbuterol 
tartrate (R - albuterol enantiomer) together with HFA 134a, dehydrated 
alcohol, and oleic acid as a suspension formulation. Table 1 summarizes the HFA 
FIGURE 1 Schematic of MDI. 
METERED DOSE INHALERS 691

692 AEROSOL DRUG DELIVERY 
products currently available in the United States and their excipients. The following 
section will focus on the current options for formulation of drugs in HFA propellant 
systems and the challenges that are encountered as products are reformulated as 
HFA formulations. 
5.8.5.3 Propellants 
The CFC propellants primarily used in MDI formulations were CFC 11, 12, and 114. 
Blends of these propellants were held liquefi ed under pressures of 50 – 80 psig within 
the canister. Flocculated drug suspensions in CFC propellants were formulated 
using a surfactant (e.g., oleic acid and lecithin). In a suspension formulation, the 
aerosol particle size is dependent upon the initial micronized drug particle size 
(typically between 2 and 5 . m) and the evaporation of the propellant droplets. It 
has long been recognized that changes in CFC propellant vapor pressure result in 
changes in droplet size and velocity of the aerosol. Newman et al. (1982) showed 
that increasing the vapor pressure of the propellant blend in the MDI signifi cantly 
increased whole - lung deposition and reduced oropharyngeal deposition [110] . 
The fi rst challenge encountered during the reformulation with HFA propellants 
was the altered physicochemical properties of HFA 134a and HFA 227 compared 
to the CFC propellants [124] . Table 2 compares the physicochemical properties of 
the CFC and HFA propellants [124] . The increased polarity of HFA 134a and HFA 
227 is illustrated by the increased dipole moments and dielectric constant. From a 
practical point of view, the altered solvency properties of the HFA propellant for 
the drug, excipient, water, and, surprisingly, components of the MDI have been the 
TABLE 1 Summary of HFA Metered Dose Inhaler Products Available in United States, 
June 2006 
Product Name Drug Approval Date Excipients Type 
Proventil HFA 
(3M) 
Albuterol sulfate August 1996 HFA 134a, ethanol, 
oleic acid 
Suspension 
Ventolin HFA 
(GSK) 
Albuterol sulfate April 2001 HFA 134a Suspension 
Proair HFA 
(IVAX) 
Albuterol sulfate October 2004 HFA 134a, ethanol Suspension 
QVAR (3M) Beclomethasone 
dipropionate 
September 2000 HFA 134a, ethanol, 
oleic acid 
Solution 
Flovent HFA 
(GSK) 
Fluticasone 
propionate 
May 2004 HFA 134a, ethanol Suspension 
Atrovent HFA 
(BI) 
Ipratropium 
bromide 
November 2004 HFA 134a, purifi ed 
water, dehydrated 
alcohol, 
anhydrous citric 
acid 
Solution 
Xopenex 
(Sepracor) 
Levalbuterol 
tartrate 
March 2005 HFA 134a, 
dehydrated 
alcohol, oleic acid 
Suspension 

METERED DOSE INHALERS 693 
major issues during reformulation. Suspension formulations of micronized drug in 
the liquefi ed CFC propellant blends with surfactants were replaced with either 
suspension or solution HFA formulations, depending upon the solubility of the 
individual drug in the HFA propellants. For suspension HFA formulations, however, 
it was observed that the conventional surfactants used in the CFC products were 
insoluble in the HFA propellants without the addition of a cosolvent (e.g., ethanol) 
[129, 130] . An alternative approach to produce suspension formulations was to 
develop a new class of surfactants suitable for use in the HFA systems [131] . It was 
also observed that some of the commonly used inhalation drugs were slightly 
soluble in the new propellants and therefore precluded their formulation as a suspension. 
The potential to formulate as a solution offered a number of advantages 
together with signifi cant problems. Perhaps most importantly, changing from a suspension 
to a solution formulation altered the mechanism of aerosol particle formation. 
In the case of solution formulations, drug is dissolved in the liquefi ed propellant 
and a suitable cosolvent (if necessary) and particle formation takes place during 
evaporation of the propellant. This leads to much smaller particles being formed 
when propellant evaporation is complete. Stein and Myrdal (2006) recently described 
the MDI aerosol generation for solution formulations as a two - step process [132] . 
Droplet formation takes place as millions of atomized droplets are produced after 
the formulation exits the metering valve through the actuator sump. Initial droplet 
size is dependent upon the vapor pressure and surface tension of the formulation, 
TABLE 2 Comparison of Physicochemical Characteristics of CFC and HFA Propellants 
Property 
CFC HFA 
11 12 114 134a 227 
Thermodynamic 
RRM 137 121 171 102 170 
Boiling point, ° C 24 . 30 4 . 26 . 16 
Vapor pressure, 20 ° C, kPa 89 566 182 572 390 
Enthalpy vap., 20 ° C, kJ/mol (J/g) 25.1 
(183) 
17.2 
(142) 
22.1 
(130) 
18.6 
(182) 
19.6 
(115) 
C p liquid, 20 ° C, J/mol · K (J/g · K) 120 
(0.88) 
118 
(0.98) 
168 
(0.98) 
143 
(1.41) 
210 
(1.24) 
Polarity 
Dielectric constant 2.3 2.1 2.2 9.5 4.1 
Dipole moment ( D ) 0.45 0.51 0.58 2.1 1.2 
Induced polarization 2.8 2.3 3.2 6.1 6.1 
Solubility parameter (Hild. units) 7.5 6.1 6.4 6.6 6.2 
Kauri - butanol value 60 18 12 9 13 
log P octanol/water 2.0 2.2 2.8 1.1 2.1 
Water solubility (ppm) 130 a 120 a 110 a 2200 b 610 b 
Liquid Phase 
Density (g/cm 3 ) 1.49 1.33 1.47 1.23 1.42 
Viscosity (mPa · s) 0.43 0.20 0.30 0.21 0.27 
Surface tension (mN/m) 18 9 11 8 7 
Source: From ref. 124 . a 30 ° b 25 ° . 

694 AEROSOL DRUG DELIVERY 
valve size, and actuator orifi ce diameter. The second step is an evaporative or 
“ aerosol maturation ” phase, as the propellant and cosolvents (e.g., ethanol) rapidly 
evaporate leaving inhalable drug particles. The fi nal size of these particles is dependent 
upon the initial droplet size, the vapor pressure of the formulation mixture, 
and the proportion of nonvolatiles in the formulation [132] . 
Leach et al. (1998) compared the pulmonary deposition of a suspension CFC 
formulation of beclomethasone diproprionate with a solution HFA formulation 
[133] . The marketed CFC product had a mass median aerodynamic diameter 
(MMAD) of 3.5 . m compared to 1.1 . m for the solution formulation, refl ecting the 
altered aerosol formation mechanism. The gamma scintigraphy profi le for the solution 
formulation showed the drug and label to be diffusely deposited throughout 
the airways with approximately 55 – 60% deposited in the lungs. In contrast, the CFC 
product was deposited mainly in the mouth and throat (90 – 94%), with only 4 – 6% 
being deposited in the airways. In many ways, this study summarized the defi ciencies 
of the suspension formulation CFC MDI and offered the alternative of improved 
delivery effi ciencies with the solution HFA formulation. A signifi cant conclusion 
from this and other studies supported the hypothesis that improved pulmonary 
deposition and reduced oropharyngeal losses of aerosols would allow reduction in 
the dose required by the patient to achieve the same therapeutic effect [108, 133, 
134] . 
The altered solubility profi le of the HFA propellant, while providing attractive 
characteristics for solution formulations, also provide signifi cant challenges with 
respect to their interactions with the basic MDI components. Leachables are compounds 
that can be transferred from MDI component parts to the formulation 
during the shelf life of the product. Berry et al. (2003) postulated that MDI orientation 
could affect the amount of leachables that entered a formulation and affect the 
particle size distribution of aerosol [135] . Extensive efforts are now required for 
extractable and leachable testing of the component materials prior to formulation 
of an MDI. Another by - product of replacing the CFC propellants was to tighten the 
impurity specifi cations required for the new propellants. A proposed U.S. Pharmacopeia 
(USP) monograph for HFA 134a has now been published detailing the 
impurity profi le [136] . 
Manufacturing processes for MDIs have also required adapting for the use with 
the new propellant system [137] . There are two main manufacturing processes used 
for MDIs: cold fi lling and pressure fi lling [138] . Cold fi lling requires cooling the 
propellants to below . 50 ° F and fi lling at that temperature prior to crimping the 
valve onto the canister. Pressure - fi lling techniques for MDIs are most commonly 
employed. These can be accomplished in either a one - or two - step process. In the 
single - step process, the formulation is placed in a pressurized mixing vessel. The 
empty canister is purged with propellant to remove the air. The valve is then crimped 
onto the canister and the formulation is metered through the valve. The absence of 
a HFA propellant that was liquid at room temperature was a major difference 
compared to the process employed for CFC manufacturing. In the two - step process, 
the formulation (excluding the propellant) are mixed together to form a concentrate. 
Previously, liquefi ed CFC 11 was used in this step of the process. However, 
there is no suitable HFA propellant that is liquid at room temperature. Therefore, 
cosolvents such as ethanol and glyercol are employed during this step to form the 
product concentrate. The concentrate is metered into the empty canister. The valve 

METERED DOSE INHALERS 695 
is then crimped onto the canister and the propellant is fi lled through the valve. 
Wilkinson (1998) provides an extensive history and review of the manufacturing 
procedures for MDIs [138] . 
5.8.5.4 Excipients 
A number of excipients have been included in MDI formulations; however, the 
nature of the excipients has changed with the introduction of the HFA propellant 
aerosols. Oleic acid and sorbitan trioleate (SPAN 85) and lecithins were used in 
CFC suspension MDIs as suspending agents and valve lubricants [112] . Typical 
concentrations ranged from 0.1 to 2.0% w/w. Ethanol is now being used in HFA 
formulations as a cosolvent for suspension and solution formulations. The addition 
of ethanol to the formulation has a number of effects [121, 139] . Increasing the 
ethanol concentration has been shown to increase the initial droplet size [121] . In 
addition, ethanol can increase the hydrophilicity of the formulation and increase 
moisture uptake. Glycerol and polyethyleneglycol have also been added as cosolvents 
but also have the effect of increasing the residual droplet particle size due to 
their lower volatility [111] . In general, a relationship can be observed between the 
fraction of nonvolatile components (drug and nonvolatile excipients) in a solution 
HFA formulation and the fi nal particle size of the aerosol. The MMAD was observed 
to be linearly proportional to the cube root of the nonvolatile concentration [119, 
121] . Oligolactic acids (OLAs) have been investigated for their use in a variety of 
functions in HFA formulations. OLAs with repeating units of 6 – 15 units have been 
proposed as suspending agents [131] . They are readily soluble in both HFA 134a 
and 227. These molecules have also been shown to act as ion pair solubilizers for 
certain drugs (e.g., albuterol). The addition of ethanol to these OLA formulations 
synergizes the solubilizing effect [131] . 
A word of caution is required when considering introduction of novel excipients 
into any inhalation drug product formulation. Due to the unique toxicological challenges 
associated with administration and clearance from the lung, the qualifi cation 
of novel excipients for inhalation has proven to be an expensive and time - 
consuming challenge. This has led to a limited number of compounds with an extensive 
“ in - use ” profi le being commonly employed. 
5.8.5.5 Valves 
Metering valves are required to accurately meter and dispense the formulation upon 
MDI actuation. In addition, they perform an important contact closure role preventing 
moisture ingress and minimizing propellant evaporation. Figure 2 and Table 3 
show the basic components of the metering valve. Currently, the most common valve 
type is the retention valve, consisting of a plastic metering chamber and two rubber 
gaskets. The remaining valve components are manufactured from plastic, metal, and 
elastomeric materials. 
Material component evaluation and selection are critical steps in the development 
of a MDI formulation [140, 141] . The materials must be chemically resistant 
and compatible with all components of the formulation. Gaskets must have appropriate 
mechanical properties and work effectively as a seal, preventing leakage of 
the formulation and moisture ingress. While the basic components themselves have 

696 AEROSOL DRUG DELIVERY 
remained unchanged during the introduction of the HFA propellants, the materials 
used to manufacture the components have required signifi cant adaptation. Nitrile 
was the most commonly employed elastomer in CFC MDIs; it has good mechanical 
and elastic properties. However, it has been shown to swell when in contact with 
HFA propellants and ethanol. Newer elastomers such as ethylene propylene diene 
monomer (EPDM), chloroprene, and bromobutyl are now used in HFA MDIs [142] . 
The ideal universal elastomer has yet to be developed and the newer materials must 
be assessed on a case - by - case basis for formulation compatibility and the desired 
moisture ingress characteristics. Among the many issues to be considered when 
screening materials are formulation – material compatibility, extractable profi les, and 
mechanical resistance. Manufacturers such as Valois, Solvay, and Bespak have extensive 
knowledge of drug/excipient/material component compatibility and should be 
used as the fi rst point of reference when considering a MDI formulation project. 
Another concern to formulators is the ingress of moisture into HFA - formulated 
MDIs [143] . HFA propellants have a higher moisture affi nity compared to the CFC 
propellants, especially HFA 134a. In addition, the inclusion of ethanol in some formulations 
increases its hydrophilicity. Moisture entering the canister can have 
several effects; it may alter the physical or chemical stability of the formulation and 
aerosolization performance of HFA MDIs. Due to its lower volatility compared to 
FIGURE 2 Schematic of components of metering valve. (Courtesy of Valois Pharm.) 
Upper stem 
First gasket 
Ring 
Ferrule 
Second gasket 
Metering chamber 
Neck gasket 
Lower stem 
Spring 
Body 
TABLE 3 Summary of Components and Materials Used in 
Metered Dose Inhaler Valves 
Component Material 
Metering chamber Polyester 
Core Polyester/acetal 
Core extension Polyester/acetal 
Body Polyester/nylon 
Seats/gaskets EPDM/nitrile/butyl/chloroprene/ 
bromobutyl 
Spring Stainless steel 
Ferrule Aluminium 

METERED DOSE INHALERS 697 
the other components, water may affect aerosol generation and alter aerosol particle 
size [144] . The increased water content may increase the solubility of suspended 
polar drug particles or decrease the solubility of hydrophobic compounds [145] . 
Corrosion in aluminum canisters may also increase over the shelf life of the product. 
Williams and Hu (2000) reported that HFA 134a had a greater tendency to take up 
moisture during storage than did HFA 227 [144] . The issue of moisture ingress 
during storage has led to certain HFA MDIs being stored in moisture - protecting 
pouches prior to initial use (e.g., Ventolin HFA). An alternative approach to minimize 
the effects of moisture ingress has been taken by SkyePharma, which has 
incorporated subtherapeutic doses of cromolyn sodium into its HFA MDI formulations. 
Cromolyn sodium is used as a hygroscopic excipient to scavenge any moisture 
that penetrates into the formulation. Cromolyn sodium has been used widely by 
inhalation over the past 30 years and has an excellent safety profi le via the inhalation 
route. Burel et al. (2004) reported that for a HFA 134a MDI formulation the 
inclusion of a polyamide (nylon 66) molded ring around the valve body reduced 
both the initial water content and the fi nal water content (6 months) when stored 
under stress conditions [40 ° C and 75% relative humidity (RH)]. A combination of 
a thermoplastic elastomer sealing gasket in the MDI valve and a polyamide ring 
produced the lowest water ingress under these stress conditions [143] . For formulations 
that might be susceptible to water - induced stability issues, HFA 227 may be 
considered a more suitable propellant than HFA 134a. Given the possibility of 
moisture ingress, there is also the issue of propellant leakage. Leak testing is among 
the array of in - process quality assurance tests that are required. These include assay 
of the suspension or solution, moisture level, consistency of fi lling of both the concentrate 
and the propellant, valve crimp measurements, quality of sealing, in - line 
leak testing under stress conditions, and performance of the valve. 
Another signifi cant issue encountered during use of MDIs was related to loss of 
prime and dose reproducibility [129, 146] . Loss of prime relates to the fact that in 
conventional capillary retention metering valves the dose is fi lled into the valve 
immediately following the last actuation. Capillary retention valves require priming 
with one or two sprays prior to their fi rst use. In addition, if there is a signifi cant 
interval between the actuations and the inhaler is stored upside down or on its side 
or shaken, then the metering valve may actually partially empty, resulting in a low 
and variable dose being delivered to the patient. A review of patient information 
leafl ets indicated varying instructions on priming MDIs. This ranged from Atrovent 
CFC and Combivent CFC requiring priming with 3 sprays “ after 24 hours of nonuse. ” 
Ventolin HFA and Proventil HFA both required priming with 4 sprays after “ 2 
weeks of nonuse. ” Flovent CFC required priming with 4 sprays after “ 4 weeks of 
nonuse. ” Clearly, such instructions add to the complexity for patients using MDIs 
and also contribute to drug waste issues. Loss of prime is also a signifi cant issue for 
breath - actuated MDIs, where the opportunity to prime the inhaler is not readily 
possible. A number of new valve designs have been developed to address this issue. 
The fast - fi ll, fast - empty valves offer a solution to the priming and loss of prime 
issues. In these valves [(e.g., 3M Shuttle valve (3M), 3M Face Seal valve (3M), ACT 
(Valois), and Easifi ll valve (Bespak)], the metering valve is only isolated from the 
formulation canister reservoir immediately prior to dose actuation. Therefore, the 
metering chamber can be emptied and refi lled with a fresh dose from the reservoir 
simply by shaking the canister prior to use. 

698 AEROSOL DRUG DELIVERY 
5.8.5.6 Actuators 
Nonvolatile component concentration has previously been described as one of the 
primary determinants of the initial droplet size for HFA solution formulations [119] . 
Perhaps, equally important is the MDI actuator [147] . The actuator consists of the 
sump block into which the metered dose is immediately delivered during MDI 
actuation. As expansion and vaporization of the propellant take place, the aerosol 
exits the sump via the actuator nozzle and then is inhaled through the actuator 
mouthpiece. From a practical perspective, in general, reducing the size of the orifi ce 
diameter for HFA solution formulations produced a relatively slower spray emitted 
with less force compared to marketed CFC products [118] . The nozzle orifi ce diameter 
has been considered to be the most important, although not the only, actuator 
variable determining the particle size distribution of HFA solution formulations 
[118, 147, 148] . Recently, Smyth et al. (2006) described three critical components of 
the actuator that could affect the aerosol performance of a solution HFA formulation. 
In addition to the orifi ce diameter, sump depth (and hence the expansion 
chamber volume) together with orifi ce length was observed to have signifi cant 
effects on the aerosol particle size distribution and should be considered for optimization 
with an HFA formulation [147] . It has also been recognized that the electrostatic 
charge of all components of the MDI and its formulation may affect the 
aerosolization properties of the aerosol spray [149, 150] . 
5.8.5.7 Canisters 
Aluminum canisters are widely used in commercial MDI products mainly due to 
their inert characteristics. Other materials, including stainless steel and glass, can be 
employed depending upon the particular formulation characteristics. These canisters 
were usually uncoated. Changes to the canister may be required when the formulation 
interacts with the interior surface of the canister altering the chemical 
stability of the formulation. The presence of ethanol in HFA formulations has also 
increased the risk of metal corrosion. Drug migration or absorption to the metal 
components of the canister and also the metal valve components has also been 
reported [141] . The loss of drug to the walls of the canister will result in variability 
in the delivered dose from the MDI during the shelf life of the inhaler. The use of 
canister coating and anodized canisters has been advocated to mitigate this problem 
[141] . 
5.8.5.8 Breath Actuation 
In order to overcome the problems associated with many patients ’ inability to coordinate 
actuating the MDI and inhaling, breath - actuated MDIs were developed [107] . 
These devices allow the MDI to be automatically actuated only when the patient 
commences inhaling through the mouthpiece. Of critical importance here is ensuring 
that the patient has suffi cient inspiratory fl ow rate to trigger actuation. While 
these devices offer little improvement for patients with a good inhaler technique, it 
has been shown that patients with poor coordination did have signifi cantly greater 
lung drug deposition when inhaling using a breath - actuated MDI [151] . The 3M 
Autohaler was the fi rst device marketed using this technology [151, 152] . In Europe, 

METERED DOSE INHALERS 699 
the Easibreathe and Autohaler breath - actuated MDIs are used to deliver . agonists 
and corticosteroids for the treatment of obstructive airway. Recently, in the United 
States, the MD Turbo has been launched, a device that allows patients to take their 
regular MDI canister and actuator and insert it into the MD Turbo. The MD Turbo 
acts as a generic breath actuator for a number of marketed MDIs and also incorporates 
a dose counter. 
5.8.5.9 Spacers 
Spacer devices have been developed as another alternative to overcome the problems 
associated with patients coordinating the beginning of their inspiratory effort 
with actuation of the MDI [153] . This problem is extenuated by the fact that the 
MDI emits a high - velocity, short - duration aerosol cloud. On actuation, the propellant 
spray is delivered into the spacer that often incorporates a one - way inhalation 
valve. The patient is now able to inhale the aerosol cloud. The large - volume spacers 
have an additional effect in that they allow evaporation of large propellant droplets 
prior to inhalation. These high - velocity droplets would previously have had a high 
probability of impacting in the patient ’ s throat. Figure 3 shows the large number of 
spacer chambers that are available [154] . Spacers are advocated for use by children 
and elderly patients and people who experience diffi culty coordinating actuation of 
the MDI. The use of spacers for the delivery of corticosteroids also minimizes oral 
deposition of the inhaled dose and therefore reduces the incidence of steroid - 
related side effects [155] . Both in vitro and clinical studies have shown the effectiveness 
of spacers with CFC MDIs [156 – 158] . It has been shown that electrostatic 
charge can have a signifi cant effect on the performance of a spacer chamber and 
where possible the charge should be minimized to maximize drug delivery [159 – 
161] . Finally, it should be noted that the use of any particular spacer – MDI combination 
should be evaluated at least in vitro to confi rm the benefi cial effect, especially 
when employed with solution - based HFA MDI formulations [162, 163] . 
FIGURE 3 Example spacer chambers available for use with MDIs. (Reproduced from 
ref. 154 with permission of Pharmacotherapy .) 
OptiHaler® ACE® 
MediSpacer® InspirEase® 
OpriChamber® 
Space ChamberTM 
EasiVent® 
EZ-Spacer® 
Ellipse® 
Gentle-Haler® AeroChamber® 
BreatheRite® 
6’’Tube

700 AEROSOL DRUG DELIVERY 
5.8.5.10 Dose Counters 
A guidance document from the Food and Drug Administration (FDA) recommends 
the addition of a dose counter to the MDI. This would overcome a long - standing 
problem with the MDI, the inability of a patient to accurately know the number of 
doses remaining in the canister [164] . Dose counters have been incorporated successfully 
into multiple - dose DPIs. In general, the counter should give a clear indication 
of when approaching end of life and the actual end of life. It should be either 
numeric or color coded. If numeric, it should count downward and should be 100% 
reliable and avoid undercounting [165] . 
5.8.6 DRY POWDER INHALERS 
5.8.6.1 Introduction 
Dry power inhalers have been in use for over 40 years. They were developed as an 
environmentally friendly alternative to the MDI. The early DPIs were simple in 
design, portable, but again, a relatively ineffi cient means of delivering drugs to the 
lungs for local action [166] . The Spinhaler, the fi rst DPI, has been prescribed in 
Europe since the late 1960s. In general, the acceptance and use of DPIs is much 
greater in Europe than in the United States. However, with the reformulation efforts 
for MDIs, there are an increasing number of DPIs becoming available in the United 
States (Figure 4 ). 
Research and development for dry powder inhalers have two main focuses: the 
optimization of the powder formulation for use in these inhalers and investigations 
of novel DPI device designs and technology. An enormous literature now exists in 
each of these areas; for more extensive reviews readers should consult refs. 33, 167, 
or 168 . 
FIGURE 4 Example DPIs available in United States. 

5.8.6.2 Size Reduction and Particle Formation Technologies 
Dry powder inhaler formulations consist usually of either a drug - only formulation 
or an ordered mixture of drug and excipient, most commonly lactose monohydrate. 
In both cases, the fi rst challenge is the production of drug particles with suitable 
size characteristics for inhalation (i.e., 1 – 5 . m). Traditionally, micronization or jet - 
milling methods have been employed as the method of choice for conventional 
small molecules. This method is identical to that employed for the production of 
fi ne particles for suspension MDIs. Using this method it is possible to produce 
primary particles between 1 – 5 . m. However, as a consequence of the particle size 
reduction there are a number of undesirable effects with respect to the powder 
properties. Micronized powders possess high intramolecular forces and are cohesive. 
They readily form aggregates that are diffi cult to disperse to the primary particles. 
Dispersion to its primary particle is essential for successful pulmonary deposition. 
In addition, they often possess high inherent electrostatic charges which cause particle 
adhesion to the components of the dry powder inhaler [169] . The high - energy 
micronization process also causes disruption of the crystal lattice and results in the 
formation of amorphous regions which may affect the long - term stability of the 
formulation [170] . Finally, it is not possible to control the drug particle morphology. 
Despite all of these problems, micronization remains the most common technique 
employed for respirable particle formation. Modifi cations to conventional micronization 
techniques have been investigated as alternative methods of particle size 
reduction [171 – 173] . 
A number of novel particle formation technologies now exist that are able to 
produce respirable drug particles for formulation in both DPIs and MDIs. Depending 
on the method of preparation, these particles offer unique and potentially 
advantageous physical and aerodynamic properties compared to conventional crystallization 
and micronization techniques. Some investigators have advocated that 
major improvements in aerosol particle performance may be achieved by lowering 
particle density and increasing particle size, as large, porous particles display less 
tendency to agglomerate than (conventional) small and nonporous particles. Also, 
large, porous particles inhaled into the lungs can potentially release therapeutic 
substances for long periods of time by escaping phagocytic clearance from the lung 
periphery, thus enabling therapeutic action for periods ranging from hours to many 
days [174] . 
Many of these techniques involve particle formation from solution formulations 
that contain novel excipients. Spray drying is the most advanced of these technologies 
and has been used to produce the powder formulation in the Exubera inhaler 
[175] . Various modifi cations of this basic technique, including co – spray drying with 
novel excipients, have been employed. 
AIR particles are low - density lipid - based particles that are produced by spray 
drying lipid – albumin – drug solutions. These particles are characterized by their 
porous surface characteristics and large geometric diameter while having a low 
aerodynamic diameter [176, 177] . This technology has been used to produce porous 
particle powder formulations of L - dopa that have been investigated for the treatment 
of Parkinson ’ s disease [178] . 
Pulmospheres are produced using a proprietary spray drying technique, with 
phosphatidylcholine as an excipient to produce hollow and porous particles with 
DRY POWDER INHALERS 701

702 AEROSOL DRUG DELIVERY 
low interparticulate forces. These particles have been formulated as suspended 
particles in HFA MDIs. In comparison with conventional suspension MDIs, the 
Pulmosphere MDI exhibited signifi cantly higher fi ne particle fractions. This technology 
has been used to produce cromolyn sodium, albuterol sulfate and formoterol 
fumarate microspheres [179] . Pulmospheres powder formulations containing tobramycin 
and budesonide have also been tested clinically [83, 180] . 
Technosphere technology has been developed as an alternative porous particle 
for pulmonary delivery [181] . These porous microspheres are formed by precipitating 
a drug - diketopiperazine derivative from an acidic solution. Para - thryroid 
hormone (PTH) Technospheres have been investigated for the treatment of osteoporosis 
following aerosol delivery [182] . 
The use of supercritical fl uid processing technology has also been widely used 
for its application in controlled microparticle formation. Conventional small molecules 
and proteins for inhalation have been generated and formulated as powders 
for inhalation. [183 – 186] . 
The application of pulmonary delivery of nanoparticles ( < 1 um) for pharmaceuticals 
remains to be developed [187 – 189] . 
5.8.6.3 Drug – Lactose Formulations 
The most common means of overcoming cohesion problems is by incorporation of 
a carrier excipient. Lactose monohydrate is used most often; it is inert, cheap, widely 
available, and a GRAS (generally regarded as safe) non - toxic excipient. A signifi - 
cant area of research has been undertaken to optimize the critical parameters 
involved in the formulation of drug – lactose blends. Micronized drug is typically 
blended with lactose (50 – 100 . m) to produce an ordered mix. The blend ratio is 
fi xed depending upon the dose of drug to be delivered and the mass of powder 
blend in each dosage unit (typically between 5 and 25 mg). The aerosolization properties 
of the blend are related to the adhesive forces between the drug and lactose 
together with the cohesive forces between the drug particles. Reproducible dispersion 
of the blend either by the dry powder inhaler (active DPI) or by the patients ’ 
inspiratory effort (passive DPI) is required. This allows the detached micronized 
drug to be inhaled and deposited in the respiratory tract while the larger lactose 
particles are deposited by inertial impaction in the oropharnyx. 
Formulators have become increasingly aware of the criticality of the drug and 
lactose powder surface characteristics and their relationship to the aerosolization 
performance in a DPI [190, 191] . A number of investigators have shown in vitro the 
importance of controlling the size of the lactose and the amount of “ fi nes ” (lactose 
particles less than 5 . m in size) in the drug – lactose blend [192 – 194] . Inherent fi nes 
are present in all lactose powders, and the fi nes are usually adhered to the surface 
of the larger lactose particle. These fi nes are believed to occupy “ active ” or high - 
energy sites on the lactose particle surface. Occupation of these sites by the lactose 
fi nes prevents the micronized drug from adhering to these positions. This allows the 
drug to adhere to less active sites and become detached easier from the lactose 
surface during inhalation. Obviously any signifi cant change in the quantity of fi nes 
present in the lactose may alter the distribution of the micronized drug on the 
lactose particle and therefore the aerosolization characteristics of the powder blend 
[193 – 195] . Batch - to - batch control of the fi nes content of inhalation lactose has been 

recognized as critical to ensuring reproducible in vitro emitted and fi ne particle 
doses. Jones and Price (2006) have recently surveyed the literature in this area and 
provided a comprehensive review [196] . Modifi cation of the surface characteristics 
of the lactose particle has been used as an alternative approach to control the adherence 
of drug particles to the lactose surface [197 – 200] . Alternative sugar carriers 
have also been investigated; these appear to possess many of the same performance - 
limiting characteristics as lactose [201] . Finally, tertiary additives have also been 
used to improve the aerosolization properties of DPI formulations [202] . The majority 
of the studies described above relate to in vitro testing of DPI formulation performance, 
and little is known about the clinical signifi cance of these studies. 
Moisture ingress into a powder formulation is a particular concern as it may signifi 
cantly decrease the aerosolization performance of the formulation [203, 204] . 
Increased adhesion of particles is often seen following exposure to high - RH environments 
[205, 206] . Moisture ingress has also been shown to affect drug stability 
[170] . The pharmaceutical industry has used a number of approaches to protect 
powder formulations from the ingress of moisture during storage and for their “ in - 
use ” life. The Turbuhaler incorporates a desiccant in the base of the inhaler to keep 
the power reservoir free from moisture [207] . Unit - dose blisters used in the Diskus 
are sealed in a foil strip pack to protect each individual dose prior to inhalation 
[208] . It is also essential that the patient not exhale into the DPI immediately prior 
to inhaling the dose. 
Electrostatic charge can also infl uence the performance of DPI formulations. A 
number of studies have investigated the interactions of drug and lactose particle 
charge with respect to aerosolization properties and drug retention by the plastic 
components within the inhaler [209 – 212] . 
5.8.6.4 Dry Powder Inhaler Design 
Inhalation Flow Rate The main function of a DPI is to facilitate dispersion and 
delivery of inhalable drug particles. An extensive patent and scientifi c literature 
exists describing the ever - increasing number of DPI device designs [33] . Powder 
dispersion in the early passive DPIs was provided in part by the inspiratory effort 
of the patient. This removed the necessity to coordinate patient inhalation with 
actuation and delivery of the dose (in contrast to MDIs). These passive DPIs were 
“ breath actuated, ” with the patients ’ inspiratory effort dispersing, aerosolizing, and 
delivering the powder during the inhalation cycle. The airfl ow rate through the 
inhaler was determined by the inherent device resistance and the inspiratory force 
exerted by the patient [18] . Devices such as the Spinhaler, Rotahaler, and Diskhaler 
are low - resistance devices requiring relatively high inspiratory fl ow rates to disperse 
the powder formulations by turbulent deaggregation. These simple devices have low 
aerosolization effi ciencies with only 5 – 20% of the dose being delivered to the lungs 
[166] The inhalation fl ow rate dependence of passive DPIs has been cited as a 
potential problem in their use, especially given the large intersubject fl ow rate variability 
within the patient population (especially for the young and older patients). 
In vitro testing revealed that for certain DPIs there was large variability in both the 
delivered dose and the aerodynamic particle size distribution as a function of the 
inhalation fl ow rate [203, 213 – 216] . Similar clinical studies also revealed a fl ow rate 
dependence for certain DPIs while others were observed to perform with a degree 
DRY POWDER INHALERS 703

704 AEROSOL DRUG DELIVERY 
of fl ow rate independence [214, 217 – 221] . When choosing a DPI, the effect of inhalation 
fl ow rate should be assessed on a case - by - case basis for each individual DPI, 
and readers should be aware of contradictory studies, especially when comparing in 
vitro and clinical performance. The Turbuhaler is one such example, where some in 
vitro studies show high variability; however, this is not refl ected in clinical studies 
[215, 220, 222] . 
From these and many other studies it can be concluded that a desirable characteristic 
for any DPI is that its dose delivery performance is independent of inhalation 
fl ow rate. A second generation of DPIs have been developed that incorporate 
a combination of improved powder formulations, more effective turbulent dispersion 
within the inhaler, and in some cases an active dispersion mechanism. The 
Exubera inhaler releases a bolus of compressed air through the formulation and 
actively generates an aerosol cloud from the powder (Figure 5 ). The cloud is held 
within a reservoir chamber from which the patient then inhales the insulin dose 
[175] . Active dispersion improves device aerosolization effi ciency, with greater than 
50% of the dose being deposited in the lungs, while minimizing the reliance on the 
patients ’ inspiratory effort. 
Single - and Multiple - Dose DPI s Inhalation powder dose metering is one of the 
problems encountered by DPI formulators. The powder dose can range from 250 . g 
in the drug - only Pulmicort Turbuhaler formulation to 25 mg in the lactose - blended 
FIGURE 5 Exubera Inhaler. (Reprinted from ref. 175 . Courtesy of Mary Ann Liebert, 
Inc.) 
Transparent 
chamber 
TransJector 
(disperser) 
Blister 
pack 
Pump 
handle 
Actuation 
button

Spinhaler formulations. In each case, accurate and reproducible metering of the 
powder is required for regulatory approval and therapeutic effi cacy. This proved to 
be a technological challenge that was solved in a number of ways. Single - unit - dose 
inhalers were the fi rst generation of DPIs, the unit dose being metered in the factory 
and subsequently loaded into the inhaler by the patient immediately prior to each 
dosing. Because metering takes place prior to batch release by the manufacturer, 
this allows for quality control and release testing, ensuring that dosage units were 
within acceptable criteria. Procedures such as capsule fi lling were common for early 
devices such as the Spinhaler and Rotahaler. This approach is still used by some of 
the newer devices being developed (e.g., Aerohaler and Cyclohaler) [185, 223] . 
While popular with the pharmaceutical industry, the single - unit - dose device required 
signifi cant patient handling to load and empty the inhaler for each inhalation (unlike 
the MDI, which often contained up to 200 doses available for inhalation on demand). 
Two approaches were taken toward the design of multiple - dose DPIs; the multiple - 
unit - dose DPI (e.g., Diskhaler and Diskus) and the powder reservoir multidose DPI 
(e.g., Turbuhaler) [207] . For the multiple - unit - dose DPI, manufacturers sought to 
address the requirement for multiple doses while retaining the control of factory 
premetering. Perhaps the most successful DPI in this respect is the Diskus, in which 
the dose is premetered into a coiled foil covered strip containing individually sealed 
blister reservoirs [208] . Each blister is opened immediately prior to inhalation and 
up to 60 doses can be help in each foil strip. For the powder reservoir multidose 
DPI, volumetric dose metering of the powder takes place within the DPI immediately 
prior to inhalation in a manner analogous to MDIs. Among the devices 
that use this approach are the Turbuhaler, Clickhaler, Pulvinal, and Easyhaler 
[220, 224 – 226] . The Turbuhaler is used with a drug - only formulation (although 
lactose blends have also been used) that employs a proprietary powder agglomeration 
process to produce loosely bound aggregates that are easily dispersed by the 
patient ’ s inhalation and by the turbulent fl ow path encountered in the DPI [227] . 
Besides the Diskus and Turbuhaler, there are four other devices currently available 
in the United States, the Asmanex Twisthaler, the Foradil Aerolizer, the Relenza 
Diskhaler, and the Spiriva Handihaler. Other devices in development include 
the Novolizer, a multidose, refi llable, breath - actuated DPI that delivers up to 
200 metered doses of drug from a single cartridge [228, 229] . The Ultrahaler 
offers yet another alternative DPI [230] . The Taifun inhaler, the JAGO inhaler, and 
the Airmax are other multidose DPIs [231 – 234] . 
5.8.6.5 Exubera 
Systemic delivery of drugs via the lungs offers a noninvasive route of administration. 
Perhaps the most important and widely investigated molecule considered for this 
route has been insulin [235, 236] . Following over a decade of development, in 
January 2006, Pfi zer and its partner Nektar received marketing approval for Exubera, 
their insulin DPI. This offered diabetics a noninvasive route of insulin administration 
rather than repeated subcutaneous injections [237] . Exubera has been indicated 
for the treatment of adult patients with diabetes mellitus for the control of hyperglycemia 
[238] . It has an onset of action similar to rapid - acting insulin analogs and 
has a duration of glucose - lowering activity comparable to subcutaneously administered 
regular human insulin [239] . Patton et al. (2004) provided an extensive review 
DRY POWDER INHALERS 705

706 AEROSOL DRUG DELIVERY 
of the clinical pharmacokinetics and pharmacodynamics of inhaled insulin [240] . In 
patients with type I diabetes, Exubera should be used in regimens that include a 
longer acting insulin [241] . In patients with type II diabetes, Exubera can be used 
as monotherapy or in combination with oral agents or longer acting insulins [242 – 
244] . Studies revealed that the same level of blood sugar control was achieved following 
inhalation compared to subcutaneous injection, although different nominal 
doses were required due to lung bioavailability issues [245] . The therapeutic effi cacy 
and safety of inhaled insulin appears to have been proven, although there are a 
signifi cant number of issues with its administration via this route [246] . It has been 
noted that asthmatics absorb less insulin from the lungs than nonasthmatics. In 
addition, smokers absorb more insulin than nonsmokers. Small and reversible 
changes in pulmonary lung function have been observed in some studies with 
inhaled insulin. Each of these issues has led to the development of specifi c prescribing 
guidelines and an intensive physician/patient education program for the inhaled 
insulin product. The Exubera insulin formulation is a spray - dried, amorphous insulin 
powder containing 60% insulin in a buffered, sugar - based matrix [175] . 
Other pharmaceutical companies are also continuing to develop their own inhalation 
insulin products. Aradigm and NovoNordisk are using a liquid insulin formulation 
in combination with the AERx IDMS inhaler [247 – 251] . Alkermes and Lily 
are developing an insulin product derived from their research on geometrically 
large, low - density particles that are formed by a spray drying process incorporating 
a natural phospholipid. MannKind is using its Technosphere technology to produce 
low - density porous insulin particles. This formulation is delivered using the MedTone 
inhaler. Other companies working in this area include Kos Pharmaceuticals, Mircodose 
Technologies, Coremed, and Biosante. 
5.8.7 NEBULIZERS 
Nebulization of liquid formulations has long been established as an effective, if not 
effi cient, means of pulmonary drug delivery. The basic principle of nebulizer aerosol 
generation has remained unchanged; however, a number of technological advances 
have been made which have improved effi ciency and reduced variability. Aerosols 
that were previously delivered in a continuous inhalation mode over 5 – 15 min are 
now delivered only during the inspiratory cycle, thus reducing drug waste. In general, 
nebulizers convert a liquid into a fi ne droplet mist, either by means of a compressed 
gas (jet nebulizer) or by high - frequency sound (ultrasonic nebulizer) [252] . Ultrasonic 
nebulizers use a piezoelectric source within the formulation reservoir to 
induce waves at the surface of the nebulizer formulation. Interference of these 
waves induces the formation of droplets which are then carried in a fl owing air 
stream that is passed over the formulation. These devices are not suitable for the 
nebulization of suspension formulations [253] . Rau (2002) also observed that ultrasonic 
nebulizers can increase the solution reservoir temperature and may cause drug 
degradation [254] . In the case of the jet nebulizer, an aerosol is produced by forcing 
compressed air through a narrow orifi ce which is positioned at the end of a capillary 
tube. The negative pressure created by the expanding jet causes formulation to be 
drawn up to the capillary tube from the reservoir in which it is immersed. As the 
liquid emerges from the tip of the capillary, it is drawn into the air stream and 

broken up into droplets by the jet to produce an aerosol. Baffl e structures within 
the nebulizer fi lter the large droplets from the aerosol by impaction and the deposited 
drug solution is recycled back into the drug reservoir [255] . Only the small 
aerosol droplets evade impaction on the baffl es and are delivered to the patient for 
inhalation. Jet nebulizers can be categorized by function, for example, the DeVilbiss 
646 is a conventional jet nebulizer with continuous drug output resulting in signifi - 
cant waste during exhalation. The Pari LC Plus system incorporated a valve system 
and operates as an active venturi jet nebulizer; although drug output is continuous, 
there is an increased output during inhalation. The patient ’ s inspiratory effort 
increases the nebulizer airfl ow, thus increasing drug output for these breath - 
enhanced nebulizers. Finally, dosimetric jet nebulizers such as the Ventstream use a 
one - way valve system to emit aerosol only during inspiration and are also breath - 
enhanced nebulizers [256, 257] . It is this last type of nebulizer that offers the most 
signifi cant advances in technology [258] . 
Jet nebulizers are commonly used in nonambulatory settings such as hospitals or 
the patient ’ s home. In vitro studies comparing the performance of commercial 
nebulizers have concluded that there were large differences in drug delivery between 
nebulizers of different classes and even between nebulizers of apparently the same 
class [259 – 261] . The aerosolization performance of different nebulizers has been 
found to be dependent upon a number of factors, including the drug being aerosolized, 
the formulation fi ll volume, the compressed airfl ow rate, and breathing pattern 
[254, 260, 262, 263] . These parameters ultimately control the aerosol droplet size and 
rate of drug output [264] . However, probably the size of the conventional nebulizer, 
the duration of the treatment cycle (5 – 15 min), and the cost of the nebulizers are 
the main reasons that they are usually reserved for nonambulatory settings and 
remain less popular than the MDI and DPI. 
Solutions or suspensions are available as nebulizer formulations. Due to the relative 
simplicity in formulating a liquid nebulizer formulation and because of the relatively 
large range of doses available for delivery, the nebulization method is often 
chosen as the aerosol method for proof - of - concept investigational studies. 
Nebulizer technology continues to be developed to miniaturize and lower the 
cost of the devices while maintaining the quality of the aerosols generated. The 
Halolite incorporates adaptive aerosol delivery which monitors patients inspiratory 
cycle and delivers drug to patients during the fi rst 50% of their inspiratory cycle 
[265 – 267] . The Pari eFlow is a hand - held device that uses a vibrating membrane 
nebulizer to generate a respirable aerosol [268] . Aerogen (now part of Nektar) has 
a range of nebulizer - based technologies, including the Aeroneb and Aerodose 
devices. Aerosols are generated as a liquid formulation passes through vibrating 
apertures [269, 270] . 
5.8.8 EMERGING TECHNOLOGIES 
5.8.8.1 Soft Mist Aerosols 
In recent years research has focused on a new method of pharmaceutical aerosol 
generation that involves passing a solution formulation through a nozzle or series 
of nozzles to generate a “ soft mist ” aerosol as a bolus dose [271] . Aerosol generation 
EMERGING TECHNOLOGIES 707

708 AEROSOL DRUG DELIVERY 
is achieved by mechanical, thermomechanical or electromechanical processes 
depending upon the particular technology employed [272] . It is worth noting that 
these devices are bolus dose delivery inhalers, rather than the new continuous - 
generation nebulizers which generate aerosols by vibrating porous membranes at 
ultrasonic frequencies. Such devices include the eFlow and Aerodose, which were 
described earlier. 
While the precise mechanism of soft mist aerosol generation may differ between 
inhalers, a number of common characteristics can be observed. They are propellant 
free and produce slow - moving aerosols over an extended duration with high in vitro 
fi ne - particle fractions compared to MDIs and DPIs. The aerosols are often generated 
from simple solution formulations containing pharmaceutically acceptable 
excipients. Water and ethanol are the most commonly employed vehicles for soft 
mist aerosols [273] . Perhaps the most simple and advantageous vehicle is water. 
There is often a well - known and established stability profi le of many pharmaceuticals 
in aqueous solutions, accelerating the route to the clinic in any development 
program. Drug solubility can be manipulated by choice of water, ethanol, or mixtures 
of the two to increase formulation options and doses. In multidose reservoir - 
type devices, a preservative would be required to prevent microbial contamination. 
This is in addition to the current federal regulations that all aqueous - based drug 
products for oral inhalation must be manufactured to be sterile. 
5.8.8.2 Respimat 
The Respimat inhaler was recently launched in Germany as a combination product 
of fenoterol and ipratropium hydrobromide (Berodual) and was licensed for the 
treatment of chronic obstructive airway disease. A large body of literature now 
exists documenting the aerosol characteristics and clinical performance of the 
Respimat inhaler with a number of different drugs [274, 275] . Aerosolized formulations 
include the steroids budesonide and fl unisolide in addition to the . agonist 
fenoterol as well as the commercially available combination product of fenoterol 
and ipratropium bromide [276 – 281] . 
The Respimat device is a multidose reservoir system that is primed by twisting 
the device base (Figure 6 ). This compresses a spring and transfers a metered volume 
of formulation from the drug cartridge to the dosing chamber. The metered volume 
is between 11 and 15 . l depending upon the drug formulation. When the device is 
actuated (in coordination with the patient ’ s inspiration), the spring is released. This 
forces a micropiston into the dosing chamber and pushes the solution through the 
uniblock. The uniblock is the heart of the aerosol generation system and consists of 
a fi lter structure with two fi ne outlet nozzle channels. The uniblock produces two 
fi ne jets of liquid that converge at a precisely set angle and then collide. This collision 
aerosolizes the liquid to form an aerosol [282] . 
Aerosols generated from the Respimat inhaler have been characterized as having 
a prolonged aerosol cloud duration compared to MDIs and have a slower cloud 
velocity as measured using video camera imaging. Hochrainer et al. (2005) measured 
the cloud duration of the Respimat aerosol to be 0.2 – 1.6 s compared to less than 
0.2 s for HFA and CFC MDIs. Aerosol velocities have been reported as less than 
1 m/s for the Respimat, compared to 6 – 8 m/s for CFC MDI inhalers [283] . While a 
degree of patient coordination is required to actuate the Respimat and to inhale, 

the longer duration of aerosol cloud generation makes this maneuver less critical 
than with MDIs. 
Aqueous and ethanolic formulations have been employed with the Respimat 
and the in vitro aerosol performance determined. Zierenberg (1999) reported fi ne - 
particle fractions of 66% for an aqueous fenoterol formulation and 81% for an 
ethanolic fl unisolide formulation. The respective MMADs were 2.0 ± 0.4 . m for 
the aqueous formulation and 1.0 ± 0.3 . m for the ethanolic formulation [284] . 
5.8.8.3 AER x 
The AERx system was developed for the systemic delivery of insulin. Unit - dose 
aqueous solution formulations were produced in a blister strip design. The fi rst - 
generation AERx device is a battery - operated device that guides the patient through 
the inhalation technique required to successfully deliver a dose. It can also monitor 
dose times and frequency together with the facility to download dosing data in 
the clinic. A number of macromolecules, including insulin, and traditional small 
molecules (e.g., morphine) have been investigated using the AERx technology 
[89, 251] . 
FIGURE 6 Respimat Inhaler. (Courtesy of Boehringer Ingelheim.) 
Mouthpiece 
Uniblock 
Dose release button 
Capillary tube 
Upper housing 
Transparent base 
spring 
Cartridge 
EMERGING TECHNOLOGIES 709

710 AEROSOL DRUG DELIVERY 
Aerosol generation using the AERx system is achieved by mechanically forcing 
a dose of the liquid formulation though a nozzle array in its disposable unit - dose 
blisters. The electronic version of the AERx inhaler guides the patient to inhale at 
the required fl ow rate. A cam - operated piston mechanism is actuated to compress 
the blister and extrude the dose as an aerosol through the nozzle array into warmed 
fl owing air. The nozzle array consists of a number of laser - drilled holes. Nozzle 
design characteristics can be altered depending upon the formulation characteristics 
and the desired droplet particle size. The single - use nature of the blister avoids 
potential problems such as microbial contamination from a dosing solution reservoir 
and nozzle - clogging issues. 
A number of prototype versions of the AERx system have been investigated. In 
general, the in vitro aerosol characteristics revealed that about 50 – 60% of the 
loaded dose was emitted from the device, of which over 90% was respirable. MMADs 
ranged from 1 to 3 . m depending upon the formulation and nozzle array [285] . In 
a scintigraphic study, lung deposition following inhalation from the AERx was 
53.3% (expressed as a percentage of the radioactivity in the AERx blister) compared 
to 21.7% for an MDI [285] . 
A number of clinical studies delivering insulin to diabetic patients using the 
AERx system are currently ongoing. Hermansen et al. (2004) concluded that in type 
II diabetics, preprandial inhaled insulin via the AERx was as effective as preprandial 
subcutaneous insulin in achieving glycemic control [286] . Clinical studies with morphine 
revealed comparable analgesic effi cacy for a matched dose of inhaled and 
intravenous morphine in a postsurgical pain model [251] . In addition, the AERx 
inhaler has been employed for the topical delivery of rhDNase to cystic fi brosis 
patients. A mean relative increase in forced expiratory volume in 1 s (FEV1) of 7.8% 
was observed after 15 days treatment compared to control [287] . 
5.8.8.4 Mystic 
The Mystic inhaler offers a soft mist aerosol generated from solution or suspension 
formulations. Unlike the previously described soft mist inhalers which use purely 
mechanical forces to generate the aerosol, the Mystic inhaler applies an electric fi eld 
to the formulation within the spray nozzle [288] . An electric charge builds on the 
fl uid surface and, as the droplets exit the nozzle, the repelling force of the surface 
charge overcomes the surface tension of the droplets to form a soft mist droplet 
aerosol. This process is known as electrohydrodynamic aerosolization or electrospray. 
The particle size characteristics of the aerosol can be controlled by adjusting 
the physical and chemical characteristics of the formulation together with the formulation 
fl ow rate and electrical fi eld properties. The inhaler consists of a number 
of components, a drug containment system, metering system, aerosol nozzle, power 
supply, and microprocessor, all enclosed in a housing. To date, Ventaira reports that 
the inhaler has been successfully employed to generate aerosols from small - 
molecule formulations (albuterol, triamcinolone, cromolyn, budesonide, and terbutaline) 
and macromolecules, including insulin [288] . 
5.8.8.5 Capillary Aerosol Generator 
In the capillary aerosol generator (CAG) system, the aerosol is formed by pumping 
the drug formulation through a small, electrically heated capillary. Upon exiting the 

capillary, the formulation is rapidly cooled by ambient air to produce an aerosol. 
The generated aerosol characteristics are dependent upon the formulation employed. 
Using propylene glycol as a condensing vehicle, drug containing condensation aerosols 
are generated [289] . When using water, ethanol, or combinations of both as 
noncondensing excipients, a stream of solid particles is delivered as a soft mist 
aerosol. In vitro studies using budesonide, cromolyn sodium, buprenophine, albuterol, 
and insulin have been performed to demonstrate various applications of the 
CAG technology. These studies are characterized by high emitted doses and high 
fi ne - particle fractions. Using noncondensing excipients, it is possible to produce 
aerosols with vastly different size characteristics, depending upon the required 
application. 
5.8.8.6 Staccato 
This technology utilizes a rapid heating technique to vaporize a thin fi lm of drug. 
Following vaporization, the drug particles condense in the inhalation fl ow stream 
to form a respirable aerosol and are inhaled. Single - and multiple - dose breath - 
actuated inhalers are currently in development. As with any method involving 
heating of a formulation, drug degradation must be minimal. Rabinowitz et al. 
(2006) described the absorption of prochlorperazine from human lungs as similar 
to the pharmacokinetic profi les observed following intravenous administration [290, 
291] . 
5.8.9 CONCLUSIONS 
Pharmaceutical aerosol drug delivery has been established for over 50 years. Pulmonary 
administration remains the route of choice for local treatment of respiratory 
diseases. Over the past decade there have been changes in both the diseases treated 
by this route and the devices used for aerosol generation. Future advances will see 
pulmonary delivery of gene therapy and vaccines, together with improved drug 
targeting within the respiratory tract using novel inhalers. 
ACKNOWLEDGMENTS 
The author would like to thank Suparna Das Choudhuri and Deepika Arora 
for their assistance and discussions during the preparation of this chapter. In 
addition, he is grateful to Guillaume Brouet (Valois Pharm), Michael Spallek 
(Boehringer Ingelheim), and Joanne Peart (RDD) for their help in obtaining 
fi gures and tables used in this chapter. Finally, the review of soft mist inhalers 
has previously been published in the Drug Delivery Company Report (Autumn/ 
Winter 2004), and the author acknowledges PharmaVentures and the Drug 
Delivery Company Report, which allowed reproduction of an abridged form of 
this paper. 
The author received a research grant from Chrysalis Technologies, a division of 
Philip Morris USA, for the development of the CAG technology. 
ACKNOWLEDGMENTS 711

712 AEROSOL DRUG DELIVERY 
REFERENCES 
1. Byron , P. R. , and Patton , J. S. ( 1994 ), Drug delivery via the respiratory tract , J. Aerosol 
Med. , 7 , 49 – 75 . 
2. Gonda , I. ( 2006 ), Systemic delivery of drugs to humans via inhalation , J. Aerosol Med. , 
19 , 47 – 53 . 
3. Douglas , R. B. ( 1987 ), The physiology of the lung , in Ganderton , D. and Jones , T. M. , 
Eds., Drug Delivery to the Respiratory Tract , Ellis Horwood , Chichester , pp 13 – 26 . 
4. Weibel , E. R. ( 1963 ), Morphometry of the Human Lung , Academic , New York . 
5. Gonda , I. ( 1990 ), Aerosols for delivery of therapeutic and diagnostic agents to the respiratory 
tract , Crit. Rev. Ther. Drug Carrier Syst. , 6 , 273 – 313 . 
6. Stuart , B. O. ( 1973 ), Deposition of inhaled aerosols , Arch. Intern. Med. , 131 , 60 – 73 . 
7. Palmes , E. D. ( 1973 ), Measurement of pulmonary air spaces using aerosols , Arch. Intern. 
Med. , 131 , 76 – 79 . 
8. Finlay , W. H. ( 2001 ), Particle deposition in the respiratory tract , in Finlay , W. H. , Ed., The 
Mechanisms of Inhaled Pharmaceutical Aerosols , Academic , London , pp 119 – 174 . 
9. Lippmann , M. , and Albert , R. E. ( 1969 ), The effect of particle size on the regional deposition 
of inhaled aerosols in the human respiratory tract , Am. Ind. Hyg. Assoc. J. , 30 , 
257 – 275 . 
10. Foord , N. , Black , A. , and Walsh , M. ( 1975 ), Pulmonary deposition of inhaled particles 
with diameters in the range 2.5 to 7.5 micron , Inhaled Part. , 4 ( Pt. 1 ), 137 – 149 . 
11. Taulbee , D. B. , and Yu , C. P. ( 1975 ), A theory of aerosol deposition in the human respiratory 
tract , J. Appl. Physiol. , 38 , 77 – 85 . 
12. Yu , C. P. , and Taulbee , D. B. ( 1975 ), A theory of predicting respiratory tract deposition 
of inhaled particles in man , Inhaled Part. , 4 ( Pt. 1 ), 35 – 47 . 
13. Pavia , D. , and Thomson , M. L. ( 1976 ), The fractional deposition of inhaled 2 and 5 mum 
particles in the alveolar and tracheobronchial regions of the healthy human lung , Ann. 
Occup. Hyg. , 19 , 109 – 114 . 
14. Bennett , W. D. , Brown , J. S. , Zeman , K. L. , Hu , S. C. , Scheuch , G. , and Sommerer , K. 
( 2002 ), Targeting delivery of aerosols to different lung regions , J. Aerosol Med. , 15 , 
179 – 188 . 
15. Dolovich , M. A. ( 2000 ), Infl uence of inspiratory fl ow rate, particle size, and airway caliber 
on aerosolized drug delivery to the lung , Respir. Care , 45 , 597 – 608 . 
16. Bennett , W. D. ( 1988 ), Human variation in spontaneous breathing deposition fraction: 
A review , J. Aerosol Med. , 1 , 67 – 80 . 
17. Hindle , M. , Newton , D. A. , and Chrystyn , H. ( 1993 ), Investigations of an optimal inhaler 
technique with the use of urinary salbutamol excretion as a measure of relative bioavailability 
to the lung , Thorax , 48 , 607 – 610 . 
18. Clark , A. R. , and Hollingworth , A. M. ( 1993 ), The relationship between powder inhaler 
resistance and peak inspiratory conditions in healthy volunteers - implications for in vitro 
testing , J. Aerosol Med. , 3 , 99 – 110 . 
19. Broeders , M. E. A. C. , Molema , J. , Vermue , N. A. , and Folgering , H. T. M. ( 2001 ), Peak 
inspiratory fl ow rate and slope of the inhalation profi les in dry powder inhalers , Eur. 
Respir. J. , 18 , 780 – 783 . 
20. Phalen , R. F. , and Oldham , M. J. ( 2001 ), Methods for modeling particle deposition as a 
function of age , Respir. Physiol. , 128 , 119 – 130 . 
21. Segal , R. A. , Martonen , T. B. , Kim , C. S. , and Shearer , M. ( 2002 ), Computer simulations 
of particle deposition in the lungs of chronic obstructive pulmonary disease patients , 
Inhal. Toxicol. , 14 , 705 – 720 . 

22. Kim , C. S. , and Hu , S. C. ( 1998 ), Regional deposition of inhaled particles in human lungs: 
Comparison between men and women , J. Appl. Physiol. , 84 , 1834 – 1844 . 
23. Kim , C. S. , and Kang , T. C. ( 1997 ), Comparative measurement of lung deposition of 
inhaled fi ne particles in normal subjects and patients with obstructive airway disease , 
Am. J. Respir. Crit. Care Med. , 155 , 899 – 905 . 
24. Bennett , W. D. , Zeman , K. L. , and Kim , C. ( 1996 ), Variability of fi ne particle deposition 
in healthy adults: Effect of age and gender , Am. J. Respir. Crit. Care Med. , 153 , 
1641 – 1647 . 
25. Kim , C. S. , Abraham , W. M. , Garcia , L. , and Sackner , M. A. ( 1989 ), Enhanced aerosol 
deposition in the lung with mild airways obstruction , Am. Rev. Respir. Dis. , 139 , 
422 – 426 . 
26. Kim , C. S. , Lewars , G. A. , and Sackner , M. A. ( 1988 ), Measurement of total lung aerosol 
deposition as an index of lung abnormality , J. Appl. Physiol. , 64 , 1527 – 1536 . 
27. Palmes , E. D. , Goldring , R. M. , Wang , C. , and Altshuler , B. ( 1970 ), Effect of chronic 
obstructive pulmonary disease on rate of deposition of aerosols in the lung during breath 
holding , Inhaled Part. , 1 , 123 – 130 . 
28. Suarez , S. , and Hickey , A. J. ( 2000 ), Drug properties affecting aerosol behavior , Respir. 
Care , 45 , 652 – 666 . 
29. Groneberg , D. A. , Witt , C. , Wagner , U. , Chung , K. F. , and Fischer , A. ( 2003 ), Fundamentals 
of pulmonary drug delivery , Respir. Med. , 97 , 382 – 387 . 
30. Labiris , N. R. , and Dolovich , M. B. ( 2003 ), Pulmonary drug delivery. part I: Physiological 
factors affecting therapeutic effectiveness of aerosolized medications , Br. J. Clin. Pharmacol. 
, 56 , 588 – 599 . 
31. Hindle , M. , and Chrystyn , H. ( 1992 ), Determination of the relative bioavailability of 
salbutamol to the lung following inhalation , Br. J. Clin. Pharmacol. , 34 , 311 – 315 . 
32. Niven , R. W. ( 2004 ), Modulated drug therapy with inhalation aerosols: Revisited , in 
Hickey , A. J. , Ed., Pharmaceutical Inhalation Aerosol Technology , 2nd ed., Marcel 
Dekker , New York , pp 551 – 570 . 
33. Niven , R. W. ( 2002 ), Powders and processing: Deagglomerating a dose of patents and 
publications , in Dalby , R. N. , Byron , P.R. , Peart , J. , and Farr , S. J. , Eds., Respiratory Drug 
Delivery VIII , Davis Horwood International Publishing , Raleigh, NC , pp 257 – 266 . 
34. Cook , R. O. , Pannu , R. K. , and Kellaway , I. W. ( 2005 ), Novel sustained release microspheres 
for pulmonary drug delivery , J. Controlled Release , 104 , 79 – 90 . 
35. Crooks , P. A. , and Al - Ghananeem , A. M. ( 2004 ), Drug targeting to the lung: Chemical 
and biological considerations , in Hickey , A. J. , Ed., Pharmaceutical Inhalation Aerosol 
Technology , 2nd ed., Marcel Dekker , New York , pp 89 – 154 . 
36. Sitar , D. S. ( 1996 ), Clinical pharmacokinetics of bambuterol , Clin. Pharmacokinet. , 31 , 
246 – 256 . 
37. Svensson , L. A. ( 1991 ), Mechanism of action of bambuterol: A beta - agonist prodrug with 
sustained lung affi nity , Agents Actions Suppl. , 34 , 71 – 78 . 
38. Taburet , A. M. , and Schmit , B. ( 1994 ), Pharmacokinetic optimisation of asthma treatment 
, Clin. Pharmacokinet. , 26 , 396 – 418 . 
39. Lipworth , B. J. ( 1996 ), Pharmacokinetics of inhaled drugs , Br. J. Clin. Pharmacol. , 42 , 
697 – 705 . 
40. Hochhaus , G. , and Mollmann , H. ( 1992 ), Pharmacokinetic/pharmacodynamic characteristics 
of the beta - 2 - agonists terbutaline, salbutamol and fenoterol , Int. J. Clin. Pharmacol. 
Ther. Toxicol. , 30 , 342 – 362 . 
41. Dahl , A. R. , and Lewis , J. L. ( 1993 ), Respiratory tract uptake of inhalants and metabolism 
of xenobiotics , Annu. Rev. Pharmacol. Toxicol. , 33 , 383 – 407 . 
REFERENCES 713

714 AEROSOL DRUG DELIVERY 
42. Witek, T. J. , Jr . (2000), The fate of inhaled drugs: The pharmacokinetics and pharmacodynamics 
of drugs administered by aerosol , Respir. Care , 45 , 826 – 830 . 
43. Issar , M. , Mobley , C. , Khan , P. , and Hochhaus , G. ( 2004 ), Pharmacokinetics and pharmacodynamics 
of drugs delivered to the lungs , in Hickey , A. J. , Ed., Pharmaceutical Inhalation 
Aerosol Technology , 2nd ed., Marcel Dekker , New York , pp 215 – 252 . 
44. Hussain , A. , Arnold , J. J. , Khan , M. A. , and Ahsan , F. ( 2004 ), Absorption enhancers in 
pulmonary protein delivery , J. Controlled Release , 94 , 15 – 24 . 
45. Nelson , H. S. , Chapman , K. R. , Pyke , S. D. , Johnson , M. , and Pritchard , J. N. ( 2003 ), 
Enhanced synergy between fl uticasone propionate and salmeterol inhaled from a single 
inhaler versus separate inhalers , J. Allergy Clin. Immunol. , 112 , 29 – 36 . 
46. Dhillon , S. , and Keating , G. M. ( 2006 ), Beclometasone dipropionate/formoterol: In an 
HFA - propelled pressurised metered - dose inhaler , Drugs , 66 , 1475 – 1483 . 
47. Nowak , R. , Emerman , C. , Hanrahan , J. P. , et al. ( 2006 ), A comparison of levalbuterol 
with racemic albuterol in the treatment of acute severe asthma exacerbations in adults , 
Am. J. Emerg. Med. , 24 , 259 – 267 . 
48. Cass , L. M. , Brown , J. , Pickford , M. , et al. ( 1999 ), Pharmacoscintigraphic evaluation of 
lung deposition of inhaled zanamivir in healthy volunteers , Clin. Pharmacokinet. , 
36 ( Suppl. 1 ), 21 – 31 . 
49. Diot , P. , Vecellio - None , L. , Varaigne , F. , Marchand , S. , and Lemarie , E. ( 2003 ), Role of 
rhDNase in cystic fi brosis , Rev. Mal. Respir. , 20 , S171 – S175 . 
50. Anonymous ( 2006 ), First inhaled insulin product approved , FDA Consum. , 40, 28 – 29 . 
51. Clark , A. R. ( 2004 ), Pulmonary delivery technology: Recent advances and potential for 
the new millennium , in Hickey , A. J. , Ed., Pharmaceutical Inhalation Aerosol Technology , 
2nd ed., Marcel Dekker , New York , pp 571 – 591 . 
52. Roth , Y. , Chapnik , J. S. , and Cole , P. ( 2003 ), Feasibility of aerosol vaccination in humans , 
Ann. Otol. Rhinol. Laryngol. , 112 , 264 – 270 . 
53. Sabin , A. B. , Fernandez de Castro , J. , Flores Arechiga , A. , Sever , J. L. , Madden , D. L. , 
and Shekarchi , I. ( 1982 ), Clinical trials of inhaled aerosol of human diploid and chick 
embryo measles vaccine , Lancet. , 2 , 604 . 
54. Sabin , A. B. ( 1983 ), Immunization against measles by aerosol , Rev. Infect. Dis. , 5 , 
514 – 523 . 
55. Sabin , A. B. , Albrecht , P. , Takeda , A. K. , Ribeiro , E. M. , and Veronesi , R. ( 1985 ), High 
effectiveness of aerosolized chick embryo fi broblast measles vaccine in seven - month - old 
and older infants , J. Infect. Dis. , 152 , 1231 – 1237 . 
56. Sabin , A. B. , Flores Arechiga , A. , Fernandez de Castro , J. , Albrecht , P. , Sever , J. L. , and 
Shekarchi , I. ( 1984 ), Successful immunization of infants with and without maternal 
antibody by aerosolized measles vaccine. II. Vaccine comparisons and evidence for 
multiple antibody response , JAMA , 251 , 2363 – 2371 . 
57. Fernandez - de Castro , J. , Kumate - Rodriguez , J. , Sepulveda , J. , Ramirez - Isunza , J. M. , and 
Valdespino - Gomez , J. L. ( 1997 ), Measles vaccination by the aerosol method in Mexico , 
Salud Publica Mex. , 39 , 53 – 60 . 
58. Bennett , J. V. , Fernandez de Castro , J. , Valdespino - Gomez , J. L. , et al. ( 2002 ), Aerosolized 
measles and measles - rubella vaccines induce better measles antibody booster responses 
than injected vaccines: Randomized trials in Mexican schoolchildren , Bull. World Health 
Organ. , 80 , 806 – 812 . 
59. LiCalsi , C. , Maniaci , M. J. , Christensen , T. , Phillips , E. , Ward , G. H. , and Witham , C. ( 2001 ), 
A powder formulation of measles vaccine for aerosol delivery , Vaccine , 19 , 2629 – 2636 . 
60. Laube , B. L. ( 2005 ), The expanding role of aerosols in systemic drug delivery, gene 
therapy, and vaccination , Respir. Care , 50 , 1161 – 1176 . 

61. Blanchard , J. D. ( 2004 ), Pulmonary drug delivery as a fi rst response to bioterrorism , in 
Dalby , R. N. , Byron , P. R. , Peart , J. , Suman , J. D. , and Farr , S. J. , Eds., Respiratory Drug 
Delivery IX , Davis Healthcare International , River Grove, IL , pp 73 – 82 . 
62. Li , H. Y. , Seville , P. C. , Williamson , I. J. , and Birchall , J. C. ( 2005 ), The use of amino acids 
to enhance the aerosolisation of spray - dried powders for pulmonary gene therapy , 
J. Gene Med. , 7 , 343 – 353 . 
63. Joseph , P. M. , O ’ Sullivan , B. P. , Lapey , A. , et al. ( 2001 ), Aerosol and lobar administration 
of a recombinant adenovirus to individuals with cystic fi brosis. I. Methods, safety, and 
clinical implications , Hum. Gene Ther. , 12 , 1369 – 1382 . 
64. Perricone , M. A. , Morris , J. E. , Pavelka , K. , et al. ( 2001 ), Aerosol and lobar administration 
of a recombinant adenovirus to individuals with cystic fi brosis. II. Transfection effi ciency 
in airway epithelium , Hum. Gene Ther. , 12 , 1383 – 1394 . 
65. Rochat , T. , and Morris , M. A. ( 2002 ), Gene therapy for cystic fi brosis by means of aerosol , 
J. Aerosol Med. , 15 , 229 – 235 . 
66. Garcia - Contreras , L. , and Hickey , A. J. ( 2003 ), Aerosol treatment of cystic fi brosis , Crit. 
Rev. Ther. Drug Carrier Syst. , 20 , 317 – 356 . 
67. Gautam , A. , Waldrep , J. C. , and Densmore , C. L. ( 2003 ), Aerosol gene therapy , Mol. 
Biotechnol. , 23 , 51 – 60 . 
68. Anson , D. S. , Smith , G. J. , and Parsons , D. W. ( 2006 ), Gene therapy for cystic fi brosis 
airway disease — is clinical success imminent ? Curr. Gene Ther. , 6 , 161 – 179 . 
69. Griesenbach , U. , Geddes , D. M. , and Alton , E. W. ( 2006 ), Gene therapy progress and 
prospects: Cystic fi brosis , Gene Ther. , 13 , 1061 – 1067 . 
70. Ziady , A. G. , and Davis , P. B. ( 2006 ), Current prospects for gene therapy of cystic fi brosis , 
Curr. Opin. Pharmacol , 6 , 515 – 521 . 
71. Cryan , S. A. ( 2005 ), Carrier - based strategies for targeting protein and peptide drugs to 
the lungs , AAPS J. , 7 , E20 – 41 . 
72. Shoyele , S. A. , and Slowey , A. ( 2006 ), Prospects of formulating proteins/peptides as 
aerosols for pulmonary drug delivery , Int. J. Pharm. , 314 , 1 – 8 . 
73. Adjei , A. , and Garren , J. ( 1990 ), Pulmonary delivery of peptide drugs: Effect of particle 
size on bioavailability of leuprolide acetate in healthy male volunteers , Pharm. Res. , 7 , 
565 – 569 . 
74. Adjei , A. , Sundberg , D. , Miller , J. , and Chun , A. ( 1992 ), Bioavailability of leuprolide 
acetate following nasal and inhalation delivery to rats and healthy humans , Pharm. Res. , 
9 , 244 – 249 . 
75. Zheng , J. Y. , Fulu , M. Y. , Lee , D. Y. , Barber , T. E. , and Adjei , A. L. ( 2001 ), Pulmonary 
peptide delivery: Effect of taste - masking excipients on leuprolide suspension metered - 
dose inhalers , Pharm. Dev. Technol. , 6 , 521 – 530 . 
76. Shahiwala , A. , and Misra , A. ( 2005 ), A preliminary pharmacokinetic study of liposomal 
leuprolide dry powder inhaler: A technical note , AAPS PharmSciTech. , 6 , E482 – 486 . 
77. Deftos , L. J. , Nolan , J. J. , Seely , B. L. , et al. ( 1997 ), Intrapulmonary drug delivery of salmon 
calcitonin , Calcif. Tissue Int. , 61 , 345 – 347 . 
78. Chan , H. K. , Clark , A. R. , Feeley , J. C. , et al. ( 2004 ), Physical stability of salmon calcitonin 
spray - dried powders for inhalation , J. Pharm. Sci. , 93 , 792 – 804 . 
79. Colthorpe , P. , Farr , S. J. , Smith , I. J. , Wyatt , D. , and Taylor , G. ( 1995 ), The infl uence of 
regional deposition on the pharmacokinetics of pulmonary - delivered human growth 
hormone in rabbits , Pharm. Res. , 12 , 356 – 359 . 
80. Bosquillon , C. , Preat , V. , and Vanbever , R. ( 2004 ), Pulmonary delivery of growth 
hormone using dry powders and visualization of its local fate in rats , J. Controlled 
Release , 96 , 233 – 244 . 
REFERENCES 715

716 AEROSOL DRUG DELIVERY 
81. Ohmori , Y. , Onoue , S. , Endo , K. , Matsumoto , A. , Uchida , S. , and Yamada , S. ( 2006 ), 
Development of dry powder inhalation system of novel vasoactive intestinal peptide 
(VIP) analogue for pulmonary administration , Life Sci. , 79 , 138 – 143 . 
82. Ramsey , B. W. , Pepe , M. S. , Quan , J. M. , et al. ( 1999 ), Intermittent administration of 
inhaled tobramycin in patients with cystic fi brosis. cystic fi brosis inhaled tobramycin 
study group , N. Engl. J. Med. , 340 , 23 – 30 . 
83. Newhouse , M. T. , Hirst , P. H. , Duddu , S. P. , et al. ( 2003 ), Inhalation of a dry 
powder tobramycin PulmoSphere formulation in healthy volunteers , Chest , 124 , 360 – 
366 . 
84. Iacono , A. T. , Johnson , B. A. , Grgurich , W. F. , et al. ( 2006 ), A randomized trial of inhaled 
cyclosporine in lung - transplant recipients , N. Engl. J. Med. , 354 , 141 – 150 . 
85. Staniforth , J. N. ( 2006 ), Nasal and pulmonary powder opportunities: New drugs and 
formulations for rapid systemic onset , in: Dalby , R. N. , Byron , P. R. , Peart , J. , Suman , 
J. D. , and Farr , S. J. , Eds., Respiratory Drug Delivery , 2006, Davis Healthcare International 
, River Grove, IL , pp. 249 – 256 . 
86. Azarmi , S. , Tao , X. , Chen , H. , et al. ( 2006 ), Formulation and cytotoxicity of doxorubicin 
nanoparticles carried by dry powder aerosol particles , Int. J. Pharm. , 319 , 155 – 161 . 
87. Ward , M. E. , Woodhouse , A. , Mather , L. E. , et al. ( 1997 ), Morphine pharmacokinetics 
after pulmonary administration from a novel aerosol delivery system , Clin. Pharmacol. 
Ther. , 62 , 596 – 609 . 
88. Mather , L. E. , Woodhouse , A. , Ward , M. E. , Farr , S. J. , Rubsamen , R. A. , and Eltherington , 
L. G. ( 1998 ), Pulmonary administration of aerosolised fentanyl: Pharmacokinetic analysis 
of systemic delivery , Br. J. Clin. Pharmacol. , 46 , 37 – 43 . 
89. Otulana , B. , Okikawa , J. , Linn , L. , Morishige , R. , and Thipphawong , J. ( 2004 ), Safety and 
pharmacokinetics of inhaled morphine delivered using the AERx system in patients 
with moderate - to - severe asthma , Int. J. Clin. Pharmacol. Ther. , 42 , 456 – 462 . 
90. Fulda , G. J. , Giberson , F. , and Fagraeus , L. ( 2005 ), A prospective randomized trial of 
nebulized morphine compared with patient - controlled analgesia morphine in the management 
of acute thoracic pain , J. Trauma , 59 , 383 – 388 ; discussion 389 – 390. 
91. Kohler , D. (1994), Aerosolized heparin, J. Aerosol Med. , 7 , 307 – 314 . 
92. Bendstrup , K. E. , Chambers , C. B. , Jensen , J. I. , and Newhouse , M. T. ( 1999 ), Lung deposition 
and clearance of inhaled (99m)Tc - heparin in healthy volunteers , Am. J. Respir. Crit. 
Care Med. , 160 , 1653 – 1658 . 
93. Qi , Y. , Zhao , G. , Liu , D. , et al. ( 2004 ), Delivery of therapeutic levels of heparin and low - 
molecular - weight heparin through a pulmonary route , Proc. Natl. Acad. Sci. U S A , 101 , 
9867 – 9872 . 
94. Olschewski , H. , Rohde , B. , Behr , J. , et al. ( 2003 ), Pharmacodynamics and pharmacokinetics 
of inhaled iloprost, aerosolized by three different devices, in severe pulmonary 
hypertension , Chest , 124 , 1294 – 1304 . 
95. Ganderton , D. ( 1997 ), General factors infl uencing drug delivery to the lung , Respir. Med. , 
91(Suppl. A), 13 – 16. 
96. Buck , H. ( 2001 ), The ideal inhaler for asthma therapy , Med. Device Technol. , 12 , 
24 – 27 . 
97. Virchow , J. C. ( 2005 ), What plays a role in the choice of inhaler device for asthma 
therapy ? Curr. Med. Res. Opin. , 21 ( Suppl. 4 ), S19 – S25 . 
98. Rau , J. L. ( 2005 ), Determinants of patient adherence to an aerosol regimen , Respir. Care , 
50 , 1346 – 1356 ; discussion 1357 – 1359. 
99. Ganderton , D. ( 1999 ), Targeted delivery of inhaled drugs: Current challenges and future 
goals , J. Aerosol Med. , 12 ( Suppl. 1 ), S3 – S8 . 

100. Dolovich , M. B. , Ahrens , R. C. , Hess , D. R. , et al. ( 2005 ), Device selection and outcomes 
of aerosol therapy: Evidence - based guidelines: American College of Chest Physicians/ 
American College of Asthma, Allergy, and Immunology , Chest , 127 , 335 – 371 . 
101. O ’ Byrne , P. M. ( 1995 ), Clinical comparisons of inhaler systems: What are the important 
aspects ? J. Aerosol Med. , 8 ( Suppl. 3 ), S39 – 46 ; discussion S47. 
102. O ’ Callaghan , C. , and Barry , P. W. ( 2000 ), How to choose delivery devices for asthma , 
Arch. Dis. Child. , 82 , 185 – 187 . 
103. Newhouse , M. ( 1991 ), Advantages of pressurized canister metered dose inhalers , 
J. Aerosol Med. , 4 , 139 – 150 . 
104. Crompton , G. K. ( 1990 ), The adult patient ’ s diffi culties with inhalers , Lung , 168 (Suppl)., 
658 – 662 . 
105. Harnor , K. J. , Perkins , A. C. , Wastie , M. , et al. ( 1993 ), Effect of vapor pressure on the 
deposition pattern from solution phase metered dose inhalers , Int. J. Pharm. , 95 , 
111 – 116 . 
106. Leach , C. L. , Davidson , P. J. , Hasselquist , B. E. , and Boudreau , R. J. ( 2002 ), Lung deposition 
of hydrofl uoroalkane - 134a beclomethasone is greater than that of chlorofl uorocarbon 
fl uticasone and chlorofl uorocarbon beclomethasone: A cross - over study in healthy 
volunteers , Chest , 122 , 510 – 516 . 
107. Leach , C. L. , Davidson , P. J. , Hasselquist , B. E. , and Boudreau , R. J. ( 2005 ), Infl uence of 
particle size and patient dosing technique on lung deposition of HFA - beclomethasone 
from a metered dose inhaler , J. Aerosol Med. , 18 , 379 – 385 . 
108. Leach , C. L. ( 1998 ), Improved delivery of inhaled steroids to the large and small airways , 
Respir. Med. , 92(Suppl. A), 3 – 8. 
109. Williams , R. O. , III , and Liu , J. ( 1998 ), Infl uence of formulation additives on the vapor 
pressure of hydrofl uoroalkane propellants , Int. J. Pharm. , 166 , 99 – 103 . 
110. Newman , S. P. , Moren , F. , Pavia , D. , Corrado , O. , and Clarke , S. W. ( 1982 ), The effects of 
changes in metered volume and propellant vapor pressure on the deposition of pressurized 
inhalation aerosols , Int. J. Pharm. , 11 , 337 – 344 . 
111. Brambilla , G. , Ganderton , D. , Garzia , R. , Lewis , D. , Meakin , B. , and Ventura , P. ( 1999 ), 
Modulation of aerosol clouds produced by pressurised inhalation aerosols , Int. J. Pharm. , 
186 , 53 – 61 . 
112. Vervaet , C. , and Byron , P. R. ( 1999 ), Drug - surfactant - propellant interactions in HFA - 
formulations , Int. J. Pharm. , 186 , 13 – 30 . 
113. Steckel , H. , and Wehle , S. ( 2004 ), A novel formulation technique for metered dose 
inhaler (MDI) suspensions , Int. J. Pharm. , 284 , 75 – 82 . 
114. Berry , J. , Kline , L. C. , Sherwood , J. K. , et al. ( 2004 ), Infl uence of the size of micronized 
active pharmaceutical ingredient on the aerodynamic particle size and stability of a 
metered dose inhaler , Drug Dev. Ind. Pharm. , 30 , 705 – 714 . 
115. Berry , J. , Kline , L. , Naini , V. , Chaudhry , S. , Hart , J. , and Sequeira , J. ( 2004 ), Infl uence of 
the valve lubricant on the aerodynamic particle size of a metered dose inhaler , Drug. 
Dev. Ind. Pharm. , 30 , 267 – 275 . 
116. Berry , J. , Heimbecher , S. , Hart , J. L. , and Sequeira , J. ( 2003 ), Infl uence of the metering 
chamber volume and actuator design on the aerodynamic particle size of a metered dose 
inhaler , Drug. Dev. Ind. Pharm. , 29 , 865 – 876 . 
117. Dunbar , C. A. ( 1998 ), Atomization mechanisms of the pressurized metered dose inhaler , 
Part. Sci. Technol. , 15 , 253 – 271 . 
118. Gabrio , B. J. , Stein , S. W. , and Velasquez , D. J. ( 1999 ), A new method to evaluate plume 
characteristics of hydrofl uoroalkane and chlorofl uorocarbon metered dose inhalers , Int. 
J. Pharm. , 186 , 3 – 12 . 
REFERENCES 717

718 AEROSOL DRUG DELIVERY 
119. Lewis , D. A. , Ganderton , D. , Meakin , B. J. , and Brambilla , G. ( 2004 ), Theory and practice 
with solution systems , in: Dalby , R. N. , Byron , P. R. , Peart , J. , Suman , J.D. , and Farr , S. J. , 
eds., Respiratory Drug Delivery IX , Davis Healthcare International , River Grove, IL , 
pp. 109 – 115 . 
120. Polli , G. P. , Grim , W. M. , Bacher , F. A. , and Yunker , M. H. ( 1969 ), Infl uence of formulation 
on aerosol particle size , J. Pharm. Sci. , 58 , 484 – 486 . 
121. Stein , S. W. , and Myrdal , P. B. ( 2004 ), A theoretical and experimental analysis of formulation 
and device parameters affecting solution MDI size distributions , J. Pharm. Sci. , 
93 , 2158 – 2175 . 
122. Purewal , T. S. , and Grant , D. J. W. , Eds. ( 1998 ), Metered Dose Inhaler Technology , CRC 
Press , Boca Raton . 
123. Rogers , D. F. , and Ganderton , D. ( 1995 ), Determining equivalence of inhaled medications, 
consensus statement from a workshop of the British association for lung research, 
held at Royal Brompton National Heart & Lung Institute, London 24 June 1994 , Respir. 
Med. , 89 , 253 – 261 . 
124. Meakin , B. J. , Lewis , D. A. , Ganderton , D. , and Brambilla , G. ( 2000 ), Countering challenges 
posed by mimicry of CFC performance using HFA systems , in: Dalby , R. N. , 
Byron , P. R. , Peart , J. , and Farr , S. J. , Eds., Respiratory Drug Delivery VII , Serentec , 
Raleigh, NC , pp. 99 – 107 . 
125. Smyth , H. D. , and Leach , C. L. ( 2005 ), Alternative propellant aerosol delivery systems , 
Crit. Rev. Ther. Drug Carrier Syst. , 22 , 493 – 534 . 
126. Pritchard , J. N. , and Genova , P. ( 2006 ), Adapting the pMDI to deliver novel drugs: Insulin 
and beyond , in: Dalby , R. N. , Byron , P. R. , Peart , J. , Suman , J. D. , and Farr , S. J. , Eds., 
Respiratory Drug Delivery , 2006, Davis Healthcare International , River Grove, IL , 
pp. 133 – 141 . 
127. Jannick , P. ( 2006 ), CFC phase - out scenarios of pressurized metered dose inhalers: 
Current status , in: Dalby , R. N. , Byron , P. R. , Peart , J. , Suman , J. D. , and Farr , S. J. , Eds., 
Respiratory Drug Delivery , 2006, Davis Healthcare International , River Grove, IL , 
pp. 789 – 792 . 
128. Anonymous ( 2006 ), A levalbuterol metered - dose inhaler (xopenex HFA) for asthma , 
Med. Lett. Drugs Ther. , 48, 21 – 22 , 24 . 
129. Byron , P. R. ( 1994 ), Dosing reproducibility from experimental albuterol suspension 
metered - dose inhalers , Pharm. Res. , 11 , 580 – 584 . 
130. Tansey , I. ( 1997 ), The technical transition to CFC - free inhalers , Br. J. Clin. Pract. Suppl. , 
89 , 22 – 27 . 
131. Stefely , J. D. , Duan , D. C. , Myrdal , P. B. , Ross , D. L. , Schultz , D. W. , and Leach , C. L. 
( 2000 ), Design and utility of a novel class of biocompatible excipients for HFA - based 
MDIs , in: Dalby , R. N. , Byron , P. R. , Peart , J. , and Farr , S. J. , Eds., Respiratory Drug 
Delivery VII , Serentec Press , Raleigh, NC , pp. 83 – 90 . 
132. Stein , S. W. , and Myrdal , P. B. ( 2006 ), The relative infl uence of atomization and evaporation 
on metered dose inhaler drug delivery effi ciency , Aerosol Sci. Tech. , 40 , 335 – 347 . 
133. Leach , C. L. , Davidson , P. J. , and Boudreau , R. J. ( 1998 ), Improved airway targeting 
with the CFC - free HFA - beclomethasone metered - dose inhaler compared with CFC - 
beclomethasone , Eur. Respir. J. , 12 , 1346 – 1353 . 
134. Seale , J. P. , and Harrison , L. I. ( 1998 ), Effect of changing the fi ne particle mass of inhaled 
beclomethasone dipropionate on intrapulmonary deposition and pharmacokinetics , 
Respir. Med. , 92(Suppl. A), 9 – 15. 
135. Berry , J. , Kline , L. C. , Hart , J. L. , and Sequeira , J. ( 2003 ), Infl uence of the storage orientation 
on the aerodynamic particle size of a suspension metered dose inhaler containing 
propellant HFA - 227 , Drug Dev. Ind. Pharm. , 29 , 631 – 639 . 

136. Anonymous (2005), Tetrafl uoroethane , Pharm. Forum. , 31, 1672 – 1679 . 
137. Ashurst , I. C. , Schultz , R. D. , and Shurkus , D. D. ( 1996 ), Optimization and scale - up of a 
new pressure fi lling manufacturing process for albuterol/HFA 134a inhaler , in: Byron , 
P. R. , Dalby , R. N. , and Farr , S. J. , Eds., Respiratory Drug Delivery V , Interpharm , Buffalo 
Grove, IL , pp. 294 – 296 . 
138. Wilkinson , A. ( 1998 ), The manufacture of metered dose inhalers , in: Purewal , T. S. , and 
Grant , D. J. W. , Eds., Metered Dose Inhaler Technology , CRC Press , Boca Raton, F.L. , 
pp. 69 – 116 . 
139. Gupta , A. , Stein , S. W. , and Myrdal , P. B. ( 2003 ), Balancing ethanol cosolvent concentration 
with product performance in 134a - based pressurized metered dose inhalers , 
J. Aerosol Med. , 16 , 167 – 174 . 
140. Tiwari , D. , Goldman , D. , Dixit , S. , Malick , W. A. , and Madan , P. L. ( 1998 ), Compatibility 
evaluation of metered - dose inhaler valve elastomers with tetrafl uoroethane (P134a), a 
non - CFC propellant , Drug Dev. Ind. Pharm. , 24 , 345 – 352 . 
141. Brouet , G. , Robins , E. , Hall , S. , Butterworth , G. , Hemy , J. , and Turner , R. ( 2006 ), Developing 
new container closure options: A suppliers perpective , in: Dalby , R. N. , Byron , 
P. R. , Peart , J. , Suman , J. D. , and Farr , S. J. , Eds., Respiratory Drug Delivery 2006 , Davis 
Healthcare International , River Grove, IL , pp. 111 – 120 . 
142. Leone , P. (2006), Review of elastomer materials used in metering valves for pharmaceutical 
applications, paper presented at the International Rubber Conference IRC, Lyon , 
pp. 1 – 14. 
143. Burel , S. , Brouet , G. , and Grandsire , T. ( 2004 ), Moisture uptake in pMDIs: The effect on 
valve component materials , paper presented at Drug Delivery to the Lungs XV, London , 
pp. 297 – 300 . 
144. Williams , R. O. III , and Hu , C. ( 2000 ), Moisture uptake and its infl uence on pressurized 
metered - dose inhalers , Pharm. Dev. Technol. , 5 , 153 – 162 . 
145. Williams , R. O. III , and Hu , C. ( 2001 ), Infl uence of water on the solubility of two steroid 
drugs in hydrofl uoroalkane (HFA) propellants , Drug Dev. Ind. Pharm. , 27 , 71 – 79 . 
146. Ross , R. N. ( 1997 ), Loss of bronchodilator medication in priming a conventional metered 
dose inhaler: A cost of treating asthma , Med. Interface , 10 , 141 – 146 . 
147. Smyth , H. , Brace , G. , Barbour , T. , Gallion , J. , Grove , J. , and Hickey , A. J. ( 2006 ), Spray 
pattern analysis for metered dose inhalers: Effect of actuator design , Pharm. Res. , 23 , 
1591 – 1596 . 
148. Smyth , H. D. , Brace , G. , Barbour , T. , Gallion , J. , Grove , J. , and Hickey , A. ( 2006 ), Actuator 
design variables for particle size modulation , in: Dalby , R. N. , Byron , P. R. , Peart , J. , 
Suman , J. D. , and Farr , S. J. , Eds., Respiratory Drug Delivery 2006 , Davis Healthcare 
International , River Grove, IL , pp. 857 – 860 . 
149. Peart , J. , Orban , J. C. , McGlynn , P. , Redmon , M. , Sargeant , C. M. , and Byron , P. R. ( 2002 ), 
MDI electrostatics: Valve and formulation interactions which really make a difference , 
in: Dalby , R. N. , Byron , P. R. , Peart , J. , and Farr , S. J. , Eds., Respiratory Drug Delivery 
XIII , Davis Horwood International , Raleigh, NC , pp. 223 – 230 . 
150. Kwok , P. C. , Glover , W. , and Chan , H. K. ( 2005 ), Electrostatic charge characteristics of 
aerosols produced from metered dose inhalers , J. Pharm. Sci. , 94 , 2789 – 2799 . 
151. Newman , S. P. , Weisz , A. W. , Talaee , N. , and Clarke , S. W. ( 1991 ), Improvement of drug 
delivery with a breath actuated pressurised aerosol for patients with poor inhaler technique 
, Thorax , 46 , 712 – 716 . 
152. Woodman , K. , Bremner , P. , Burgess , C. , Crane , J. , Pearce , N. , and Beasley , R. ( 1993 ), A 
comparative study of the effi cacy of beclomethasone dipropionate delivered from a 
breath activated and conventional metered dose inhaler in asthmatic patients , Curr. Med. 
Res. Opin. , 13 , 61 – 69 . 
REFERENCES 719

720 AEROSOL DRUG DELIVERY 
153. Terzano , C. ( 2001 ), Pressurized metered dose inhalers and add - on devices , Pulm. Pharmacol. 
Ther. , 14 , 351 – 366 . 
154. Asmus , M. J. , Liang , J. , Coowanitwong , I. , and Hochhaus , G. ( 2004 ), In vitro performance 
characteristics of valved holding chamber and spacer devices with a fl uticasone metered - 
dose inhaler , Pharmacotherapy , 24 , 159 – 166 . 
155. Goldberg , S. , Einot , T. , Algur , N. , et al. ( 2002 ), Adrenal suppression in asthmatic children 
receiving low - dose inhaled budesonide: Comparison between dry powder inhaler and 
pressurized metered - dose inhaler attached to a spacer , Ann. Allergy Asthma Immunol. , 
89 , 566 – 571 . 
156. Dalby , R. N. , Somaraju , S. , Chavan , V. S. , and Jarvis , D. ( 1998 ), Evaluation of aerosol drug 
output from the OptiChamber and AeroChamber spacers in a model system , J. Asthma. , 
35 , 173 – 177 . 
157. Newman , S. P. , Millar , A. B. , Lennard - Jones , T. R. , Moren , F. , and Clarke , S. W. ( 1984 ), 
Improvement of pressurised aerosol deposition with nebuhaler spacer device , Thorax , 
39 , 935 – 941 . 
158. Hindle , M. , and Chrystyn , H. ( 1994 ), Relative bioavailability of salbutamol to the lung 
following inhalation using metered dose inhalation methods and spacer devices , Thorax , 
49 , 549 – 553 . 
159. Wildhaber , J. H. , Waterer , G. W. , Hall , G. L. , and Summers , Q. A. ( 2000 ), Reducing electrostatic 
charge on spacer devices and bronchodilator response , Br. J. Clin. Pharmacol. , 
50 , 277 – 280 . 
160. Pierart , F. , Wildhaber , J. H. , Vrancken , I. , Devadason , S. G. , and Le Souef , P. N. ( 1999 ), 
Washing plastic spacers in household detergent reduces electrostatic charge and greatly 
improves delivery , Eur. Respir. J. , 13 , 673 – 678 . 
161. Coppolo , D. P. , Mitchell , J. P. , and Nagel , M. W. ( 2006 ), Levalbuterol aerosol delivery 
with a nonelectrostatic versus a nonconducting valved holding chamber , Respir. Care , 
51 , 511 – 514 . 
162. Smyth , H. D. , Beck , V. P. , Williams , D. , and Hickey , A. J. ( 2004 ), The infl uence of formulation 
and spacer device on the in vitro performance of solution chlorofl uorocarbon - free 
propellant - driven metered dose inhalers , AAPS PharmSciTech , 5 , E7 . 
163. Rahmatalla , M. F. , Zuberbuhler , P. C. , Lange , C. F. , and Finlay , W. H. ( 2002 ), In vitro effect 
of a holding chamber on the mouth - throat deposition of QVAR (hydrofl uoroalkane - 
beclomethasone dipropionate) , J. Aerosol Med. , 15 , 379 – 385 . 
164. Sander , N. , Fusco - Walkert , S. J. , Harder , J. M. , and Chipps , B. E. ( 2006 ), Dose counting 
and the use of pressurized metered - dose inhalers: Running on empty , Ann. Allergy 
Asthma Immunol. , 97 , 34 – 38 . 
165. Bradshaw , D. R. S. ( 2006 ), Developing dose counters: An appraisal based on regulator, 
pharma, and user needs , in: Dalby , R. N. , Byron , P. R. , Peart , J. , Suman , J. D. , and Farr , 
S. J. , Eds., Respiratory Drug Delivery 2006 , Davis Healthcare International , River Grove, 
IL , pp. 121 – 131 . 
166. Vidgren , M. , Paronen , P. , Vidgren , P. , Vainio , P. , and Nuutinen , J. ( 1990 ), In vivo evaluation 
of the new multiple dose powder inhaler and the rotahaler using the gamma scintigraphy 
, Acta Pharm. Nord. , 2 , 3 – 10 . 
167. Smith , I. J. , and Parry - Billings , M. ( 2003/2004 ), The inhalers of the future? A review of 
dry powder devices on the market today , Pulm. Pharmacol. Ther. , 16 , 79 – 95 . 
168. Telko , M. J. , and Hickey , A. J. ( 2005 ), Dry powder inhaler formulation , Respir. Care , 50 , 
1209 – 1227 . 
169. Clarke , M. J. , Peart , J. , Cagnani , S. , and Byron , P. R. ( 2002 ), Adhesion of powders for 
inhalation: An evaluation of drug detachment from surfaces following deposition from 
aerosol streams , Pharm. Res. , 19 , 322 – 329 . 

170. Ward , G. H. , and Schultz , R. K. ( 1995 ), Process - induced crystallinity changes in albuterol 
sulfate and its effect on powder physical stability , Pharm. Res. , 12 , 773 – 779 . 
171. Giry , K. , Pean , J. M. , Giraud , L. , Marsas , S. , Rolland , H. , and Wuthrich , P. ( 2006 ), Drug/ 
lactose co - micronization by jet milling to improve aerosolization properties of a powder 
for inhalation , Int. J. Pharm , 321 , 162 – 166 . 
172. Rasenack , N. , and Muller , B. W. ( 2004 ), Micron - size drug particles: Common and novel 
micronization techniques , Pharm. Dev. Technol. , 9 , 1 – 13 . 
173. Steckel , H. , Rasenack , N. , Villax , P. , and Muller , B. W. ( 2003 ), In vitro characterization 
of jet - milled and in - situ - micronized fl uticasone - 17 - propionate , Int. J. Pharm. , 258 , 65 – 
75 . 
174. Edwards , D. A. , Ben - Jebria , A. , and Langer , R. ( 1998 ), Recent advances in pulmonary 
drug delivery using large, porous inhaled particles , J. Appl. Physiol. , 85 , 379 – 385 . 
175. White , S. , Bennett , D. B. , and Cheu , S. , et al. ( 2005 ), EXUBERA: Pharmaceutical development 
of a novel product for pulmonary delivery of insulin , Diabetes Technol. Ther. , 7 , 
896 – 906 . 
176. Vanbever , R. , Mintzes , J. D. , Wang , J. , et al. ( 1999 ), Formulation and physical characterization 
of large porous particles for inhalation , Pharm. Res. , 16 , 1735 – 1742 . 
177. Dunbar , C. , Scheuch , G. , Sommerer , K. , DeLong , M. , Verma , A. , and Batycky , R. ( 2002 ), 
In vitro and in vivo dose delivery characteristics of large porous particles for inhalation , 
Int. J. Pharm. , 245 , 179 – 189 . 
178. Bartus , R. T. , Emerich , D. , Snodgrass - Belt , P. , et al. ( 2004 ), A pulmonary formulation of 
l - dopa enhances its effectiveness in a rat model of parkinson ’ s disease , J. Pharmacol. 
Exp. Ther. , 310 , 828 – 835 . 
179. Dellamary , L. A. , Tarara , T. E. , Smith , D. J. , et al. ( 2000 ), Hollow porous particles in 
metered dose inhalers , Pharm. Res. , 17 , 168 – 174 . 
180. Duddu , S. P. , Sisk , S. A. , Walter , Y. H. , et al. ( 2002 ), Improved lung delivery from a passive 
dry powder inhaler using an engineered PulmoSphere powder , Pharm. Res. , 19 , 
689 – 695 . 
181. Pfutzner , A. , Mann , A. E. , and Steiner , S. S. ( 2002 ), Technosphere/Insulin — A new 
approach for effective delivery of human insulin via the pulmonary route , Diabetes 
Technol. Ther. , 4 , 589 – 594 . 
182. Pfutzner , A. , Flacke , F. , Pohl , R. , et al. ( 2003 ), Pilot study with technosphere/PTH 
(1 - 34) — A new approach for effective pulmonary delivery of parathyroid hormone (1 - 
34) , Horm. Metab. Res. , 35 , 319 – 323 . 
183. Young , P. M. , and Price , R. ( 2004 ), The infl uence of humidity on the aerosolisation of 
micronised and SEDS produced salbutamol sulphate , Eur. J. Pharm. Sci. , 22 , 235 – 240 . 
184. Rehman , M. , Shekunov , B. Y. , York , P. , et al. Optimisation of powders for pulmonary 
delivery using supercritical fl uid technology , Eur. J. Pharm. Sci. , 22 , 1 – 17 . 
185. Lobo , J. M. , Schiavone , H. , Palakodaty , S. , York , P. , Clark , A. , and Tzannis , S. T. ( 2005 ), 
SCF - engineered powders for delivery of budesonide from passive DPI devices , J. Pharm. 
Sci. , 94 , 2276 – 2288 . 
186. Schiavone , H. , Palakodaty , S. , Clark , A. , York , P. , and Tzannis , S. T. ( 2004 ), Evaluation of 
SCF - engineered particle - based lactose blends in passive dry powder inhalers , Int. J. 
Pharm. , 281 , 55 – 66 . 
187. Pandey , R. , Sharma , A. , Zahoor , A. , Sharma , S. , Khuller , G. K. , and Prasad , B. ( 2003 ), 
Poly ( dl - lactide - co - glycolide) nanoparticle - based inhalable sustained drug delivery 
system for experimental tuberculosis , J. Antimicrob. Chemother. , 52 , 981 – 986 . 
188. Dickinson , P. A. , Howells , S. W. , and Kellaway , I. W. ( 2001 ), Novel nanoparticles for pulmonary 
drug administration , J. Drug Target. , 9 , 295 – 302 . 
REFERENCES 721

722 AEROSOL DRUG DELIVERY 
189. Sham , J. O. , Zhang , Y. , Finlay , W. H. , Roa , W. H. , and Lobenberg , R. ( 2004 ), Formulation 
and characterization of spray - dried powders containing nanoparticles for aerosol delivery 
to the lung , Int. J. Pharm. , 269 , 457 – 467 . 
190. Begat , P. , Morton , D. A. , Staniforth , J. N. , and Price , R. ( 2004 ), The cohesive - adhesive 
balances in dry powder inhaler formulations I: Direct quantifi cation by atomic force 
microscopy , Pharm. Res. , 21 , 1591 – 1597 . 
191. Davies , M. , Brindley , A. , Chen , X. , et al. ( 2005 ), Characterization of drug particle surface 
energetics and Young ’ s modulus by atomic force microscopy and inverse gas chromatography 
, Pharm. Res. , 22 , 1158 – 1166 . 
192. Larhrib , H. , Zeng , X. M. , Martin , G. P. , Marriott , C. , and Pritchard , J. ( 1999 ), The use of 
different grades of lactose as a carrier for aerosolised salbutamol sulphate , Int. J. Pharm. , 
191 , 1 – 14 . 
193. Zeng , X. M. , Pandhal , K. H. , and Martin , G. P. ( 2000 ), The infl uence of lactose carrier on 
the content homogeneity and dispersibility of beclomethasone dipropionate from dry 
powder aerosols , Int. J. Pharm. , 197 , 41 – 52 . 
194. Lucas , P. , Anderson , K. , and Staniforth , J. N. ( 1998 ), Protein deposition from dry powder 
inhalers: Fine particle multiplets as performance modifi ers , Pharm. Res. , 15 , 562 – 569 . 
195. Begat , P. , Morton , D. A. , Staniforth , J. N. , and Price , R. ( 2004 ), The cohesive - adhesive 
balances in dry powder inhaler formulations II: Infl uence on fi ne particle delivery characteristics 
, Pharm. Res. , 21 , 1826 – 1833 . 
196. Jones , M. , and Price , R. ( 2006 ), The infl uence of fi ne excipient particles on the performance 
of carrier - based dry powder inhalation formulations , Pharm. Res. , 23 , 
1665 – 1674 . 
197. Ming Zeng , X. , Martin , G. P. , Marriott , C. , and Pritchard , J. ( 2001 ), The use of lactose 
recrystallised from carbopol gels as a carrier for aerosolised salbutamol sulphate , Eur. 
J. Pharm. Biopharm. , 51 , 55 – 62 . 
198. Flament , M. P. , Leterme , P. , and Gayot , A. ( 2004 ), The infl uence of carrier roughness on 
adhesion, content uniformity and the in vitro deposition of terbutaline sulphate from 
dry powder inhalers , Int. J. Pharm. , 275 , 201 – 209 . 
199. Zeng , X. M. , Martin , G. P. , Marriott , C. , and Pritchard , J. ( 2001 ), Lactose as a carrier in 
dry powder formulations: The infl uence of surface characteristics on drug delivery , 
J. Pharm. Sci. , 90 , 1424 – 1434 . 
200. El - Sabawi , D. , Price , R. , Edge , S. , and Young , P. M. ( 2006 ), Novel temperature controlled 
surface dissolution of excipient particles for carrier based dry powder inhaler formulations 
, Drug Dev. Ind. Pharm. , 32 , 243 – 251 . 
201. Steckel , H. , and Bolzen , N. ( 2004 ), Alternative sugars as potential carriers for dry powder 
inhalations , Int. J. Pharm. , 270 , 297 – 306 . 
202. Lucas , P. , Anderson , K. , Potter , U. J. , and Staniforth , J. N. ( 1999 ), Enhancement of small 
particle size dry powder aerosol formulations using an ultra low density additive , Pharm. 
Res. , 16 , 1643 – 1647 . 
203. Hindle , M. , Jashnani , R. N. , and Byron , P. R. ( 1994 ), Dose emissions from marketed dry 
powder inhalers: Infl uence of fl ow, volume and environment , in: Byron , P. R. , Dalby , R. 
N. , and Farr , S. J. , Eds., Respiratory Drug Delivery IV , Interpharm , Buffalo Grove, IL , 
pp. 137 – 142 . 
204. Borgstrom , L. , Asking , L. , and Lipniunas , P. ( 2005 ), An in vivo and in vitro comparison 
of two powder inhalers following storage at hot/humid conditions , J. Aerosol Med. , 18 , 
304 – 310 . 
205. Price , R. , Young , P. M. , Edge , S. , and Staniforth , J. N. ( 2002 ), The infl uence of relative 
humidity on particulate interactions in carrier - based dry powder inhaler formulations , 
Int. J. Pharm. , 246 , 47 – 59 . 

206. Young , P. M. , Price , R. , Tobyn , M. J. , Buttrum , M. , and Dey , F. ( 2003 ), Investigation into 
the effect of humidity on drug - drug interactions using the atomic force microscope , 
J. Pharm. Sci. , 92 , 815 – 822 . 
207. Borgstrom , L. , Asking , L. , and Thorsson , L. ( 2005 ), Idealhalers or realhalers? A comparison 
of diskus and turbuhaler , Int. J. Clin. Pract. , 59 , 1488 – 1495 . 
208. Fuller , R. (1995), The diskus: A new multi -dose powder device — Effi cacy and comparison 
with turbuhaler , J. Aerosol Med. , 8 ( Suppl. 2 ), S11 – 17 . 
209. Bennett , F. S. , Carter , P. A. , Rowley , G. , and Dandiker , Y. ( 1999 ), Modifi cation of electrostatic 
charge on inhaled carrier lactose particles by addition of fi ne particles , Drug 
Dev. Ind. Pharm. , 25 , 99 – 103 . 
210. Byron , P. R. , Peart , J. , and Staniforth , J. N. ( 1997 ), Aerosol electrostatics. I: Properties of 
fi ne powders before and after aerosolization by dry powder inhalers , Pharm. Res. , 14 , 
698 – 705 . 
211. Carter , P. A. , Rowley , G. , Fletcher , E. J. , and Stylianopoulos , V. (1998), Measurement of 
electrostatic charge decay in pharmaceutical powders and polymer materials used in dry 
powder inhaler devices , Drug Dev. Ind. Pharm. , 24 , 1083 – 1088 . 
212. Murtomaa , M. , Mellin , V. , Harjunen , P. , Lankinen , T. , Laine , E. , and Lehto , V. P. ( 2004 ), 
Effect of particle morphology on the triboelectrifi cation in dry powder inhalers , Int. J. 
Pharm. , 282 , 107 – 114 . 
213. Taylor , A. , and Gustafsson , P. ( 2005 ), Do all dry powder inhalers show the same pharmaceutical 
performance ? Int. J. Clin. Pract. Suppl. , 59 , 7 – 12 . 
214. Cegla , U. H. ( 2004 ), Pressure and inspiratory fl ow characteristics of dry powder inhalers , 
Respir. Med. , 98(Suppl. A), S22 – 28. 
215. Hindle , M. , and Byron , P. R. ( 1995 ), Dose emissions from marketed dry powder inhalers , 
Int. J. Pharm. , 116 , 169 – 177 . 
216. Hindle , M. , and Byron , P. R. ( 1996 ), Impaction and impingement techniques for powder 
inhalers — Comparisons, problems and validation , in: Byron , P. R. , Dalby , R. N. , and Farr , 
S. J. , Eds., Respiratory Drug Delivery V , Interpharm, Buffalo Grove, IL , pp. 263 – 272 . 
217. Pitcairn , G. R. , Lim , J. , Hollingworth , A. , and Newman , S. P. ( 1997 ), Scintigraphic assessment 
of drug delivery from the ultrahaler dry powder inhaler , J. Aerosol Med. , 10 , 
295 – 306 . 
218. Pedersen , S. , Hansen , O. R. , and Fuglsang , G. ( 1990 ), Infl uence of inspiratory fl ow rate 
upon the effect of a turbuhaler , Arch. Dis. Child. , 65 , 308 – 310 . 
219. Tukiainen , H. , and Terho , E. O. ( 1985 ), Comparison of inhaled salbutamol powder and 
aerosol in asthmatic patients with low peak expiratory fl ow level , Eur. J. Clin. Pharmacol. 
, 27 , 645 – 647 . 
220. Borgstrom , L. ( 1994 ), Deposition patterns with turbuhaler , J. Aerosol Med. , 7 , S49 – 53 . 
221. Finlay , W. H. , and Gehmlich , M. G. ( 2000 ), Inertial sizing of aerosol inhaled from two 
dry powder inhalers with realistic breath patterns versus constant fl ow rates , Int. J. 
Pharm. , 210 , 83 – 95 . 
222. Borgstrom , L. , Bengtsson , T. , Derom , E. , and Pauwels , R. ( 2000 ), Variability in lung 
deposition of inhaled drug, within and between asthmatic patients, with a pMDI and a 
dry powder inhaler, turbuhaler , Int. J. Pharm. , 193 , 227 – 230 . 
223. Nielsen, K. G. , Skov , M. , Klug , B. , Ifversen, M. , and Bisgaard, H. (1997), Flow- dependent 
effect of formoterol dry - powder inhaled from the aerolizer , Eur. Respir. J. , 10 , 2105 – 
2109 . 
224. Koskela , T. , Malmstrom , K. , Sairanen , U. , Peltola , S. , Keski - Karhu , J. , and Silvasti , M. 
( 2000 ), Effi cacy of salbutamol via easyhaler unaffected by low inspiratory fl ow , Respir. 
Med. , 94 , 1229 – 1233 . 
REFERENCES 723

724 AEROSOL DRUG DELIVERY 
225. Newhouse , M. T. , Nantel , N. P. , Chambers , C. B. , Pratt , B. , and Parry - Billings , M. ( 1999 ), 
Clickhaler (a novel dry powder inhaler) provides similar bronchodilation to pressurized 
metered - dose inhaler, even at low fl ow rates , Chest , 115 , 952 – 956 . 
226. Meakin , B. J. , Ganderton , D. , Panza , I. , and Ventura , P. ( 1998 ), The effect of fl ow rate on 
drug delivery from the pulvinal, a high - resistance dry powder inhaler , J. Aerosol Med. , 
11 , 143 – 152 . 
227. Newman , S. P. , Moren , F. , Trofast , E. , Talaee , N. , and Clarke , S. W. (1989), Deposition and 
clinical effi cacy of terbutaline sulphate from turbuhaler, a new multi - dose powder 
inhaler , Eur. Respir. J. , 2 , 247 – 252 . 
228. Kohler , D. ( 2004 ), The novolizer: Overcoming inherent problems of dry powder inhalers , 
Respir. Med. , 98(Suppl. A), S17 – 21. 
229. O ’ Connor , B. J. ( 2004 ), The ideal inhaler: Design and characteristics to improve outcomes 
, Respir. Med. , 98(Suppl. A), S10 – 16. 
230. Lim , J. G. , Shah , B. , Rohatagi , S. , and Bell , A. ( 2006 ), Development of a dry powder 
inhaler, the ultrahaler, containing triamcinolone acetonide using in vitro – in vivo relationships 
, Am. J. Ther. , 13 , 32 – 42 . 
231. McCormack , P. L. , and Plosker , G. L. ( 2006 ), Inhaled mometasone furoate: A review of 
its use in persistent asthma in adults and adolescents , Drugs , 66 , 1151 – 1168 . 
232. Pitcairn , G. R. , Lankinen , T. , Seppala , O. P. , and Newman , S. P. ( 2000 ), Pulmonary drug 
delivery from the taifun dry powder inhaler is relatively independent of the patient ’ s 
inspiratory effort , J. Aerosol Med. , 13 , 97 – 104 . 
233. Keating , G. M. , and Faulds , D. ( 2002 ), Airmax: A multi - dose dry powder inhaler , Drugs , 
62 , 1887 – 1895 ; discussion 1896 – 1897. 
234. Iida , K. , Leuenberger , H. , Fueg , L. M. , Muller - Walz , R. , Okamoto , H. , and Danjo , K. 
( 2000 ), Effect of mixing of fi ne carrier particles on dry powder inhalation property of 
salbutamol sulfate (SS) , Yakugaku Zasshi , 120 , 113 – 119 . 
235. Patton , J. S. , Fishburn , C. S. , and Weers , J. G. ( 2004 ), The lungs as a portal of entry for 
systemic drug delivery , Proc. Am. Thorac. Soc. , 1 , 338 – 344 . 
236. Patton , J. S. ( 2005 ), Unlocking the opportunity of tight glycaemic control: Innovative 
delivery of insulin via the lung , Diabetes Obes. Metab. , 7 ( Suppl. 1 ), S5 – 8 . 
237. Rosenstock , J. , Cappelleri , J. C. , Bolinder , B. , and Gerber , R. A. ( 2004 ), Patient satisfaction 
and glycemic control after 1 year with inhaled insulin (exubera) in patients with 
type 1 or type 2 diabetes , Diabetes Care , 27 , 1318 – 1323 . 
238. Bellary , S. , and Barnett , A. H. ( 2006 ), Inhaled insulin: New technology, new possibilities , 
Int. J. Clin. Pract. , 60 , 728 – 734 . 
239. Dunn , C. , and Curran , M. P. ( 2006 ), Inhaled human insulin (exubera): A review of its 
use in adult patients with diabetes mellitus , Drugs , 66 , 1013 – 1032 . 
240. Patton , J. S. , Bukar , J. G. , and Eldon , M. A. ( 2004 ), Clinical pharmacokinetics and pharmacodynamics 
of inhaled insulin , Clin. Pharmacokinet. , 43 , 781 – 801 . 
241. Quattrin , T. , Belanger , A. , Bohannon , N. J. , Schwartz , S. L. , and Exubera Phase III Study 
Group ( 2004 ), Effi cacy and safety of inhaled insulin (exubera) compared with subcutaneous 
insulin therapy in patients with type 1 diabetes: Results of a 6 - month, randomized, 
comparative trial , Diabetes Care , 27 , 2622 – 2627 . 
242. Barnett , A. H. , Dreyer , M. , Lange , P. , and Serdarevic - Pehar , M. ( 2006 ), An open, randomized, 
parallel - group study to compare the effi cacy and safety profi le of inhaled human 
insulin (exubera) with glibenclamide as adjunctive therapy in patients with type 2 diabetes 
poorly controlled on metformin , Diabetes Care , 29 , 1818 – 1825 . 
243. Rosenstock , J. , Zinman , B. , Murphy , L. J. , et al. ( 2005 ), Inhaled insulin improves glycemic 
control when substituted for or added to oral combination therapy in type 2 diabetes: 
A randomized, controlled trial , Ann. Intern. Med. , 143 , 549 – 558 . 

244. DeFronzo , R. A. , Bergenstal , R. M. , Cefalu , W. T. , et al. ( 2005 ), Effi cacy of inhaled insulin 
in patients with type 2 diabetes not controlled with diet and exercise: A 12 - week, randomized, 
comparative trial , Diabetes Care , 28 , 1922 – 1928 . 
245. Rave , K. , Bott , S. , Heinemann , L. , et al. ( 2005 ), Time - action profi le of inhaled insulin in 
comparison with subcutaneously injected insulin lispro and regular human insulin , Diabetes 
Care , 28 , 1077 – 1082 . 
246. Davidson , M. B. , Mehta , A. E. , and Siraj , E. S. ( 2006 ), Inhaled human insulin: An inspiration 
for patients with diabetes mellitus ? Cleve. Clin. J. Med. , 73 , 569 – 578 . 
247. An , B. , and Reinhardt , R. R. ( 2003 ), Effects of different durations of breath holding after 
inhalation of insulin using the AERx insulin diabetes management system , Clin. Ther. , 
25 , 2233 – 2244 . 
248. Boyd , B. , Noymer , P. , Liu , K. , et al. ( 2004 ), Effect of gender and device mouthpiece shape 
on bolus insulin aerosol delivery using the AERx pulmonary delivery system , Pharm. 
Res. , 21 , 1776 – 1782 . 
249. Cramer , J. A. , Okikawa , J. , Bellaire , S. , and Clauson , P. ( 2004 ), Compliance with inhaled 
insulin treatment using the AERx iDMS insulin diabetes management system , Diabetes 
Technol. Ther. , 6 , 800 – 807 . 
250. Farr , S. J. , McElduff , A. , Mather , L. E. , et al. ( 2000 ), Pulmonary insulin administration 
using the AERx system: Physiological and physicochemical factors infl uencing insulin 
effectiveness in healthy fasting subjects , Diabetes Technol. Ther. , 2 , 185 – 197 . 
251. Thipphawong , J. , Otulana , B. , Clauson , P. , Okikawa , J. , and Farr , S. J. ( 2002 ), Pulmonary 
insulin administration using the AERx insulin diabetes system , Diabetes Technol. Ther. , 
4 , 499 – 504 . 
252. Ferron , G. A. , Kerrebijn , K. F. , and Weber , J. ( 1976 ), Properties of aerosols produced 
with three nebulizers , Am. Rev. Respir. Dis. , 114 , 899 – 908 . 
253. Nikander , K. , Turpeinen , M. , and Wollmer , P. ( 1999 ), The conventional ultrasonic nebulizer 
proved ineffi cient in nebulizing a suspension , J. Aerosol Med. , 12 , 47 – 53 . 
254. Rau , J. L. ( 2002 ), Design principles of liquid nebulization devices currently in use , Respir. 
Care , 47 , 1257 – 1275 ; discussion 1275 – 1278. 
255. Nerbrink , O. , and Dahlback , M. ( 1994 ), Basic nebulizer function , J. Aerosol Med. , 7 , 
S7 – 11 . 
256. Nerbrink , O. , Dahlback , M. , and Hansson , H. C. ( 1994 ), Why do medical nebulizers differ 
in their output and particle size characteristics ? J. Aerosol Med. , 7 , 259 – 276 . 
257. Newnham , D. M. , and Lipworth , B. J. ( 1994 ), Nebuliser performance, pharmacokinetics, 
airways and systemic effects of salbutamol given via a novel nebuliser delivery system 
( “ ventstream ” ) , Thorax , 49 , 762 – 770 . 
258. Leung , K. , Louca , E. , and Coates , A. L. ( 2004 ), Comparison of breath - enhanced 
to breath - actuated nebulizers for rate, consistency, and effi ciency , Chest , 126 , 1619 – 
1627 . 
259. Barry , P. W. , and O ’ Callaghan , C. ( 1999 ), An in vitro analysis of the output of salbutamol 
from different nebulizers , Eur. Respir. J. , 13 , 1164 – 1169 . 
260. O ’ Callaghan , C. , White , J. , Jackson , J. , Barry , P. W. , and Kantar , A. ( 2005 ), Delivery of 
nebulized budesonide is affected by nebulizer type and breathing pattern , J. Pharm. 
Pharmacol. , 57 , 787 – 790 . 
261. Rau , J. L. , Ari , A. , and Restrepo , R. D. ( 2004 ), Performance comparison of 
nebulizer designs: Constant - output, breath - enhanced, and dosimetric , Respir. Care , 49 , 
174 – 179 . 
262. Raabe , O. G. , Wong , T. M. , Wong , G. B. , Roxburgh , J. W. , Piper , S. D. , and Lee , J. I. ( 1998 ), 
Continuous nebulization therapy for asthma with aerosols of beta2 agonists , Ann. Allergy 
Asthma Immunol. , 80 , 499 – 508 . 
REFERENCES 725

726 AEROSOL DRUG DELIVERY 
263. Dolovich , M. B. ( 2002 ), Assessing nebulizer performance , Respir. Care , 47 , 1290 – 1301 ; 
discussion 1301 – 1304. 
264. Le Brun , P. P. , de Boer , A. H. , Heijerman , H. G. , and Frijlink , H. W. ( 2000 ), A review of 
the technical aspects of drug nebulization , Pharm. World Sci. , 22 , 75 – 81 . 
265. Byrne , N. M. , Keavey , P. M. , Perry , J. D. , Gould , F. K. , and Spencer , D. A. ( 2003 ), Comparison 
of lung deposition of colomycin using the HaloLite and the pari LC plus nebulisers 
in patients with cystic fi brosis , Arch. Dis. Child. , 88 , 715 – 718 . 
266. Denyer , J. , Nikander , K. , and Smith N. J. ( 2004 ), Adaptive aerosol delivery (AAD) technology 
, Expert Opin. Drug Deliv. , 1 , 165 – 176 . 
267. Nikander , K. , Arheden , L. , Denyer , J. , and Cobos , N. ( 2003 ), Parents ’ adherence with 
nebulizer treatment of their children when using an adaptive aerosol delivery (AAD) 
system , J. Aerosol Med. , 16 , 273 – 281 . 
268. Knoch , M. , and Keller , M. ( 2005 ), The customised electronic nebuliser: A new category 
of liquid aerosol drug delivery systems , Expert Opin. Drug Deliv. , 2 , 377 – 390 . 
269. Kim , D. , Mudaliar , S. , Chinnapongse , S. , et al. ( 2003 ), Dose - response relationships of 
inhaled insulin delivered via the aerodose insulin inhaler and subcutaneously injected 
insulin in patients with type 2 diabetes , Diabetes Care , 26 , 2842 – 2847 . 
270. Lipworth , B. J. , Sims , E. J. , Taylor , K. , Cockburn , W. , and Fishman , R. ( 2005 ), Dose - 
response to salbutamol via a novel palm sized nebuliser (aerodose inhaler), conventional 
nebuliser (pari LC plus) and metered dose inhaler (ventolin evohaler) in moderate to 
severe asthmatics , Br. J. Clin. Pharmacol. , 59 , 5 – 13 . 
271. Hindle , M. ( 2004 ), Soft mist inhalers: A review of current technology , The Drug Delivery 
Companies Report , 13 , 31 – 34 . 
272. Geller , D. E. ( 2002 ), New liquid aerosol generation devices: Systems that force pressurized 
liquids through nozzles , Respir. Care , 47 , 1392 – 1404 ; discussion 1404 – 1405. 
273. Patel , K. R. , Pavia , D. , Lowe , L. , and Spiteri , M. ( 2006 ), Inhaled ethanolic and aqueous 
solutions via respimat soft mist inhaler are well - tolerated in asthma patients , Respiration , 
73 , 434 – 440 . 
274. Voshaar , T. , Hausen , T. , Kardos , P. , et al. ( 2005 ), Inhalation therapy with respimat soft 
inhaler in patients with COPD and asthma , Pneumologie , 59 , 25 – 32 . 
275. Kassner , F. , Hodder , R. , and Bateman , E. D. ( 2004 ), A review of ipratropium 
bromide/fenoterol hydrobromide (berodual) delivered via respimat soft mist inhaler in 
patients with asthma and chronic obstructive pulmonary disease , Drugs , 64 , 1671 – 
1682 . 
276. Pitcairn , G. , Reader , S. , Pavia , D. , and Newman , S. ( 2005 ), Deposition of corticosteroid 
aerosol in the human lung by respimat soft mist inhaler compared to deposition by 
metered dose inhaler or by turbuhaler dry powder inhaler , J. Aerosol Med. , 18 , 
264 – 272 . 
277. Vincken , W. , Dewberry , H. , and Moonen , D. ( 2003 ), Fenoterol delivery by respimat soft 
mist inhaler versus CFC metered dose inhaler: Cumulative dose - response study in 
asthma patients , J. Asthma , 40 , 721 – 730 . 
278. Newman , S. P. , Brown , J. , Steed , K. P. , Reader , S. J. , and Kladders , H. ( 1998 ), Lung deposition 
of fenoterol and fl unisolide delivered using a novel device for inhaled medicines: 
Comparison of RESPIMAT with conventional metered - dose inhalers with and without 
spacer devices , Chest , 113 , 957 – 963 . 
279. Newman, S. P. , Steed, K. P. , Reader , S. J. , Hooper , G. , and Zierenberg , B. (1996), Effi cient 
delivery to the lungs of fl unisolide aerosol from a new portable hand - held multidose 
nebulizer , J. Pharm. Sci. , 85 , 960 – 964 . 

280. Schurmann , W. , Schmidtmann , S. , Moroni , P. , Massey , D. , and Qidan , M. ( 2005 ), Respimat 
soft mist inhaler versus hydrofl uoroalkane metered dose inhaler: Patient preference and 
satisfaction , Treat. Respir. Med. , 4 , 53 – 61 . 
281. Pavia , D. , and Moonen , D. ( 1999 ), Preliminary data from phase II studies with respimat, 
a propellant - free soft mist inhaler , J. Aerosol Med. , 12 ( Suppl. 1 ), S33 – 39 . 
282. Dalby , R. , Spallek , M. , and Voshaar , T. ( 2004 ), A review of the development of respimat 
soft mist inhaler , Int. J. Pharm. , 283 , 1 – 9 . 
283. Hochrainer , D. , Holz , H. , Kreher , C. , Scaffi di , L. , Spallek , M. , and Wachtel , H. ( 2005 ), 
Comparison of the aerosol velocity and spray duration of respimat soft mist inhaler and 
pressurized metered dose inhalers , J. Aerosol Med. , 18 , 273 – 282 . 
284. Zierenberg , B. ( 1999 ), Optimizing the in vitro performance of respimat , J. Aerosol Med. , 
12 ( Suppl. 1 ), S19 – 24 . 
285. Farr , S. J. , Warren , S. J. , Lloyd , P. , et al. ( 2000 ), Comparison of in vitro and in vivo effi - 
ciencies of a novel unit - dose liquid aerosol generator and a pressurized metered dose 
inhaler , Int. J. Pharm. , 198 , 63 – 70 . 
286. Hermansen , K. , Ronnemaa , T. , Petersen , A. H. , Bellaire , S. , and Adamson , U. ( 2004 ), 
Intensive therapy with inhaled insulin via the AERx insulin diabetes management 
system: A 12 - week proof - of - concept trial in patients with type 2 diabetes , Diabetes Care , 
27 , 162 – 167 . 
287. Geller , D. , Thipphawong , J. , Otulana , B. , et al. ( 2003 ), Bolus inhalation of rhDNase with 
the AERx system in subjects with cystic fi brosis , J. Aerosol Med. , 16 , 175 – 182 . 
288. Zimlich , W. C. , Ding , J. Y. , Busick , D. R. , et al. ( 2000 ), The development of a novel electrohydrodynamic 
pulmonary drug delivery device , in Dalby , R. N. , Byron , P. R. , Peart , 
J. , and Farr , S. J. , Eds., Respiratory Drug Delivery VII , Serentec , Raleigh, NC , pp. 
241 – 246 . 
289. Shen , X. , Hindle , M. , and Byron P. R. ( 2004 ), Effect of energy on propylene glycol aerosols 
using the capillary aerosol generator , Int. J. Pharm. , 275 , 249 – 258 . 
290. Rabinowitz , J. D. , Lloyd , P. M. , Munzar , P. , et al. ( 2006 ), Ultra - fast absorption of amorphous 
pure drug aerosols via deep lung inhalation , J. Pharm. Sci , 95 , 2438 – 2451 . 
291. Rabinowitz , J. D. , Wensley , M. , Lloyd , P. , et al. ( 2004 ), Fast onset medications through 
thermally generated aerosols , J. Pharmacol. Exp. Ther. , 309 , 769 – 775 . 
REFERENCES 727


729 
5.9 
OCULAR DRUG DELIVERY 
Ilva D. Rupenthal and Raid G. Alany 
The University of Auckland, Auckland, New Zealand 
Contents 
5.9.1 Introduction 
5.9.2 Challenges in Ocular Drug Delivery 
5.9.2.1 Anatomical and Physiological Considerations 
5.9.2.2 Pharmacokinetic Considerations 
5.9.2.3 Formulation Considerations 
5.9.3 Formulation Approaches to Improve Ocular Bioavailability 
5.9.3.1 Conventional Dosage Forms 
5.9.3.2 Polymeric Delivery Systems 
5.9.3.3 Colloidal Delivery Systems 
5.9.3.4 Other Delivery Approaches 
5.9.4 Conclusion 
References 
5.9.1 INTRODUCTION 
Due to the accessibility of the eye surface, topical administration of ophthalmic 
medications is the most common method for treating conditions affecting the exterior 
eye surface. However, the unique anatomy and physiology of the eye renders 
it diffi cult to achieve an effective drug concentration at the target site. Therefore, 
effi cient delivery of a drug past the protective ocular barriers accompanied with 
minimization of its systemic side effects remains a major challenge. 
Conventional eye drops currently account for more than 90% of the marketed 
ophthalmic formulations [1] . However, after instillation of an eye drop, typically less 
than 5% of the applied drug penetrates the cornea and reaches the intraocular 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

730 OCULAR DRUG DELIVERY 
tissues. This is due to the rapid and extensive precorneal loss caused by drainage 
and high tear fl uid turnover. 
As a consequence, the typical corneal contact time is limited to 1 – 2 min and the 
ocular bioavailability is usually less than 10% [2] . Furthermore, there is an initial 
peak dose of the drug, which is usually higher than that needed for a therapeutic 
effect, followed by a sharp drop - off in concentration to subtherapeutic levels. 
Various ocular delivery systems, such a ointments, suspensions, micro - and nanocarriers, 
and liposomes, have been investigated during the past two decades pursuing 
two main strategies: to increase the corneal permeability and to prolong the contact 
time on the ocular surface [3] . 
On the other hand, the most effi cient method for drug delivery to the posterior 
chamber of the eye so far has been intravitreal injection. This chapter focuses on 
the topical application of drugs to the surface of the eye and discusses the most 
recent formulation approaches in this area. 
5.9.2 CHALLENGES IN OCULAR DRUG DELIVERY 
5.9.2.1 Anatomical and Physiological Considerations 
In order to research and develop an effective ophthalmic delivery system, a good 
understanding of the anatomy and physiology of the eye (the globe) is necessary. 
Figure 1 shows a cross section through the human eye. This chapter will mainly focus 
on the precorneal area and the transport barriers present in the eye. 
FIGURE 1 Cross section of the human eye and cornea. 

Precorneal Area 
Precorneal Tear Film Corneal transparency and good visual function require a 
uniform eye surface. This is achieved by the tear fi lm, which covers and lubricates 
the cornea and the external globe. It is about 7 – 8 . m thick and is the fi rst structure 
encountered by topically applied drugs. The trilaminar structure of the tear fi lm is 
shown in Figure 2 . 
Attached to the glycocalix of the corneal/conjunctival surface is a mucous layer, 
which consists mainly of glycoproteins. This layer is produced by the conjunctival 
goblet cells and the lacrimal gland. It plays an important role in the stability of the 
tear fi lm as well as in the wetting of the corneal and conjunctival epithelium. The 
middle aqueous layer constitutes about 98% of the tear fi lm and is mostly secreted 
by the main and accessory lacrimal glands [4] . It is composed of water, electrolytes, 
and various proteins such as lipocalin, lysozyme, and lactoferrin [5 – 8] . The outermost 
lipid layer is derived from the Meibomian and sebaceous Zeiss glands and 
prevents the evaporation of the tear fl uid. It consists of sterol esters, triacylglycerols, 
and phospholipids and is spread over the aqueous layer during blinking. 
Nasolacrimal Drainage System Figure 3 illustrates the nasolacrimal drainage 
system. The lacrimal gland, which is situated in the superior temporal angle of the 
orbit, is responsible for most of the tear fl uid secretion. Secreted fl uid is spread over 
the surface of the cornea during blinking and ends up in the puncta when the upper 
eye lid approaches the lower lid. The blinking process creates a suction mechanism 
which results in tears fl owing through the lacrimal canaliculi into the lacrimal sac. 
Fluid from the lacrimal sac then drains into the 12 - mm - long nasolacrimal duct, 
which empties into the inferior nasal passage. This passage is a highly vascular area 
FIGURE 2 Structure of the precorneal tear fi lm. 
CHALLENGES IN OCULAR DRUG DELIVERY 731

732 OCULAR DRUG DELIVERY 
and is responsible for most of the systemic drug absorption and subsequent systemic 
side effects. 
The cul - de - sac normally holds 7 – 9 . L of tear fl uid, with the normal tear fl ow rate 
being 1.2 – 1.5 . L/min [4] . The loss from the precorneal area by drainage, tear fl uid 
turnover, and noncorneal absorption plays an important role in determining the 
ocular bioavailability of a drug. As the drainage rate is much faster than the ocular 
absorption rate, most of the topically applied drug is eliminated from the precorneal 
area within the fi rst minute [4] . 
Tear production can be divided into basal, refl ex, and emotional tearing [9] . 
Refl ex tearing can be induced by many pharmaceutical/formulation factors, including 
the drug itself as well as pH and tonicity of the ocular dosage form. 
Transport Barriers in the Eye Topical administration is the most common route 
for ocular drug delivery. Consequently, the cornea, conjunctiva, and sclera form the 
most essential barriers for drug penetration into the intraocular tissues. 
Cornea The cornea is an important mechanical barrier protecting the intraocular 
tissues. It is considered to be the main pathway for ocular penetration of topically 
applied drugs. However, due to its unique structure, with the hydrophilic stroma 
sandwiched between the highly lipophilic epithelium and the less lipophilic endothelium, 
the penetration of compounds through the cornea depends on their n - 
octanol – water partition coeffi cient. Only drugs with a partition coeffi cient between 
10 and 100 that show both lipid - and water - soluble properties can readily pass 
through the cornea. 
The cornea is composed of fi ve layers (see Figure 1 ): epithelium, Bowman ’ s 
membrane stroma, Descemet ’ s membrane, and endothelium. 
The epithelium is the outermost layer and consists of fi ve to six cell layers. These 
can be subdivided into one to two outermost layers of fl attened superfi cial cells with 
microvilli on their anterior surface enhancing the cohesion and stability of the tear 
FIGURE 3 Schematic of the nasolacrimal drainage system. 

fi lm [10] , two to three layers of polygonal wing cells, and a single layer of basal 
columnar cells, allowing for minimal paracellular transport. The superfi cial cells 
adhere to one another via desmosomes and the cells are encircled by tight junctions 
[11] . Due to the nature of the tight junctions, the epithelium represents the rate - 
limiting barrier for hydrophilic compounds. 
Bowman ’ s membrane is composed of a layer of collagen fi bers which form a relatively 
tight and impermeable barrier against microorganisms and therefore protect 
the stroma. 
The stroma makes up approximately 90% of the corneal thickness and is mainly 
composed of hydrated collagen fi brils. It is highly hydrophilic, porous, and can be 
considered as an open structure, as it allows free passage of hydrophilic substances 
with a molecular weight below 500,000 Da but acts as a diffusion barrier to all lipophilic 
drugs [4, 12] . 
Descemet ’ s membrane is the basement membrane of the endothelial cells. It is 
comprised of collagen fi bers arranged in a hexagonal pattern and embedded in a 
matrix. 
The endothelium is a single layer of fl attened polygonal cells with microvilli 
which increase the surface area for removal of waste and absorption of nutrients 
[12] . It plays an important role in the maintenance of corneal hydration and transparency 
via active ion and fl uid transport mechanisms. 
Since the cornea exhibits hydrophilic as well as lipophilic characteristics, it represents 
an effective barrier for diffusion of both hydrophilic and lipophilic substances 
[3, 13] . 
Conjunctiva The conjunctiva is a thin and vascular mucous membrane consisting 
of two to three layers of epithelial cells overlying a loose, highly vascular connective 
tissue. The tight junctions present on the apical surface of the epithelium act as the 
main barrier for drug penetration (molecules > 20,000 Da) across the tissue, although 
not as tight as the corneal epithelium, which is impermeable to molecules larger 
than 5000 Da [14, 15] . The conjunctiva covers the anterior surface of the globe 
(bulbar conjunctiva), with the exception of the cornea, and is folded at the fornix 
(fornix conjunctiva) to form the palpebral conjunctiva, which lines the inner surface 
of the eyelids. The bulbar conjunctiva represents the fi rst barrier for permeation of 
topically applied drugs via the noncorneal route [16] . 
Sclera The sclera is the outermost fi rm coat of the eye that serves as a protective 
barrier for the sensitive inner parts. It is composed of the same type of collagen 
fi bers as the corneal stroma. However, the fi bers are arranged in an irregular network 
rather than a lattice pattern, which makes the tissue appear opaque compared to 
the transparent cornea. The white sclera constitutes the posterior fi ve - sixths of the 
globe, whereas the transparent cornea comprises the anterior one - sixth [17] . 
Iris – Ciliary Body The ciliary body comprises the ciliary muscle, which mainly 
enables accommodation, and the ciliary processes, which produce the aqueous 
humor. 
The iris separates the anterior and posterior chambers and consists of the pigmented 
epithelial cell layer, the iridial sphincter and dilator muscles, and the stroma. 
The amount of melanin present in the stroma determines the color of the iris: few 
CHALLENGES IN OCULAR DRUG DELIVERY 733

734 OCULAR DRUG DELIVERY 
melanocytes exhibit blue, grey, or green, while many melanocytes are responsible 
for the brown appearance of the iris. There is often a considerable quantitative difference 
in drug response between light and heavily pigmented eyes [18] . The binding 
of drugs with melanin can decrease the aqueous humor concentration of free drug 
and is therefore likely to reduce the pharmacological response [19] . 
Lens The lens is the transparent biconvex structure situated behind the iris and 
in front of the vitreous. It plays an important role in the visual function of the eye 
and also enables accommodation together with the ciliary muscle. The lens is made 
up of slightly more than 30% protein (water - soluble crystallins) and therefore has 
the highest protein content of all tissues in the body [20] . The lens receives its nutrients 
from the aqueous humor and its transparency depends on the geometry of the 
lens fi bres. 
Blood – Ocular Barriers The blood – ocular barriers can be divided into the blood – 
aqueous barrier and the blood – retinal barrier. 
The blood – aqueous barrier is located in the anterior part of the eye and is formed 
by the endothelial cells of the blood vessels in the iris and the nonpigmented cell 
layer of the ciliary epithelium [21] . It regulates the solute exchange between the 
blood and the intraocular fl uid, preventing unspecifi c passage of solutes that could 
infl uence the transparency of the ocular tissues. The outward movement into the 
systemic blood circulation is less restricted, allowing especially small and lipophilic 
drug molecules to enter the uveal blood circulation [22] . These molecules are consequently 
removed more rapidly from the anterior chamber than larger, hydrophilic 
molecules, which are eliminated by the aqueous humor turnover only [23] . 
The blood – retinal barrier can be found in the posterior part of the eye. It prevents 
toxic molecules, plasma components, and water from entering the retina. It also 
forms a barrier for passage of systemically administered drugs into the vitreous, 
typically resulting in only 1 – 2% of the drug ’ s plasma concentration in the intraocular 
tissues [24] . 
5.9.2.2 Pharmacokinetic Considerations 
After topical application of an ophthalmic solution, the solution is instantly mixed 
with the tear fl uid and then spread over the eye surface. However, various precorneal 
factors such as the drainage of the instilled solution, induced lacrimation, 
normal tear turnover, noncorneal absorption, drug metabolism, and enzymatic degradation 
limit the ocular absorption by shortening the contact time of the applied 
drug with the corneal surface [25] . As a result, typically less than 10% of the instilled 
dose is delivered into the intraocular tissues, whereas the rest is absorbed into the 
systemic circulation, leading to various side effects [3, 25, 26] . A summary of the 
drug deposition model in the eye after topical application as described by Lee and 
Robinson [27] is given below. 
Upon instillation, the topically applied drug solution is instantly diluted by the 
resident tears, resulting in a signifi cant decrease in the concentration gradient 
(driving force) and hence in the reduction of the transcorneal fl ux. Drainage of 
lacrimal fl uid towards the nasolacrimal sac during blinking leads to a rapid elimination 
of the ocular solution via the canaliculi. 

Any drug remaining on the ocular surface for a suffi cient period of time can be 
absorbed into the anterior chamber via either the corneal or the conjunctival – scleral 
route. 
Corneal absorption is considered to be the major penetration pathway for topically 
applied drugs. There are two mechanisms for absorption across the corneal 
epithelium, namely transcellular and paracellular diffusion [28] . Lipophilic drugs 
prefer the transcellular route while hydrophilic drugs penetrate primarily via the 
paracellular route. Transcorneal transport includes simple diffusion, facilitated diffusion, 
active transport, and endocytosis. Transport along the paracellular route is 
passive and is only limited by the pore size and the charge of the intracellular spaces. 
Only relatively small molecules can permeate through the pores. Negatively charged 
molecules permeate at a slower rate than positively charged and neutral ones [29] . 
In addition, a positive charge may also decrease the permeation due to the possible 
ionic interaction with the negatively charged carboxylic acid residues of the tight 
junction proteins [30] . 
The noncorneal route of absorption via the conjunctiva and the sclera is usually 
nonproductive, as most of the drug reaches the systemic circulation before gaining 
access to the intraocular tissues. As the surface area of the conjunctiva is much larger 
than that of the cornea and with the highly vascularized conjunctiva being more 
permeable, especially to larger hydrophilic molecules, drug loss through this route 
of absorption may be signifi cant. Both transconjunctival and transnasal absorption 
via the nasolacrimal duct are generally undesirable, not only because of the loss of 
drug into the systemic circulation, but also because of the possible side effects [1] . 
5.9.2.3 Formulation Considerations 
Irritation of the eye following the use of an ocular delivery system can be induced 
by a number of factors, including the instilled volume, the pH, and the osmolality 
of the formulation, as well as by the drug itself [31] . All these factors may induce 
refl ex tearing and as a result increase lacrimal drainage. This is likely to reduce the 
ocular bioavailability of the drug and thus needs to be considered during the formulation 
process. In addition, general safety considerations such as sterility, ocular 
toxicity and irritation, and the amount of preservative used, need to be taken into 
account when formulating an ocular dosage form. 
Physicochemical Drug Properties On the one hand, factors such as the chemical 
nature or the concentration of a drug can cause irritation of the ocular tissues, inducing 
refl ex tearing and therefore reducing the retention time of the formulation in 
front of the eye. 
However, more important physicochemical properties in terms of ocular bioavailability 
are the ones that affect the corneal permeability of the active compound. 
These include the lipophilicity of the drug as refl ected by its n - octanol – water partition 
coeffi cient [32] , the molecular size and shape [33] , the charge [34] , and the 
acid – base properties as determined by its p Ka [35] . 
According to Kaur and Smitha [36] , the optimum lipophilicity for corneal absorption 
is found in drugs with an n - octanol – water partition coeffi cient between 10 and 
100. For drugs with smaller partition coeffi cients (highly hydrophilic drugs), the 
lipophilic epithelium forms the rate - limiting barrier, whereas the hydrophilic stroma 
CHALLENGES IN OCULAR DRUG DELIVERY 735

736 OCULAR DRUG DELIVERY 
represents the primary barrier for transcorneal diffusion of highly lipophilic drugs 
[1, 3, 25] . In general, more lipophilic drugs penetrate via the transcellular pathway 
while the more hydrophilic drugs enter the cornea via the paracellular route. 
In the case of ionized compounds (weak acids and bases), drug permeation 
depends on the chemical equilibrium between the ionized and the un - ionized form, 
both in the delivery system itself and in the lacrimal fl uid. In general, un - ionized 
molecules penetrate lipid membranes more readily than ionized ones. 
Besides lipophilicity and degree of ionization, the charge of the drug molecule 
may have an effect on its penetration. Cationic drugs permeate the cornea more 
easily than anionic compounds, which are repelled by the negative charge of the 
mucin layer on the ocular surface as well as the negatively charged pores present 
in the corneal epithelium [1, 3, 25, 29] . However, a positive charge may also decrease 
permeation in some cases, due to possible ionic interactions between the positively 
charged molecules and the negatively charged carboxylic acid groups of the tight 
junction proteins [21, 30] . 
Finally, the molecular size of the drug has an effect on the corneal absorption. 
The cornea is impermeable to molecules larger than 5000 Da, whereas the conjunctival 
tissues allow compounds of up to 20,000 Da to penetrate [14, 15] . 
Buffer Capacity and pH The normal pH of the tear fl uid is 7.4. Ocular formulations 
should ideally be formulated between pH 7.0 and 7.7 to avoid irritation of the 
eye [31] . However, in most cases the pH necessary for maximal solubility or stability 
of the drug is well outside this range. The tear fl uid has only a limited buffering 
capacity, which is mainly due to the dissolved carbon dioxide and bicarbonate. It is 
therefore recommended to formulate using buffers with a low buffering capacity to 
allow the tears to regain their normal pH more rapidly [31] . 
Instillation Volume The cul - de - sac normally holds 7 – 9 . L [37] , but can momentarily 
accommodate up to 30 . L without overfl owing. Most commercial eye droppers 
however, deliver a volume of approximately 50 . L. The excess volume is rapidly 
removed, either by spillage from the conjunctival sac or through the puncta to the 
nasolacrimal drainage system, until the tears return to their normal volume [38] . 
Chrai et al. [39, 40] determined the infl uence of drop size on the rate of drainage of 
a solution instilled into the conjunctival sac of rabbits. The authors reported that 
the drainage process followed fi rst - order kinetics and found that the rate of solution 
drainage from the conjunctival sac (as refl ected by the elimination rate constant) 
was directly proportional to the instilled volume [40] . Similar observations were 
reported with other dosage forms such as suspensions [41] and liposomes [42] . 
Therefore, keeping the applied dose constant while decreasing the instilled volume 
substantially increases ocular bioavailability and decreases systemic absorption 
[3, 26, 43] . 
Osmotic Pressure The osmolality of the lacrimal fl uid is mainly dependent on the 
number of ions dissolved in the aqueous layer of the tear fi lm and normally ranges 
between 310 and 350 mOsm/kg [44] . When an ophthalmic solution is instilled into 
the eye, it mixes with the tear fl uid, resulting in an osmotic pressure that is dependent 
on the osmolality of the tears as well as that of the formulation and the amount 
of the formulation instilled. In general, hypotonic solutions are better tolerated by 

the ocular tissues than hypertonic ones, which lead to increased lacrimation. If the 
tonicity of the formulation is lower than 260 mOsm/kg or higher than 480 mOsm/kg, 
the formulation becomes irritant [45] , induces refl ex tearing and blinking, and is 
therefore likely to reduce the bioavailability of topically instilled drugs. 
5.9.3 FORMULATION APPROACHES TO 
IMPROVE OCULAR BIOAVAILABILITY 
One of the major problems encountered with topical administration is the rapid 
precorneal loss caused by nasolacrimal drainage and high tear fl uid turnover, 
which leads to drug concentrations of typically less than 10% of the applied drug. 
Approaches to improve the ocular bioavailability have been attempted in two 
directions: to increase the corneal permeability by using penetrations enhancers or 
prodrugs and to prolong the contact time with the ocular surface by using viscosity - 
enhancing or in situ gelling polymers. Table 1 summarizes conventional and novel 
ocular drug delivery approaches. Marketed ophthalmic delivery systems based on 
recent formulation approaches are listed in Table 2 . An optimal ocular delivery 
systems would be administered in the form of an eye drop, causing neither blurred 
vision nor ocular irritancy, and would only need to be instilled once a day [1] . 
5.9.3.1 Conventional Dosage Forms 
Conventional dosage forms such as solutions, suspensions, and ointments account 
for almost 90% of the currently accessible ophthalmic formulations on the market 
[1, 169] . They offer some advantages such as their ease of administration by the 
patient, ease of preparation, and the low production costs. However, there are also 
signifi cant disadvantages associated with the use of conventional solutions in particular, 
including the very short contact time with the ocular surface and the fast 
nasolacrimal drainage, both leading to a poor bioavailability of the drug. Various 
ophthalmic delivery systems have been investigated to increase the corneal permeability 
and prolong the contact time with the ocular surface. However, conventional 
eye drops prepared and administered as aqueous solutions remain the most commonly 
used dosage form in ocular disease management. 
Solutions The reasons behind choosing solutions over other dosage forms are 
their favorable cost advantage, the simplicity of formulation development and production, 
and the high acceptance by patients [170] . However, there are also a few 
drawbacks, such as rapid and extensive precorneal loss, high absorption via the 
conjunctiva and the nasolacrimal duct leading to systemic side effects, as well as the 
increased installation frequency resulting in low patient compliance. 
Some of these problems have been encountered by addition of viscosity - 
enhancing agents such as cellulose derivates, which are believed to increase the 
viscosity of the preparation and consequently reduce the drainage rate. The use of 
viscosity enhancers will be discussed later in this section. 
Suspensions Suspensions of the micronized drug ( < 10 . m) in a suitable aqueous 
vehicle are formulated, where the active compound is water insoluble. This is the 
FORMULATION APPROACHES TO IMPROVE OCULAR BIOAVAILABILITY 737

738 OCULAR DRUG DELIVERY 
TABLE 1 Summary of Conventional and Novel Ocular Drug Delivery Approaches 
Drug 
Formulation 
Approach Polymers/Bases References 
Pilocarpine Viscosity enhancer Methylcellulose [ 40 ] 
Fluorescein Viscosity enhancer + 
collagen shields 
Collagen shields in 
methylcellulose vehicle 
[ 46 ] 
Ciprofl oxacin Viscosity enhancers 
+ penetration 
enhancer 
Carbopol 934P/HPMC + 
dodecylmaltoside 
[ 47 ] 
Pilocarpine Viscosity enhancers, 
mucoadhesives, 
in situ gelling 
systems 
Gellan gum, xanthan gum, HEC, 
HPMC, PVA 
[ 48 ] 
Progesterone Mucoadhesive Cross - linked acrylic acid [ 49 ] 
Levobetaxolol Mucoadhesive Polyacrylic acid [ 50 ] 
Tropicamide Mucoadhesive CMC, HPCL, HPCM, PVP, PVA [ 51 ] 
CMC - Na, HA - Na, PAA [ 52 ] 
Pilocarpine, 
tropicamide 
Mucoadhesive Hyaluronic acid, polyacrylic acid [ 53 ] 
Tertrahydrozoline Mucoadhesive Hyaluronic acid, polyacrylic acid, 
chitosan, gelatin 
[ 54 ] 
Tobramycin Mucoadhesive Chitosan [ 55 ] 
Tobramycin, 
ofl oxacin 
Mucoadhesive Chitosan [ 56 ] 
Ofl oxacin Mucoadhesive N - Trimethyl and N - carboxy - 
methyl chitosan 
[ 57, 58 ] 
Pilocarpine Mucoadhesive Carbopol 934P [ 59 ] 
Carbomer 974P and 1342 [ 60 ] 
PVA, PVP, dextran, HPMC, HEC, 
MC, PAA, Na - hyaluronate, Na - 
alginate, gellan gum, chitosan 
[ 61 ] 
Timolol Mucoadhesive Xyloglucan [ 62 ] 
Xanthan gum [ 63 ] 
Carrageenan, gellan gum [ 64 ] 
Carrageenan, locust bean gum, 
guar gum, xanthan gum, 
scleroglucan, xanthan gum, 
sodium alginate, . - cyclodextrin 
[ 65 ] 
Hyaluronic acid [ 66 ] 
HPMC, PVA, hyaluronic acid [ 67 ] 
Carbomer, hyaluronic acid [ 68 ] 
PAA, PVP [ 69 ] 
In situ gelling system Gellan gum [ 70 – 78 ] 
Indomethacine In situ gelling system Gellan gum [ 79 ] 
Pefl oxacin 
mesylate 
In situ gelling system Gellan gum [ 80 ] 
Gatifl oxacin In situ gelling system Alginate/HPMC [ 81 ] 
Ofl oxacin In situ gelling system Carbopol 940/HPMC [ 82 ] 
Pilocarpine In situ gelling system Alginate [ 83 ] 
Carteolol In situ gelling system Alginate [ 84 ] 
Ciprofl oxacin In situ gelling system Poloxamer/hyaluronic acid [ 85 ] 

Drug 
Formulation 
Approach Polymers/Bases References 
Pilocarpine In situ gelling system Pluronic F127, MC, HPMC [ 86 ] 
Pluronic F127/carbopol [ 87 ] 
Pluronic F127, xyloglycan [ 88 ] 
Timolol In situ gelling system Pluronic F127, MC, HPMC, CMC [ 89 ] 
Doxorubicin In situ gelling system Chitosan/glycerophosphate [ 90 ] 
Pluronic F127 and F68, sodium 
hyaluronate 
[ 91 ] 
Pilocarpine, 
hydrocortisone 
Nanoparticles Gelatin [ 92 ] 
Hydrocortisone Nanoparticles Albumin [ 93 ] 
Pilocarpine Nanoparticles Albumin [ 94, 95 ] 
Poly(methyl)methacrylate – acrylic 
acid copolymer (Piloplex) 
[ 96 – 98 ] 
Cellulose acetate hydrogen 
phthalate (CAP) 
[ 99 – 101 ] 
Betaxolol Nanoparticles Poly - . - caprolactone (PECL), poly 
(isobutyl)cyanoacrylate,polylac 
tic -co - glycolic acid (PLGA) 
[ 102 ] 
Carteolol Nanoparticles Poly - . - caprolactone (PECL) [ 103 ] 
Pilocarpine Nanoparticles Poly(butyl)cyanoacrylate 
(PBCA) 
[ 104 – 107 ] 
Poly(hexyl)cyanoacrylate 
(PHCA) 
[ 104, 106, 
107 ] 
Ciclosporine Nanoparticles Chitosan [ 108 ] 
Indomethacine Nanoparticles Chitosan - and poly - L - lysine - 
coated poly - . - caprolactone 
nanocapsules 
[ 109 ] 
Rhodamine Nanoparticles Chitosan - and PEG - coated poly - 
. - caprolactone nanocapsules 
[ 110 ] 
Aciclovir Nanoparticles PEG - coated PLA nanospheres [ 111 ] 
Epinephrine Nanoparticles Poly - N - isopropylacrylamide [ 112 ] 
Ibuprofen Nanoparticles Eudragit RS100 [ 113, 114 ] 
Flurbiprofen Nanoparticles Eudragit RS100 and RL100 [ 115 ] 
Diclofenac Nanoparticles Eudragit RLPM and RSPM [ 116 ] 
Tobramycin Nanoparticles Various lipids [solid lipid 
nanoparticles (SLN)] 
[ 117 ] 
Gentamicin Microspheres Eudragit RS100 and RL100 [ 118 ] 
Piroxicam Microspheres Pectin [ 119 ] 
Albumin [ 120 ] 
Pilocarpine Liposomes + 
mucoadhesive 
coating 
Carbopol - coated liposomes [ 121, 122 ] 
Radioactive - 
labeled BSA 
Liposomes + 
mucoadhesive 
coating 
Chitosan - coated liposomes [ 123 ] 
Oligonucleotides 
(pdT16) 
Liposomes + in situ 
gelling system 
Liposomes in poloxamer 407 gel [ 124 – 126 ] 
Tropicamide Liposomes + in situ 
gelling system 
Liposomes in polycarbophil gel [ 127 ] 
TABLE 1 Continued 
FORMULATION APPROACHES TO IMPROVE OCULAR BIOAVAILABILITY 739

740 OCULAR DRUG DELIVERY 
Drug 
Formulation 
Approach Polymers/Bases References 
Timolol Niosomes Chitosan - and carbopol - coated 
niosomes 
[ 128 ] 
Indomethacine Submicrometer 
emulsions 
Phospholipids, 
lauroamphodiacetate 
[ 129 ] 
Pilocarpine Submicrometer 
emulsions 
Mono - dodecylphosphoric acid [ 130 ] 
Microemulsions Lecithin - based microemulsions [ 131 ] 
Retinol Microemulsions Tween 60 and 80, soy bean 
lecithin, n - butanol, triacetin, 
PG 
[ 132 ] 
Crodamol EO, Crill 1 and 
Crillet 4 
[ 133 ] 
Epinephrine Prodrug Dipivalyl epinephrine [ 134, 135 ] 
Pilocarpine Prodrug O, O. (Xylylene)bispilocarpic 
acid esters 
[ 136 ] 
Ganciclovir Prodrug Ganciclovir acyl ester [ 137 ] 
Atenolol, 
betaxolol, 
Timolol 
Penetration 
enhancers 
Polyoxyethylene alkyl ethers 
(Brij), bile salts 
[ 138 ] 
Cromoclycin Penetration 
enhancers 
EDTA [ 33 ] 
Carbonic 
anhydrase 
inhibitors 
Cyclodextrins . - Cyclodextrin, 
hydroxypropyl -. - cyclodextrin 
[ 139 ] 
Ciprofl oxacin Cyclodextrins Hydroxypropyl - . - cyclodextrin [ 140 ] 
Pilocarpine Cyclodextrins Hydroxypropyl - . - cyclodextrin [ 141 ] 
Ocular fi lms Hydroxypropyl cellulose [ 142 ] 
Poly(2-hydroxypropyl-methacrylate) [ 143 ] 
Pefl oxacin 
mesylate 
Ocular fi lms HPC, HPMC, PVP, PVA [ 144 ] 
Ocular inserts PVP, Eudragit RS and RL [ 145 ] 
Mitomycin C Ocular inserts Collagen implant [ 146 ] 
Pilocarpine Ocular inserts Collagen shield [ 147 ] 
PAA, PVP, HPMC [ 148 ] 
PVA, glyceryl behenate, xanthan 
gum, carrageenan, HPMC, HA; 
coated with Eudragit RS and 
RL 
[ 149 ] 
Timolol Ocular inserts HPC, coated with Eudragit RS 
and RL 
[ 150 ] 
Tilisolol Ocular inserts Poly(hydroxypropyl-methacrylate) [ 151 ] 
Poly(2 - hydroxypropyl - 
methacrylate), polypropylene 
tape 
[ 152 ] 
Ciprofl oxacin Ocular inserts Sodium alginate, Eudragit, 
polyvinyl acetate 
[ 153 ] 
Pradofl oxacin Ocular inserts Hydrogel coating on thin metallic 
wire (OpthaCoil) 
[ 154 ] 
TABLE 1 Continued

Drug 
Formulation 
Approach Polymers/Bases References 
Fluorescein Ocular inserts HPMC lyophilisate on carrier 
strip 
[ 155 ] 
HPMC lyophilisate on poly(tetra 
fl uoroethylene) carrier strip 
[ 156 ] 
Carbopol 974P, maize starch [ 157, 158 ] 
Gentamicin Ocular inserts CAP, carbomer, HPMC, HPC, EC [ 159, 160 ] 
Oxytetracycline Ocular inserts Silicone, PAA, PMA [ 161 ] 
Oxytetracycline, 
prednisolone, 
gentamicin 
Ocular inserts PEO 400, PEO 900 [ 162 ] 
Ofl oxacin Ocular inserts PEO 200, 400, 900, and 2000 [ 163 ] 
PEO 400, Eudragit L100 [ 164 ] 
PEO 900, PEO 2000, chitosan - 
thiolated PAA 
[ 165 ] 
Fluorescein, 
diclofenac 
Ocular inserts [ 166 ] 
Gentamicin Iontophoresis Hydroxyethyl methacrylate [ 167 ] 
Pilocarpine, 
tropicamide 
Dendrimers Various poly(amidoamine) 
(PAMAM) dendrimers 
[ 168 ] 
TABLE 1 Continued 
case for most of the steroids. It is assumed that the drug particles remain in the 
conjunctival sac, thus promoting a sustained release effect [171] . There have been 
many studies trying to prove this assumption, but none of them has revealed a pronounced 
prolonged release profi le [35, 172, 173] . 
According to Davies [174] , topical ophthalmic suspensions have a number of 
limitations compared to solutions. They need to be adequately shaken before use 
to ensure correct dosing, a process which can result in poor patient compliance. In 
addition, they need to be sterilized, which may cause physical instability of the formulation. 
Furthermore, the amount of drug required to achieve only a moderate 
increase in bioavailability is very high, rendering suspensions expensive in terms of 
their production costs [175] . 
The drug particle size plays the most important role in the formulation process 
of suspensions. Particles greater than 10 . m cause patient discomfort. As they are 
perceived as foreign substances, they cause refl ex tearing in order to eliminate the 
particles from the ocular surface [176] . A study by Schoenwald and Stewart [177] 
showed the infl uence of the particle size of dexamethasone on its bioavailability. 
The in vivo dissolution rate decreased with increasing particle size to the point when 
particles were removed from the conjunctival sac before the dissolution was 
complete. 
As a result, achieving a near - solution state with small particles that are easy to 
resuspend and show minimal sedimentation remains the goal when formulation of 
a suspension is unavoidable [176] . 
FORMULATION APPROACHES TO IMPROVE OCULAR BIOAVAILABILITY 741

742 OCULAR DRUG DELIVERY 
TABLE 2 Marketed Ophthalmic Delivery Systems Based on Recent Formulation 
Approaches 
Formulation 
Approach Polymer/Base Product Company 
Suspensions/ 
microparticulates 
Carbomer ion exchange 
resin 
Betoptic S Alcon 
Ointments Wool fat, paraffi n Polyvisc Alcon 
Liquid paraffi n, white soft 
paraffi n 
LacriLube Allergan 
Viscosity enhancers/ 
mucoadhesives 
Polyethylene glycol (PEG), 
propylene glycol (PG), 
HP-guar 
Systane Alcon 
Dextran, HPMC Bion Tears Alcon 
Carboxymethylcellulose 
sodium (CMC-Na) 
Refresh Celluvisc Allergan 
Refresh Liquigel Allergan 
Polyvinyl alcohol (PVA) Liquifi lm Tears Allergan 
HPMC Lacrigel Sunways 
Carbomer Viscotears Novartis 
Carbomer, polyvinyl 
alcohol (PVA) 
Nyogel Novartis 
Hyaluronic acid (HA) Hy - Drop Bausch & Lomb 
Fidia Oftal 
Sodium hyaluronate Vismed TRB 
Chemedica 
Carbomer Pilopine HS Alcon 
Polyacrylic acid (PAA) Fucithalmic Leo Pharma 
In situ gelling 
systems 
Gellan gum Timoptic XE Merck 
Polycarbophil DuraSite InSite Vision 
Polyacrylic acid, poloxamer Smart Hydrogel Advanced 
Medical 
Solutions 
Prodrugs Dipivefrin hydrochloride 
(epinephrin prodrug) 
Propine Allergan 
Ocular inserts Alginic acid Ocusert Alza 
Hydroxylpropyl cellulose Lacrisert Merck 
Silicone elastomer Ocufi t SR Escalon Medical 
Collagen shield MediLens Chiron 
ProShield Alcon 
Ointments Ointments generally consist of a dissolved or dispersed drug in an 
appropriate vehicle base. They are the most commonly used semisolid preparations 
as they are well tolerated and fairly safe and increase the ocular bioavailability of 
the drug. The instilled ointment breaks up into small oily droplets that remain in 
the cul - de - sac as a drug depot. The drug eventually gets to the ointment – tear interface 
due to the shearing action of the eyelids [178] . 
Sieg and Robinson [35] compared the bioavailability of fl uorometholone in a 
solution, a suspension, and an ointment. They found that the peak concentration 

(cmax ) of the drug in the aqueous humor of rabbits was comparable with all three 
formulations, whereas the time to peak concentration ( tmax ) occurred much later 
with the ointment, leading to a signifi cantly greater total bioavailability of the 
drug. 
Overall, ophthalmic ointments offer the following advantages: reduced dilution 
of the medication via the tear fi lm, resistance to nasolacrimal drainage, and an 
increased precorneal contact time [179, 180] . However, oily viscous preparations for 
ophthalmic use (such as ointments) can cause blurred vision and matting of the 
eyelids and may also be associated with discomfort by the patient as well as occasional 
ocular mucosal irritation. Ointments are therefore generally used in combination 
with eye drops, which can be administered during the day, while the ointment 
is applied at night, when clear vision is not required. 
5.9.3.2 Polymeric Delivery Systems 
Polymeric systems used for ocular drug delivery can be divided into three groups: 
viscosity - enhancing polymers, which simply increase the formulation viscosity, 
resulting in decreased lacrimal drainage and enhanced bioavailability; mucoadhesive 
polymers, which interact with the ocular mucin, therefore increasing the contact 
time with the ocular tissues; and in situ gelling polymers, which undergo sol - to - gel 
phase transition upon exposure to the physiological conditions present in the eye. 
However, there are no defi ned boundaries between the different groups and most 
polymers exhibit more than one of these properties. 
Viscosity -Enhancing Polymers In order to reduce the lacrimal clearance (drainage) 
of ophthalmic solutions, various polymers have been added to increase the 
viscosity of conventional eye drops, prolong precorneal contact time, and subsequently 
improve ocular bioavailability of the drug [40, 51, 181 – 184] . Among the 
range of hydrophilic polymers investigated in the area of ocular drug delivery are 
polyvinyl alcohol (PVA) and polyvinyl pyrrolidone (PVP), cellulose derivates such 
as methylcellulose (MC), and polyacrylic acids (carbopols). 
Chrai and Robinson [40] evaluated the use of an MC solution of pilocarpine in 
albino rabbits and found a decrease in the drainage rate with increasing viscosity. 
Patton and Robinson [185] investigated the relationship between the viscosity and 
the contact time or drainage rate and demonstrated an optimum viscosity of 12 – 
15 cps for an MC solution in rabbits. The infl uence of different polymers on the 
activity of pilocarpine in rabbits and human was reported by Saettone et al. [182] . 
Trueblood et al. [183] used lacrimal microscintigraphy to evaluate the corneal 
contact time for saline, PVA, and hydroxpropyl methylcellulose (HPMC) and 
observed the longest contact time for the formulation with HPMC as a viscosity - 
enhancing agent. 
The ocular shear rate ranges from 0.03 s . 1 during interblinking periods to 4250 – 
28,500 s . 1 during blinking [186] . It has a great infl uence on the rheological properties 
of viscous ocular dosage forms and consequently the bioavailability of the incorporated 
drug [187] . Newtonian systems do not show any real improvement of bioavailability 
below a certain viscosity and blinking becomes painful, followed by refl ex 
tearing, if the viscosity is too high [188] . While the viscosity of Newtonian systems 
is independent from the shear rate, non - Newtonian pseudoplastic or so - called 
FORMULATION APPROACHES TO IMPROVE OCULAR BIOAVAILABILITY 743

744 OCULAR DRUG DELIVERY 
shear - thinning systems exhibit a decrease in viscosity with increasing shear rates. 
This pseudoplastic behavior is favorable for ocular drug delivery systems as it offers 
less resistance to blinking and therefore shows greater acceptance by patients than 
Newtonian systems of the same viscosity [189] . 
Mucoadhesive Polymers Bioadhesion refers to the attachment of a drug molecule 
or a delivery system to a specifi c biological tissue by means of interfacial forces. If 
the surface of the tissue is covered by a mucin fi lm, as is the case for the external 
globe, it is more commonly referred to as mucoadhesion. 
In order to be an effective mucoadhesive excipient, polymers must show one or 
more of the following features [190] : strong hydrogen binding group, strong anionic 
charge, high molecular weight, suffi cient chain fl exibility, surface energy properties 
favoring spreading onto the mucus, and near - zero contact angle to allow maximum 
contact with the mucin coat. 
The most commonly used bioadhesives are macromolecular hydrocolloids with 
numerous hydrophilic functional groups capable of forming hydrogen bonds (such 
as carboxyl, hydroxyl, amide, and sulfate groups) [191] . Hui and Robinson [49] were 
the fi rst to demonstrate the usefulness of bioadhesive polymers in improving the 
ocular bioavailability of progesterone. Saettone et al. [53] evaluated a series of 
bioadhesive dosage forms for ocular delivery of pilocarpine and tropicamide and 
found hyaluronic acid to be the most promising mucoadhesive polymer. Lehr et al. 
[192] suggested that cationic polymers, which are able to interact with the negative 
sialic acid residues of the mucin, would probably show better mucoadhesive properties 
than anionic or neutral polymers. They investigated the polycationic polymer 
chitosan and demonstrated that the mucoadhesive performance of chitosan was 
signifi cantly higher in neutral or slightly alkaline pH as it is present in the tear 
fl uid. 
However, according to Park and Robinson [193] , polyanions are better than 
polycations in terms of binding and potential toxicity. In general, both anionic and 
cationic charged polymers demonstrate better mucoadhesive properties than nonionic 
polymer, such as cellulose derivates or PVA [194, 195] . 
The mechanism of mucoadhesion involves a series of different steps. First, the 
mucoadhesive formulation needs to establish an intimate contact with the corneal 
surface. Prerequisites are either good wetting or swelling of the mucoadhesive 
polymer as well as suffi cient spreading across the cornea. The second stage involves 
the penetration of the mucoadhesive polymer chains into the crevices of the tissue 
surface and also the entanglement with the mucous chains [196] . On a molecular 
level, mucoadhesion is a results of van - der - Waals forces, electrostatic attractions, 
hydrogen bonding, and hydrophobic interactions [36] . 
Mucoadhesive polymers increase the contact time of a formulation with the tear 
fi lm and simulate the continuous delivery of tears due to a high water - restraining 
capacity. As such, they allow a decrease in the instillation frequency compared to 
common eye drops and are therefore useful as artifi cial tear products [1, 197, 198] . 
In Situ Gelling Systems In situ gelling systems are viscous polymer - based liquids 
that exhibit sol - to - gel phase transition on the ocular surface due to change in a 
specifi c physicochemical parameter (ionic strength, temperature, pH, or solvent 
exchange). They are highly advantageous over preformed gels as they can easily be 

instilled in liquid form but are capable of prolonging the residence time of the formulation 
on the surface of the eye due to gelling [199] . 
The principal advantage of in situ gelling systems is the easy, accurate, and reproducible 
administration of a dose compared to the application of preformed gels 
[198] . 
The concept of forming gels in situ (e.g., in the cul - de - sac of the eye) was fi rst 
suggested in the early 1980s, and ever since then various triggers of in situ gelling 
have been further investigated. 
Polymers that may undergo sol - to - gel transition triggered by a change in pH are 
cellulose acetate phthalate (CAP) and cross - linked polyacrylic acid derivates such 
as carbopols, methacrylates and polycarbophils. CAP latex is a free - running solution 
at pH 4.4 which undergoes sol - to - gel transition when the pH is raised to that of the 
tear fl uid. This is due to neutralization of the acid groups contained in the polymer 
chains, which leads to a massive swelling of the particles. The use of pH - sensitive 
latex nanoparticles has been described by Gurny et al. [100, 200] . Carbopols have 
apparent p Ka values in the range of 4 – 5 resulting in rapid gelation due to rise in pH 
after ocular administration. 
Gellan gum is an anionic polysaccharide which undergoes phase transition under 
the infl uence of an increased ionic strength. In fact, the gel strength increases proportionally 
with the amount of mono - or divalent cations present in the tear fl uid. 
As a consequence, the usual refl ex tearing, which leads to a dilution of common 
viscous solutions, further enhances the viscosity of gellan gum formulations due to 
the increased amount of tear fl uid and thus higher cation concentration [201] . 
Several studies have been performed comparing Gelrite formulations (low acetyl 
gellan gum) to conventional ophthalmic solutions of the same active compound. 
Shedden et al. [76] compared the plasma concentrations of timolol following multiple 
applications of Timoptic - XE and a timolol maleate solution. They found that a 
once - daily application of the in situ gelling formulation was suffi cient to reduce the 
intraocular pressure to levels comparable to a twice - daily application of the solution, 
leading to better patient compliance as well as a reduction in systemic side 
effects. 
Poloxamers or pluronics are block copolymers consisting of poly(oxyethylene) 
and poly(oxypropylene) units. They rapidly undergo thermal gelation when the 
temperature is raised to that of the ocular surface (32 ° C), while they remain liquid 
at refrigerator temperature. Poloxamers exhibit surface active properties, but even 
if used in high concentrations (usually between 20 and 30%), Pluronic F127 was 
found no more damaging to the cornea than a physiological saline solution [202] . 
In order to reduce the total polymer concentration and achieve better gelling properties, 
several poloxamer combinations have been tested. Wei et al. [91] used a 
mixture of Pluronic F127 and F68 resulting in a more suitable phase transition 
temperature with a free - fl owing liquid under 25 ° C. 
Combining thermal - with pH - dependent gelation, Kumar et al. [86] developed 
a combination of methylcellulose 15% and carbopol 0.3%. This composition exhibited 
a sol - to - gel transition between 25 and 37 ° C with a pH increase from 4 to 7.4 
[203] . A possible mechanism for the thermal effect could be the decrease in the 
degree of the methylcellulose hydration, while the polyacrylic acid can transform 
into a gel upon an increase in pH due to the buffering properties of the tear 
fl uid [1] . 
FORMULATION APPROACHES TO IMPROVE OCULAR BIOAVAILABILITY 745

746 OCULAR DRUG DELIVERY 
5.9.3.3 Colloidal Delivery Systems 
Colloidal carriers have been investigated as drug delivery systems for the past 30 
years in order to achieve specifi c drug targeting, facilitate the bioavailability of drugs 
through biological membranes, and protect the drug against enzymatic degradation. 
Their use in topical administration and especially in ocular delivery however has 
only been studied for the last two decades [1, 3] . 
Colloidal carriers are small particulate systems ranging in size from 100 to 400 nm. 
As they are usually suspended in an aqueous solution, they can easily be administered 
as eye drops, thus avoiding the potential discomfort resulting from bigger 
particles present in ocular suspensions or from viscous or sticky preparations [38] . 
Most efforts in ophthalmic drug delivery have been made with the aim of increasing 
the corneal penetration of the drug. Calvo et al. [204] have shown that colloidal 
particles are preferably taken up by the corneal epithelium via endocytosis. It has 
also been stated by Lallemand and co - workers [205] , that the cornea acts as a drug 
reservoir, slowly releasing the active compound present in the colloidal delivery 
system to the surrounding ocular tissues. 
Nanoparticles Nanoparticles have been among the most widely studied particulate 
delivery systems over the past three decades. They are defi ned as submicrometer 
- sized polymeric colloidal particles ranging from 10 to 1000 nm in which the drug 
can be dissolved, entrapped, encapsulated, or adsorbed [206] . Depending on the 
preparation process, nanospheres or nanocapsules can be obtained. Nanospheres 
have a matrixlike structure where the drug can either be fi rmly adsorbed at the 
surface of the particle or be dispersed/dissolved in the matrix. Nanocapsules, on the 
other hand, consist of a polymer shell and a core, where the drug can either be dissolved 
in the inner core or be adsorbed onto the surface [207] . 
The fi rst nanoparticulate delivery system studied was Piloplex, consisting of pilocarpine 
ionically bound to poly(methyl)methacrylate – acrylic acid copolymer 
nanoparticles [44] . Klein et al. [1, 98] found that a twice - daily application of Piloplex 
in glaucoma patients was just as effective as three to six instillations of conventional 
pilocarpine eye drops. However, the formulation was never accepted for 
commercialization due to various formulation - related problems, including the nonbiodegradability, 
local toxicity, and diffi culty of preparing a sterile formulation 
[208] . 
Another early attempt to formulate a nanoparticulate system for the delivery of 
pilocarpine was made by Gurny [99] . This formulation was based on pilocarpine 
dispersed in a hydrogen CAP pseudolatex formulation. Gurny and co - workers [101] 
compared the formed nanoparticles to a 0.125% solution of hyaluronic acid some 
years after their fi rst investigation and found that the viscous hyaluronic acid system 
showed a signifi cantly longer retention time in front of the eye than the pseudolatex 
formulation. 
The most commonly used biodegradable polymers in the preparation of nanoparticulate 
systems for ocular drug delivery are poly - alkylcyanoacrylates, poly - . - 
caprolactone, and polylactic - co - glycolic acid copolymers. Marchal - Heussler et al. 
[102, 103] compared the three particulate delivery systems using antiglaucoma drugs 
including betaxolol and cartechol. Results showed that poly - . - caprolactone (nanospheres 
and nanocapsules) exhibited the highest pharmacological activity when 

loaded with betaxolol. It seemed that the higher ocular activity was related to the 
hydrophobic nature of the carrier and that the mechanism of action seemed to be 
directly linked to the agglomeration of the particles in the conjunctival sac [1] . In 
general, nanocapsules displayed a much better effect than nanospheres probably 
due to the fact that the active compound was in its un - ionized form in the oily core 
and could diffuse faster into the cornea. Diffusion of the drug from the oily core of 
the nanocapsule to the corneal epithelium seems to be more effective than diffusion 
from the internal, more hydrophilic matrix of the nanospheres [1, 209] . 
In order to achieve a sustained drug release and a prolonged therapeutic activity, 
nanoparticles must be retained in the cul - de - sac and the entrapped drug must be 
released from the particles at a certain rate. If the release is too fast, there is no 
sustained release effect. If it is too slow, the concentration of the drug in the tears 
might be too low to achieve penetration into the ocular tissues [208] . The major 
limiting issues for the development of nanoparticles include the control of particle 
size and drug release rate as well as the formulation stability. 
So far, there is only one microparticulate ocular delivery system on the market. 
Betoptic S is obtained by binding of betaxolol to ion exchange resin particles. Betoptic 
S 0.25% was found to be bioequivalent to the Betoptic 0.5% solution in lowering 
the intraocular pressure [208] . 
Liposomes Liposomes were fi rst reported by Bangham in the 1960s and have been 
investigated as drug delivery systems for various routes ever since then. They offer 
some promising features for ophthalmic drug delivery as they can be administered 
as eye drops but will localize and maintain the pharmacological activity of the drug 
at its site of action [1] . Due to the nature of the lipids used, conventional liposomes 
are completely biodegradable, biocompatible, and relatively nontoxic [1] . 
A liposome or so - called vesicle consists of one or more concentric spheres of 
lipid bilayers separated by water compartments with a diameter ranging from 80 nm 
to 100 . m. Owing to their amphiphilic nature, liposomes can accommodate both 
lipohilic (in the lipid bilayer) and hydrophilic (encapsulated in the central aqueous 
compartment) drugs [207] . According to their size, liposomes are classifi ed as either 
small unilamellar vesicles (SUVs) (10 – 100 nm) or large unilamellar vesicles (LUVs) 
(100 – 300 nm). If more than one bilayer is present, they are referred to as multilamellar 
vesicles (MLVs). Depending on their lipid composition, they can have a positive, 
negative, or neutral surface charge. 
Liposomes are potentially valuable as ocular drug delivery systems due to their 
simplicity of preparation and versatility in physical characteristics. However, their 
use is limited by instability (due to hydrolysis of the phospholipids), limited drug - 
loading capacity, technical diffi culties in obtaining sterile preparations, and blurred 
vision due to their size and opacity [42] . 
In addition, liposomes are subject to the same rapid precorneal clearance as 
conventional ocular solutions, especially the ones with a negative or no surface 
charge [127] . Positively charged liposomes, on the other hand, were reported to 
exhibit a prolonged precorneal retention due to electrostatic interactions with the 
negative sialic acid residues of the mucin layer [2, 127, 208, 210 – 213] . 
There have been several attempts to use liposomes in combination with other 
newer formulation approaches, such as incorporating them into mucoadhesive gels 
or coating them with mucoadhesive polymers [210] . Mucoadhesive polymers inves- 
FORMULATION APPROACHES TO IMPROVE OCULAR BIOAVAILABILITY 747

748 OCULAR DRUG DELIVERY 
tigated in this regard were poly(acrylic acid) (PAA), hyaluronic acid (HA), chitosan, 
and poloxamer [36, 43, 121 – 122, 214] . 
Durrani and co - workers [122] reported on the infl uence of a carbopol coating on 
the corneal retention of pilocarpine - loaded liposomes, and demonstrated a biphasic 
response with an initial low intensity followed by a sustained reaction. 
Bochot et al. [124, 125] developed a novel delivery system for oligonucleotides 
by incorporating them into liposomes and then dispersing them into a thermosensitive 
gel composed of poloxamer 407. They compared the in vitro release of the 
model oligonucleotides pdT16 from simple poloxamer gels (20 and 27% poloxamer) 
with the ones where pdT16 was encapsulated into liposomes and then dispersed 
within the gels. They found that the release of the oligonucleotides from the gels 
was controlled by the poloxamer dissolution, whereas the dispersion of liposomes 
within 27% poloxamer gel was shown to slow down the diffusion of pdT16 out of 
the gel. 
Niosomes In order to circumvent some of the limitations encountered with 
liposomes, such as their chemical instability, the cost and purity of the natural 
phospholipids, and oxidative degradation of the phospholipids, niosomes have been 
developed. Niosomes are nonionic surfactant vesicles which exhibit the same bilayered 
structures as liposomes. Their advantages over liposomes include improved 
chemical stability and low production costs. Moreover, niosomes are biocompatible, 
biodegradable, and nonimmunogenic [215] . They were also shown to increase the 
ocular bioavailability of hydrophilic drugs signifi cantly more than liposomes. This is 
due to the fact that the surfactants in the niosomes act as penetrations enhancers 
and remove the mucous layer from the ocular surface [209] . 
A modifi ed version of niosomes are the so - called discomes, which vary from the 
conventional niosomes in size and shape. The larger size of the vesicles (12 – 60 . m) 
prevents their drainage into the nasolacrimal drainage system. Furthermore, their 
disclike shape provides them with a better fi t in the cul - de - sac of the eye [26] . 
Vyas et al. [216] demonstrated that discomes entrapped higher amounts of timolol 
maleate than niosomes and that both niosomes and discomes signifi cantly increased 
the bioavailability of timolol maleate when compared to a conventional timolol 
maleate solution. 
Microemulsions Microemulsions (MEs) are colloidal dispersions composed of an 
oil phase, an aqueous phase, and one or more surfactants. They are optically isotropic 
and thermodynamically stable and appear as transparent liquids as the droplet 
size of the dispersed phase is less than 150 nm. One of their main advantages is their 
ability to increase the solubilization of lipophilic and hydrophilic drugs accompanied 
by a decrease in systemic absorption [217] . Moreover, MEs are transparent 
systems thus enable monitoring of phase separation and/or precipitation. In addition, 
MEs possess low surface tension and therefore exhibit good wetting and 
spreading properties. 
While the presence of surfactants is advantageous due to an increase in cellular 
membrane permeability, which facilitates drug absorption and bioavailability [218] , 
caution needs to be taken in relation to the amount of surfactant incorporated, as 
high concentrations can lead to ocular toxicity. In general, nonionic surfactants are 
preferred over ionic ones, which are generally too toxic to be used in ophthalmic 

formulations. Surfactants most frequently utilized for the preparation of MEs are 
poloxamers, polysorbates, and polyethylene glycol derivatives [219] . 
Similar to all other colloidal delivery systems discussed above it was hypothesized 
by numerous research teams that a positive charge (provided by cationic surfactants 
[220] ) would increase the ocular residence time of the formulation due to electrostatic 
interactions with the negatively charged mucin residues. However, toxicological 
studies contradicted this assumption regarding the ocular effects, and so far there 
has been no publication demonstrating a distinct benefi cial effect of charged surfactants 
incorporated into MEs. 
Microemulsions can be classifi ed into three different types depending on their 
microstructure: oil - in - water (o/w ME), water - in - oil (w/o ME), and bicontinuous ME. 
They have been investigated by physical chemists since the 1940s but have only 
gained attention as potential ocular drug delivery carriers within the last two 
decades. 
Gasco and co - workers [221] investigated the potential application of o/w lecithin 
MEs for ocular administration of timolol, in which the drug was present as an ion 
pair with octanoate. The ocular bioavailability of the timolol ion pair incorporated 
into the ME was compared to that of an ion pair solution as well as a simple timolol 
solution. Areas under the curve for the ME and the ion pair solution respectively 
were 3.5 and 4.2 times higher than that of the simple timolol solution. A prolonged 
absorption was achieved using the ME with detectable amounts of the drug still 
present 120 min after instillation. 
Various lecithin - based MEs were also characterized by Hasse and Keipert [131] . 
The formulations were tested in terms of their physicochemical parameters (pH, 
refractive index, osmolality, viscosity, and surface tension) and physiological compatibility 
(HET - CAM and Draize test). In addition, in vitro and in vivo evaluations 
were performed. The tested MEs showed favorable physicochemical parameters 
and no ocular irritation as well as a prolonged pilocarpine release in vitro and in 
vivo. 
Muchtar and co - workers [129] prepared MEs with poloxamer 188 and 
soybean lecithin to deliver indometacin to the ocular tissues. They found a threefold 
increased indomethacin concentration in the cornea and aqueous humor 1 h 
post - instillation. 
Beilin et al. [222] demonstrated a prolonged ocular retention of a submicrometer 
emulsion (SME) in the conjunctival sac using a fl uorescent marker (0.01% calcein) 
as well as the miotic response of New Zealand Albino rabbits to pilocarpine. They 
found that the fl uorescence intensity of calcein in SME was signifi cantly higher than 
that of a calcein solution at all time points. Moreover, the pilocarpine SME exhibited 
a longer duration of miosis than the simple pilocarpine solution. It should be mentioned 
that SMEs are true emulsions, being different from MEs. They do not form 
spontaneously and are kinetically rather than thermodynamically stable. They generally 
require high - shear homogenization to form and are more susceptible to phase 
inversion. Furthermore, they are neither transparent nor translucent but rather 
turbid due their larger droplet size compared to MEs. While the two terms are used 
interchangeably in the the scientifi c literature, they actually refer to two distinct 
categories of dispersed systems and should be differentiated from each other. 
The w/o MEs composed of water, Crodamol EO, Crill 1, and Crillet 4 were investigated 
as potential ocular delivery systems by Alany et al. [133] . It was hypothesized 
FORMULATION APPROACHES TO IMPROVE OCULAR BIOAVAILABILITY 749

750 OCULAR DRUG DELIVERY 
that w/o MEs undergo phase transition into lamellar liquid crystals (LCs) upon 
aqueous dilution by the tears, prolonging the precorneal retention time due to an 
increase in the formulation ’ s viscosity. HET - CAM studies revealed no ocular irritancy 
by the excipients used. Ocular drainage was evaluated via . - scintigraphy and 
demonstrated a signifi cantly higher precorneal retention of the tested microemulsions 
compared to an aqueous solution. 
The use of MEs in ocular delivery is very attractive due to all the advantages 
offered by these formulations. They are thermodynamically stable and transparent, 
possess low viscosity, and thus are easy to instill, formulate, and sterilize (via fi ltration). 
Moreover, they offer the possibility of reservoir and/or localizer effects. All 
these factors, in addition to the ones previously mentioned, render MEs promising 
ocular delivery systems. 
5.9.3.4 Other Delivery Approaches 
Many other ocular delivery approaches have been investigated over the past decades, 
including the use of prodrugs, penetration enhancers, cyclodextrins, as well as different 
types of ocular inserts. In addition, iontophoresis, which is an active drug 
delivery approach utilizing electrical current of only 1 – 2 mA to transport ionized 
drugs across the cornea, offers an effective, noninvasive method for ocular delivery. 
Another more recent approach is the use of dendrimers in ocular therapy. Dendrimers 
are synthetic spherical molecules named after their characteristic treelike 
branching around a central core with a size ranging from 2 to 10 nm in diameter 
[223] . So far, PAMAM (polyamidoamine) has been the most commonly studied 
dendrimer system for ocular use [224, 225] . 
Prodrugs Prodrugs are pharmacologically inactive derivates of drug molecules 
that require a chemical or enzymatic transformation into their active parent drug 
[226] . To be effective, an ocular prodrug should show an appropriate lipophilicity to 
facilitate corneal absorption, posses suffi cient aqueous solubility and stability to be 
formulated as an eye drop, and demonstrate the ability to be converted to the active 
parent drug at a rate that meets therapeutic needs [227] . 
When considering ophthalmic drug molecules as prodrug candidates, the following 
factors need to be considered: the pathway and mechanism of ocular drug 
penetration, the functional groups of the drug candidate amenable to prodrug 
derivatization, and the enzymes present in the ocular tissues, which are necessary 
for prodrug activation [28] . 
The majority of ophthalmic prodrugs developed so far are chemically classifi ed 
as ester. They are derived from the esterifi cation of the hydroxyl or carboxylic acid 
groups present in the parent molecule. Of all enzymes participating in the activation 
of prodrugs, esterases, which are present in all anterior segment tissues except the 
tear fi lm, have received the most attention [228, 229] . 
Prodrugs were introduced into the area of ocular drug delivery about 25 years 
ago [230] , and steroids were probably the fi rst ones to be utilized as prodrugs. 
However, the concept of prodrugs was not fully exploited until the introduction of 
dipivefrin (epinephrine prodrug) in the late 1970s. Kaback and co - workers [134] 
found that a 0.1% dipivalyl epinephrine solution lowered the intraocular pressure 
as effective as a 2% epinephrine solution, while signifi cantly lowering the systemic 

side effects. Wei et al. [135] compared the ocular penetration, distribution, and 
metabolism of epinephrine and dipivalyl epinephrine and found the partition coef- 
fi cient of the later to be 100 – 600 times higher than that of epinephrine, therefore 
leading to a 10 - times faster absorption into the rabbit eye. 
Dipivefrin was the only commercially available ophthalmic prodrug at that time. 
However, numerous prodrug derivates have been designed to improve the effi cacy 
of ophthalmic drugs ever since. 
Jarvinen and co - workers [136] synthesized unique O, O. - (xylylene)bispilocarpic 
acid esters containing two pilocarpic acid monoesters linked with one moiety. The 
found that prodrug showed a two - to seven - fold higher corneal permeability than 
pilocarpine itself despite the high molecular weight. 
Tirucherai et al. [137] formulated an acyl ester prodrug of ganciclovir. The 
increased permeability was associated with a linear increased susceptibility of the 
ganciclovir esters to the esterases present in the cornea. 
So far, aims that have been achieved by using prodrugs include the modifi cation 
of the drug ’ s duration of action, reduction of the systemic absorption, and reduction 
of ocular and systemic side effects. Although prodrugs are commonly used to treat 
diseases of the anterior segment, there have also been attempts to treat conditions 
associated with the posterior segment of the eye. 
Penetration Enhancers The transport process across the corneal tissue is the rate - 
limiting step in ocular drug absorption. Increasing the permeability of the corneal 
epithelium by penetration enhancers is likely to enhance the drug transport across 
the corneal tissues and therefore improve ocular bioavailability of the drug. 
Penetration enhancers act by increasing the permeability of the corneal cell 
membrane and/or loosening the tight junctions between the epithelial cells, which 
primarily restrict the entry of molecules via the paracellular pathway. Classes of 
penetration enhancers include surfactants, bile salts, calcium chelators, preservatives, 
fatty acids, and some glycosides such a saponin. 
Surfactants are perceived to enhance drug absorption by disturbing the integrity 
of the plasma membranes. When present at low concentrations, surfactants are 
incorporated into the lipid bilayer, leading to polar defects in the membrane, which 
change the membrane ’ s physical properties. When the lipid bilayer is saturated, 
micelles start to form, enclosing phospholipids from the membranes, hence leading 
to membrane solubilization [36] . Saettone et al. [138] found an increased corneal 
permeability for atenolol, timolol, and betaxolol by including 0.05% Brij 35, Brij 78, 
and Brij 98 into their formulations. 
Bile salts are amphiphilic molecules that are surface active and self - associate to 
form micelles in aqueous solution. They increase corneal permeability by changing 
the rheological properties of the bilayer [231] . A number of bile salts such as deoxycholate, 
taurodeoxycholate, and glycocholate have been tested so far, and it was 
suggested, that a difference in their physicochemical properties (solubilizing activity, 
lipophilicity, Ca 2+ sequestration capacity) is probably related to their performance 
as permeability - enhancing agents [36] . 
Another class of penetration enhancers includes calcium chelators such as ethylenediaminetetraacetic 
acid (EDTA). These molecules induce Ca 2+ depletion in the 
cells. This leads to a global change within the cell and as a result loosens the tight 
junctions between superfi cial epithelial cells, thus facilitating paracellular transport 
FORMULATION APPROACHES TO IMPROVE OCULAR BIOAVAILABILITY 751

752 OCULAR DRUG DELIVERY 
[138, 232] . Grass et al. [33] were among the fi rst to emphasize the enhancing effects 
of chelating agents for ocular drug delivery. They found that 0.5% EDTA doubled 
the corneal absorption of topically applied glycerol and cromoclycin sodium. 
Large numbers of penetration enhancers have been investigated to date. However, 
the unique structure of the corneal/conjunctival tissues requires caution. When 
selecting penetration enhancers for ocular delivery, their capacity to affect the 
integrity of the epithelial surfaces needs to be considered. Studies have shown that 
penetration enhancers themselves can penetrate ocular tissues, which could lead to 
potential toxicity. EDTA concentrations in the iris – ciliary body, for example, were 
found to be high enough to alter the permeability of the blood vessels in the uveal 
tract, therefore indirectly accelerating the drug ’ s removal from the aqueous humor 
[233] . Similarly, benzalkonium chloride (BAC), a cationic surfactant which shows 
the highest penetration - enhancing effect among the currently used preservatives, 
was found to accumulate in the cornea for several days. 
Cyclodextrins Cyclodextrins were introduced into the area of ocular drug delivery 
in the early 1990s. They are a group of homologous cyclic oligosaccharides with a 
hydrophilic outer surface and a lipophilic cavity in the center. Their initial aim 
was to increase the solubility of lipophilic drugs by forming inclusion complexes. 
Cyclodextrin complexation generally results in improved wettability, dissolution, 
solubility, and stability in solution as well as reduced side effects [234] . 
It is assumed that cyclodextrins are too large and hydrophilic to penetrate biological 
membranes. However, they act as penetration enhancers by assuring a high 
drug concentration at the corneal surface, from where the drug then partitions into 
the ocular tissues [235] . 
Even though cyclodextrins drug complexes seem to decrease ocular toxicity of 
irritant drugs, cyclodextrins themselves may exhibit ocular toxicity and should therefore 
be screened by performing corneal sensitivity studies. Among all cyclodextrin 
derivates investigated, hydroxy - propyl - . - cyclodextrin showed the most favorable 
properties in terms of toxicity [1] . 
Nijhawan and Agarwal [140] investigated inclusion complexes of ciprofl oxacin 
hydrochloride and hydroxy - propyl - . - cyclodextrin and found that the complexes 
exhibited better stability, biological activity, and ocular tolerance than the uncomplexed 
drug in solution. 
Aktas et al. [141] showed an increased permeation of pilocarpine nitrate complexed 
with hydroxy - propyl - . - cyclodextrin using isolated rabbit cornea. They found 
a signifi cant reduction in the pupil diameter compared to a simple aqueous solution 
of the same active compound. 
Cyclodextrins improve chemical stability, increase the drug ’ s bioavailability, and 
decrease local irritation. However, the improvement of ocular bioavailability seems 
to be limited by the very slow dissociation of the complexes in the precorneal tear 
fl uid. 
Several studies have shown that combinations of cyclodextrins drug complexes 
and viscosity enhancers can signifi cantly improved ocular absorption [141, 236 – 237] 
and should therefore be further investigated. 
Ocular Inserts Solid ocular dosage forms such as fi lms, erodible and nonerodible 
inserts, rods, and shields have been developed to overcome the typical pulse - entry - 
type drug release associated with conventional ocular dosage forms. This pulse entry 

is characterized by a transient overdose, a relatively short period of appropriate 
dosing, followed by a prolonged period of underdosing. Ocular inserts were developed 
in order to overcome these disadvantages by providing a more controlled, 
sustained, and continuous drug delivery by maintaining an effective drug concentration 
in the target tissues and yet minimizing the number of applications [238] . 
Ocular inserts probably represent one of the oldest ophthalmic formulation 
approaches. In 1948 the British Pharmacopoeia described an atropine - in - gelatin 
wafer and ever since then numerous systems have been developed applying various 
polymers and different release principals. However, the diffi culty of insertion by the 
patient, foreign - body sensation, and inadvertent loss of inserts from the eye make 
these systems less popular, especially among the elderly. Furthermore, the high cost 
involved in manufacture prevented the insert market from taking off [197] . 
Two products, Alza Ocusert and Merck Lacrisert, have been marketed, although 
Ocusert is no longer available. Ocusert is a membrane - controlled reservoir system 
for the treatment of glaucoma. It contains pilocarpine and alginic acid in the core 
reservoir, sandwiched between two transparent, lipophilic ethylenevinyl acetate 
(EVA) rate - controlling membranes, which allow the drug to diffuse from the reservoir 
at a precisely determined rate for a period of seven days. This system is nonbiodegradable 
and must therefore be removed after use. Lacrisert, on the other 
hand, is a soluble minirod of hydroxypropylmethyl cellulose without any active 
ingredient. The system is placed in the conjunctival sac, where it softens within an 
hour and completely dissolves within 14 – 18 h. Lacrisert stabilizes and thickens the 
precorneal tear fi lm and prolongs the tear fi lm break - up time, which is usually 
accelerated in patients with dry - eye syndrome (keratoconjunctivitis sicca) [239] . 
A number of ocular inserts using different techniques, namely soluble, erodible, 
nonerodible, and hydrogel inserts with polymers such as cellulose derivates, acrylates, 
and poly(ethylene oxide), have been investigated over the last few decades. 
An example of a degradable matrix system is the pilocarpine - containing inserts 
formulated by Saettone et al. [148] . Pilocarpine nitrate and polyacrylic acid were 
incorporated into a matrix containing polyvinyl alcohol and two types of hydroxypropyl 
methylcellulose. It was shown that all inserts signifi cantly increased the pharmacological 
effect (miotic response) compared to a solution of pilocarpine nitrate. 
Sasaki et al. [151] prepared nondegradable disc - type ophthalmic inserts of 
. - blockers using different polymers. They found that inserts made from 
poly(hydroxypropyl methacrylate) were able to control the release of tilisolol 
hydrochloride. 
Numerous studies have also been performed on soluble collagen shields. Collagen 
shields are fabricated from porcine scleral tissue, which has a similar collagen 
composition to that of the human cornea. Drug loading is typically achieved by 
soaking the collagen shield in the drug solution prior to application. Designed to 
slowly dissolve within 12, 24, or 72 h, collagen shields have attracted much interest 
as potential sustained ocular drug delivery systems over the last years [240] . 
5.9.4 CONCLUSION 
Although conventional eye drops still represent about 90% of all marketed ophthalmic 
dosage forms, there have been signifi cant efforts towards the development 
of new drug delivery systems. 
CONCLUSION 753

754 OCULAR DRUG DELIVERY 
Only a few of these new ophthalmic drug delivery systems have been commercialized 
over the past decades, but research in the different areas of ocular drug 
delivery has provided important impetus and dynamism, with the promise of some 
new and exciting developments in the fi eld. 
An ideal ophthalmic delivery system should be able to achieve an effective drug 
concentration at the target site for an extended period of time while minimizing 
systemic side effects. In addition, the system should be comfortable and easy to use, 
as the patient ’ s acceptance will continue to play an important role in the design of 
future ocular formulations. 
All delivery technologies mentioned in this chapter hold unlimited potential for 
clinical ophthalmology. However, each of them still bears its own drawbacks. To 
circumvent these, newer trends are directed toward combinations of the different 
drug delivery approaches. Examples for this include the incorporation of particulates 
into in situ gelling systems or coating of nanoparticles with mucoadhesive 
polymers. 
These combinations will open new directions for the improvement of ocular 
bioavailability, but they will also increase the complexity of the formulations, thus 
increasing the diffi culties in understanding the mechanism of action of the drug 
delivery systems. 
Many interesting delivery approaches have been investigated during the past 
decades in order to optimize ocular bioavailability, but much remains to be learned 
before the perfect ocular drug delivery system can be developed. 
REFERENCES 
1. Le Bourlais , C. , Acar , L. , Zia , H. , Sado , P. A. , Needham , T. , and Leverge , R. ( 1998 ), 
Ophthalmic drug delivery systems — Recent advances , Prog. Ret. Eye Res. , 17 ( 1 ), 33 – 
58 . 
2. Le Bourlais , C. A. , Treupel - Acar , L. , Rhodes , C. T. , Sado , P. A. , and Leverge , R. ( 1995 ), 
New ophthalmic drug delivery systems , Drug Dev. Ind. Pharm. , 21 ( 1 ), 19 – 59 . 
3. Jarvinen , K. , Jarvinen , T. , and Urtti , A. ( 1995 ), Ocular absorption following topical delivery 
, Adv. Drug Deliv. Rev. , 16 ( 1 ), 3 – 19 . 
4. Sasaki , H. , Yamamura , K. , Mukai , T. , Nishida , K. , Nakamura , J. , Nakashima , M. , et al. 
( 1999 ), Enhancement of ocular drug penetration , Crit. Rev. Ther. Drug Carrier Syst. , 
16 ( 1 ), 85 – 146 . 
5. Baeyens , V. , and Gurny , R. ( 1997 ), Chemical and physical parameters of tears relevant 
for the design of ocular drug delivery formulations , Pharm. Acta Helv. , 72 ( 4 ), 191 – 202 . 
6. Van Haeringen , N. J. ( 1981 ), Clinical biochemistry of tears , Surv. Ophthalmol. , 26 ( 2 ), 
84 – 96 . 
7. Nagyova , B. , and Tiffany , J. M. ( 1999 ), Components responsible for the surface tension 
of human tears , Curr. Eye Res. , 19 ( 1 ), 4 – 11 . 
8. Tiffany , J. M. (2003), Tears in health and disease , Eye , 17 ( 8 ), 923 – 926 . 
9. Murube , J. , Murube , L. , and Murube , A. ( 1999 ), Origin and types of emotional tearing , 
Eur. J. Ophthalmol. , 9 ( 2 ), 77 – 84 . 
10. Versura , P. , Bonvicini , F. , Caramazza , R. , and Laschi , R. ( 1985 ), Scanning electron microscopy 
of normal human corneal epithelium obtained by scraping - off in vivo , Acta 
Ophthalmol. , 63 ( 3 ), 361 – 365 . 

REFERENCES 755 
11. Klyce , S. D. , and Crosson , C. E. ( 1985 ), Transport processes across the rabbit corneal 
epithelium: A review , Curr. Eye Res. , 4 ( 4 ), 323 – 331 . 
12. Lens , A. ( 1999 ), Ocular Anatomy and Physiology , Slack , New York . 
13. Ahmed , I. ( 2003 ), The noncorneal route in ocular drug delivery , in Mitra , A. K. , 
Ed., Ophthalmic Drug Delivery Systems , 2nd ed., Marcel Dekker , New York , pp. 
335 – 363 . 
14. Greaves , J. L. , and Wilson , C. G. ( 1993 ), Treatment of diseases of the eye with mucoadhesive 
delivery systems , Adv. Drug Deliv. Rev. , 11 ( 3 ), 349 – 383 . 
15. Huang , A. J. W. , Tseng , S. C. G. , and Kenyon , K. R. ( 1989 ), Paracellular permeability of 
corneal and conjunctival epithelia , Invest. Ophthalmol. Vis. Sci. , 30 ( 4 ), 684 – 689 . 
16. Ahmed , I. , and Patton , T. F. ( 1985 ), Importance of the noncorneal absorption route in 
topical ophthalmic drug delivery , Invest. Ophthalmol. Vis. Sci. , 26 ( 4 ), 584 – 587 . 
17. Macha , S. , Hughes , P. M. , and Mitra , A. K. ( 2003 ), Overview of ocular drug delivery , in 
Mitra , A. K. , Ed., Ophthalmic Drug Delivery Systems , 2nd ed., Marcel Dekker , New 
York , pp. 1 – 11 . 
18. Robinson , J. C. ( 1993 ), Ocular anatomy and physiology relevant to ocular drug delivery , 
in Mitra , A. K. , Ed., Ophthalmic Drug Delivery Systems , Marcel Dekker , New York , pp. 
29 – 58 . 
19. Mikkelson , T. J. , Chrai , S. S. , and Robinson , J. R. ( 1973 ), Altered bioavailability of drugs 
in the eye due to drug - protein interaction , J. Pharm. Sci. , 62 ( 10 ), 1648 – 1653 . 
20. Stjernschantz , J. , and Astin , M. ( 1993 ), Anatomy and physiology of the eye. Physiological 
aspects of ocular drug therapy , in Edman , P. , Ed., Biopharmaceutics of Ocular Drug 
Delivery , CRC Press , Boca Raton, FL , pp. 1 – 25 . 
21. Hornof , M. , Toropainen , E. , and Urtti , A. ( 2005 ), Cell culture models of the ocular barriers 
, Eur. J. Pharm. Biopharm. , 60 ( 2 ), 207 – 225 . 
22. Jumbe , N. L. , and Miller , M. H. ( 2003 ), Ocular drug transfer following systemic drug 
administration , in Mitra , A. K. , Ed., Ophthalmic Drug Delivery Systems , 2nd ed., Marcel 
Dekker , New York , pp. 109 – 133 . 
23. Urtti , A. , and Salminen , L. ( 1993 ), Animal pharmacokinetic studies , in Mitra , A. K. , Ed., 
Ophthalmic Drug Delivery Systems , Marcel Dekker , New York , pp. 121 – 136 . 
24. Duvvuri , S. , Majumdar , S. , and Mitra , A. K. ( 2003 ), Drug delivery to the retina: 
Challenges and opportunities , Expert Opin. Biol. Ther. , 3 ( 1 ), 45 – 56 . 
25. Loftssona , T. , and Jarvinen , T. ( 1999 ), Cyclodextrins in ophthalmic drug delivery , Adv. 
Drug Deliv. Rev. , 36 ( 1 ), 59 – 79 . 
26. Kaur , I. P. , Garg , A. , Singla , A. K. , and Aggarwal , D. ( 2004 ), Vesicular systems in ocular 
drug delivery: An overview , Int. J. Pharm. , 269 ( 1 ), 1 – 14 . 
27. Lee , V. H. , and Robinson , J. R. ( 1979 ), Mechanistic and quantitative evaluation of precorneal 
pilocarpine disposition in albino rabbits , J. Pharm. Sci. , 68 ( 6 ), 673 – 684 . 
28. Lee , V. H. L., and Li , V. H. K. ( 1989 ), Prodrugs for improved ocular drug delivery , Adv. 
Drug Deliv. Rev. , 3 ( 1 ), 1 – 38 . 
29. Rojanasakul , Y. , and Robinson , J. R. ( 1989 ), Transport mechanisms of the cornea: 
Characterization of barrier permselectivity , Int. J. Pharm. , 55 ( 2 – 3 ), 237 – 246 . 
30. Palmgren , J. J. , Toropainen , E. , Auriola , S. , and Urtti , A. ( 2002 ), Liquid chromatographic - 
electrospray ionization mass spectrometric analysis of neutral and charged polyethylene 
glycols , J. Chromatogr. A , 976 ( 1 – 2 ), 165 – 170 . 
31. Van Ooteghem , M. M. M. ( 1993 ), Formulation of ophthalmic solutions and suspensions. 
Problems and Advantages , in Edman , P. , Ed., Biopharmaceutics of Ocular Drug Delivery , 
CRC Press , Boca Raton, FL , pp. 27 – 42 . 

756 OCULAR DRUG DELIVERY 
32. Schoenwald , R. D. , and Huang , H. S. ( 1983 ), Corneal penetration behavior of beta - 
blocking agents I: Physiochemical factors , J. Pharm. Sci. , 72 ( 11 ), 1266 – 1272 . 
33. Grass , G. M. , Wood , R. W. , and Robinson , J. R. ( 1985 ), Effects of calcium chelating agents 
on corneal permeability , Invest. Ophthalmol. Vis. Sci. , 26 ( 1 ), 110 – 113 . 
34. Liaw , J. , Rojanasakul , Y. , and Robinson , J. R. ( 1992 ), The effect of drug charge type and 
charge density on corneal transport , Int. J. Pharm. , 88 ( 1 – 3 ), 111 – 124 . 
35. Sieg , J. W. , and Robinson , J. R. ( 1975 ), Vehicle effects on ocular drug bioavailability: 
Evaluation of fl uorometholone , J. Pharm. Sci. , 64 ( 6 ), 931 – 936 . 
36. Kaur , I. P. , and Smitha , R. ( 2002 ), Penetration enhancers and ocular bioadhesives: Two 
new avenues for ophthalmic drug delivery , Drug Dev. Ind. Pharm. , 28 ( 4 ), 353 – 369 . 
37. Mishima , S. , Gasset , A. , Klyce , S. D. , Jr , and Baum , J. L. ( 1966 ), Determination of tear 
volume and tear fl ow , Invest. Ophthalmol. , 5 ( 3 ), 264 – 276 . 
38. Mainardes , R. M. , Urban , M. C. C. , Cinto , P. O. , Khalil , N. M. , Chaud , M. V. , Evangelista , 
R. C. , et al. ( 2005 ), Colloidal carriers for ophthalmic drug delivery , Curr. Drug Targets , 
6 ( 3 ), 363 – 371 . 
39. Chrai , S. S. , Patton , T. F. , Mehta , A. , and Robinson , J. R. ( 1973 ), Lacrimal and instilled 
fl uid dynamics in rabbit eyes , J. Pharm. Sci. , 62 ( 7 ), 1112 – 1121 . 
40. Chrai , S. S. , and Robinson , J. R. ( 1974 ), Ocular evaluation of methylcellulose vehicle in 
albino rabbits , J. Pharm. Sci. , 63 ( 8 ), 1218 – 1223 . 
41. Sieg , J. W. , and Triplett , J. W. ( 1980 ), Precorneal retention of topically instilled micronized 
particles , J. Pharm. Sci. , 69 ( 7 ), 863 – 864 . 
42. Lee , V. H. L. , Urrea , P. T. , Smith , R. E. , and Schanzlin , D. J. ( 1985 ), Ocular drug bioavailability 
from topically applied liposomes , Surv. Ophthalmol. , 29 ( 5 ), 335 – 348 . 
43. Zimmer , A. , and Kreuter , J. ( 1995 ), Microspheres and nanoparticles used in ocular 
delivery systems , Adv. Drug Deliv. Rev. , 16 ( 1 ), 61 – 73 . 
44. Ludwig , A. ( 2005 ), The use of mucoadhesive polymers in ocular drug delivery , Adv. Drug 
Deliv. Rev. , 57 ( 11 ), 1595 – 1639 . 
45. Ludwig , A. , and Van Ooteghem , M. ( 1987 ), The infl uence of the osmolality on the precorneal 
retention of ophthalmic solutions , J. Pharm. Belg. , 42 ( 4 ), 259 – 266 . 
46. Kaufman , H. E. , Steinemann , T. L. , Lehman , E. , Thompson , H. W. , Varnell , E. D. , 
Jacob - LaBarre , J. T. , et al. ( 1994 ), Collagen - based drug delivery and artifi cial tears , 
J. Ocul. Pharmacol. , 10 ( 1 ), 17 – 27 . 
47. Ke , T. - L. , Cagle , G. , Schlech , B. , Lorenzetti , O. J. , and Mattern , J. ( 2001 ), Ocular bioavailability 
of ciprofl oxacin in sustained release formulations , J. Ocul. Pharmacol. Ther. , 17 ( 6 ), 
555 – 563 . 
48. Meseguer , G. , Buri , P. , Plazonnet , B. , Rozier , A. , and Gurny , R. ( 1996 ), Gamma scintigraphic 
comparison of eyedrops containing pilocarpine in healthy volunteers , J. Ocul. 
Pharmacol. Ther. , 12 ( 4 ), 481 – 488 . 
49. Hui , H. - W. , and Robinson , J. R. ( 1985 ), Ocular delivery of progesterone using a bioadhesive 
polymer , Int. J. Pharm. , 26 ( 3 ), 203 – 213 . 
50. Lele , B. S. , and Hoffman , A. S. ( 2000 ), Insoluble ionic complexes of polyacrylic acid with 
a cationic drug for use as a mucoadhesive, ophthalmic drug delivery system , J. Biomater. 
Sci. Polym. Ed. , 11 ( 12 ), 1319 – 1331 . 
51. Saettone , M. F. , Giannaccini , B. , Ravecca , S. , La Marca , F. , and Tota , G. ( 1984 ), Polymer 
effects on ocular bioavailability — The infl uence of different liquid vehicles on the mydriatic 
response of tropicamide in humans and in rabbits , Int. J. Pharm. , 20 ( 1 – 2 ), 187 – 202 . 
52. Herrero - Vanrell , R. , Fernandez - Carballido , A. , Frutos , G. , and Cadorniga , R. ( 2000 ), 
Enhancement of the mydriatic response to tropicamide by bioadhesive polymers , 
J. Ocul. Pharmacol. Ther. , 16 ( 5 ), 419 – 428 . 

REFERENCES 757 
53. Saettone , M. F. , Chetoni , P. , Tilde Torracca , M. , Burgalassi , S. , and Giannaccini , B. ( 1989 ), 
Evaluation of muco - adhesive properties and in vivo activity of ophthalmic vehicles 
based on hyaluronic acid , Int. J. Pharm. , 51 ( 3 ), 203 – 212 . 
54. Sandri , G. , Bonferoni , M. C. , Chetoni , P. , Rossi , S. , Ferrari , F. , Ronchi , C. , et al. ( 2006 ), 
Ophthalmic delivery systems based on drug - polymer - polymer ionic ternary interaction: 
In vitro and in vivo characterization , Eur. J. Pharm. Biopharm. , 62 ( 1 ), 59 – 69 . 
55. Felt , O. , Furrer , P. , Mayer , J. M. , Plazonnet , B. , Buri , P. , and Gurny , R. ( 1999 ), Topical use 
of chitosan in ophthalmology: Tolerance assessment and evaluation of precorneal retention 
, Int. J. Pharm. , 180 ( 2 ), 185 – 193 . 
56. Felt , O. , Baeyens , V. , Buri , P. , and Gurny , R. ( 2001 ), Delivery of antibiotics to 
the eye using a positively charged polysaccharide as vehicle , Aaps Pharmsci , 3 ( 4 ), 
E34 . 
57. Di Colo , G. , Zambito , Y. , Burgalassi , S. , Nardini , I. , and Saettone , M. F. ( 2004 ), Effect of 
chitosan and of N - carboxymethylchitosan on intraocular penetration of topically applied 
ofl oxacin , Int. J. Pharm. , 273 ( 1 – 2 ), 37 – 44 . 
58. Di Colo , G. , Zambito , Y. , Burgalassi , S. , and Saettone , M. F. ( 2003 ), Effects of chitosan 
and of its N - trimethyl and N - carboxymethyl derivatives on the ocular pharmacokinetics 
of ofl oxacin in rabbits , paper presented at the 30th International Symposium on Controlled 
Release of Bioactive Materials, Glasgow, Scottland. 
59. Davies , N. M. , Farr , S. J. , Hadgraft , J. , and Kellaway , I. W. ( 1991 ), Evaluation of mucoadhesive 
polymers in ocular drug delivery. I. Viscous solutions , Pharm. Res. , 8 ( 8 ), 
1039 – 1043 . 
60. Edsman , K. , Carlfors , J. , and Harju , K. ( 1996 ), Rheological evaluation and ocular 
contact time of some carbomer gels for ophthalmic use , Int. J. Pharm. , 137 ( 2 ), 
233 – 241 . 
61. Hartmann , V. , and Keipert , S. ( 2000 ), Physico - chemical, in vitro and in vivo characterisation 
of polymers for ocular use , Pharmazie , 55 ( 6 ), 440 – 443 . 
62. Burgalassi , S. , Chetoni , P. , Panichi , L. , Boldrini , E. , and Saettone , M. F. ( 2000 ), Xyloglucan 
as a novel vehicle for timolol: Pharmacokinetics and pressure lowering activity in rabbits , 
J. Ocul. Pharmacol. Ther. , 16 ( 6 ), 497 – 509 . 
63. Ceulemans , J. , Vinckier , I. , and Ludwig , A. ( 2002 ), The use of xanthan gum in an ophthalmic 
liquid dosage form: Rheological characterization of the interaction with mucin , 
J. Pharm. Sci. , 91 ( 4 ), 1117 – 1127 . 
64. Verschueren , E. , Van Santvliet , L. , and Ludwig , A. ( 1996 ), Evaluation of various carrageenans 
as ophthalmic viscolysers, STP Pharma Sci. , 6 ( 3 ), 203 – 210 . 
65. Albasini , M. , and Ludwig , A. ( 1995 ), Evaluation of polysaccharides intended for ophthalmic 
use in ocular dosage forms , Farmaco , 50 (Sept.), 633 – 642 . 
66. Snibson , G. R. , Greaves , J. L. , Soper , N. D. , Prydal , J. I. , Wilson , C. G. , and Bron , A. J. 
( 1990 ), Precorneal residence times of sodium hyaluronate solutions studied by quantitative 
gamma scintigraphy , Eye , 4 ( 4 ), 594 – 602 . 
67. Snibson , G. R. , Greaves , J. L. , Soper , N. D. , Tiffany , J. M. , Wilson , C. G. , and Bron , A. J. 
( 1992 ), Ocular surface residence times of artifi cial tear solutions , Cornea , 11 ( 4 ), 
288 – 293 . 
68. Debbasch , C. , De La Salle , S. B. , Brignole , F. , Rat , P. , Warnet , J. M. , and Baudouin , C. 
( 2002 ), Cytoprotective effects of hyaluronic acid and Carbomer 934P in ocular surface 
epithelial cells , Invest. Ophthalmol. Vis. Sci. , 43 ( 11 ), 3409 – 3415 . 
69. Oechsner , M. , and Keipert , S. ( 1999 ), Polyacrylic acid/polyvinylpyrrolidone bipolymeric 
systems. I. Rheological and mucoadhesive properties of formulations potentially useful 
for the treatment of dry - eye - syndrome , Eur. J. Pharm. Biopharm. , 47 ( 2 ), 113 – 118 . 

758 OCULAR DRUG DELIVERY 
70. Shibuya , T. , Kashiwagi , K. , and Tsukahara , S. ( 2003 ), Comparison of effi cacy and tolerability 
between two gel - forming timolol maleate ophthalmic solutions in patients with 
glaucoma or ocular hypertension , Ophthalmologica , 217 ( 1 ), 31 – 38 . 
71. Rosenlund , E. F. ( 1996 ), The intraocular pressure lowering effect of timolol in gel - 
forming solution , Acta Ophthalmol. Scand. , 74 ( 2 ), 160 – 162 . 
72. Stewart , W. C. , Leland , T. M. , Cate , E. A. , and Stewart , J. A. ( 1998 ), Effi cacy and safety 
of timolol solution once daily versus timolol gel in treating elevated intraocular pressure , 
J. Glaucoma , 7 ( 6 ), 402 – 407 . 
73. Shedden , A. , Laurence , J. , and Tipping , R. ( 2001 ), Effi cacy and tolerability of timolol 
maleate ophthalmic gel - forming solution versus timolol ophthalmic solution in adults 
with open - angle glaucoma or ocular hypertension: A six - month, double - masked, multicenter 
study , Clin. Ther. , 23 ( 3 ), 440 – 450 . 
74. Carlfors , J. , Edsman , K. , Petersson , R. , and Jornving , K. ( 1998 ), Rheological evaluation 
of Gelrite(R) in situ gels for ophthalmic use , Eur. J. Pharm. Sci. , 6 ( 2 ), 113 – 119 . 
75. Schenker , H. I. , and Silver , L. H. ( 2000 ), Long - term intraocular pressure - lowering effi - 
cacy and safety of timolol maleate gel - forming solution 0.5% compared with timoptic 
XE 0.5% in a 12 - month study , Am. J. Ophthalmol. , 130 ( 2 ), 145 – 150 . 
76. Shedden , A. H. , Laurence , J. , Barrish , A. , and Olah , T. V. ( 2001 ), Plasma timolol concentrations 
of timolol maleate: Timolol gel - forming solution (TIMOPTIC - XE) once daily 
versus timolol maleate ophthalmic solution twice daily , Doc. Ophthalmol. , 103 ( 1 ), 
73 – 79 . 
77. Lindell , K. , and Engstrom , S. ( 1993 ), In vitro release of timolol maleate from an in situ 
gelling polymer system , Int. J. Pharm. , 95 ( 1 – 3 ), 219 – 228 . 
78. Rozier , A. , Mazuel , C. , Grove , J. , and Plazonnet , B. ( 1989 ), Gelrite(R): A novel, ion - 
activated, in - situ gelling polymer for ophthalmic vehicles. Effect on bioavailability of 
timolol , Int. J. Pharm. , 57 ( 2 ), 163 – 168 . 
79. Balasubramaniam , J. , Kant , S. , and Pandit , J. K. ( 2003 ), In vitro and in vivo evaluation 
of the Gelrite gellan gum - based ocular delivery system for indomethacin , Acta Pharm. , 
53 ( 4 ), 251 – 261 . 
80. Sultana , Y. , Aqil , M. , and Ali , A. ( 2006 ), Ion - activated, Gelrite - based in situ ophthalmic 
gels of pefl oxacin mesylate: Comparison with conventional eye drops , Drug Deliv. , 13 ( 3 ), 
215 – 219 . 
81. Liu , Z. , Li , J. , Nie , S. , Liu , H. , Ding , P. , and Pan , W. ( 2006 ), Study of an alginate/HPMCbased 
in situ gelling ophthalmic delivery system for gatifl oxacin , Int. J. Pharm. , 315 ( 1 – 2 ), 
12 – 17 . 
82. Srividya , B. , Cardoza , R. M. , and Amin , P. D. ( 2001 ), Sustained ophthalmic delivery of 
ofl oxacin from a pH triggered in situ gelling system , J. Controlled Release , 73 ( 2 – 3 ), 
205 – 211 . 
83. Cohen , S. , Lobel , E. , Trevgoda , A. , and Peled , Y. ( 1997 ), A novel in situ - forming ophthalmic 
drug delivery system from alginates undergoing gelation in the eye , J. Controlled 
Release , 44 ( 2 – 3 ), 201 – 208 . 
84. Demailly , P. , Allaire , C. , and Trinquand , C. ( 2001 ), Ocular hypotensive effi cacy and safety 
of once daily carteolol alginate , Br. J. Ophthalmol. , 85 ( 8 ), 921 – 924 . 
85. Cho , K. Y. , Chung , T. W. , Kim , B. C. , Kim , M. K. , Lee , J. H. , Wee , W. R. , et al. ( 2003 ), 
Release of ciprofl oxacin from poloxamer - graft - hyaluronic acid hydrogels in vitro , Int. J. 
Pharm. , 260 ( 1 ), 83 – 91 . 
86. Desai , S. D. , and Blanchard , J. ( 1998 ), Evaluation of pluronic F127 - based sustained - 
release ocular delivery systems for pilocarpine using the albino rabbit eye model , J. 
Pharm. Sci. , 87 ( 10 ), 1190 – 1195 . 

REFERENCES 759 
87. Lin , H. R. , and Sung , K. C. ( 2000 ), Carbopol/pluronic phase change solutions for ophthalmic 
drug delivery , J. Controlled Release , 69 ( 3 ), 379 – 388 . 
88. Miyazaki , S. , Suzuki , S. , Kawasaki , N. , Endo , K. , Takahashi , A. , and Attwood , D. ( 2001 ), 
In situ gelling xyloglucan formulations for sustained release ocular delivery of pilocarpine 
hydrochloride , Int. J. Pharm. , 229 ( 1 – 2 ), 29 – 36 . 
89. El - Kamel , A. H. ( 2002 ), In vitro and in vivo evaluation of Pluronic F127 - based ocular 
delivery system for timolol maleate , Int. J. Pharm. , 241 ( 1 ), 47 – 55 . 
90. Wu , J. , Su , Z. - G. , and Ma , G. - H. ( 2006 ), A thermo - and pH - sensitive hydrogel composed 
of quaternized chitosan/glycerophosphate , Int. J. Pharm. , 315 ( 1 – 2 ), 1 – 11 . 
91. Wei , G. , Xu , H. , Ding , P. T. , Li , S. M. , and Zheng , J. M. ( 2002 ), Thermosetting gels with 
modulated gelation temperature for ophthalmic use: The rheological and gamma scintigraphic 
studies , J. Controlled Release , 83 ( 1 ), 65 – 74 . 
92. Vandervoort , J. , and Ludwig , A. ( 2004 ), Preparation and evaluation of drug - loaded 
gelatin nanoparticles for topical ophthalmic use , Eur. J. Pharm. Biopharm. , 57 ( 2 ), 251 – 
261 . 
93. Zimmer , A. K. , Maincent , P. , Thouvenot , P. , and Kreuter , J. ( 1994 ), Hydrocortisone delivery 
to healthy and infl amed eyes using a micellar polysorbate 80 solution or albumin 
nanoparticles , Int. J. Pharm. , 110 ( 3 ), 211 – 222 . 
94. Zimmer , A. K. , Zerbe , H. , and Kreuter , J. ( 1994 ), Evaluation of pilocarpine - loaded 
albumin particles as drug delivery systems for controlled delivery in the eye I. In vitro 
and in vivo characterisation , J. Controlled Release , 32 ( 1 ), 57 – 70 . 
95. Zimmer , A. K. , Chetoni , P. , Saettone , M. F. , Zerbe , H. , and Kreuter , J. ( 1995 ), Evaluation 
of pilocarpine - loaded albumin particles as controlled drug delivery systems for the eye. 
II. Coadministration with bioadhesive and viscous polymers , J. Controlled Release , 33 ( 1 ), 
31 – 46 . 
96. Mazor , Z. , Ticho , U. , Rehany , U. , and Rose , L. ( 1979 ), Piloplex, a new long - acting pilocarpine 
polymer salt. B: Comparative study of the visual effects of pilocarpine and 
Piloplex eye drops , Br. J. Ophthalmol. , 63 ( 1 ), 48 – 51 . 
97. Ticho , U. , Blementhal , M. , Zonis , S. , Gal , A. , Blank , I. , and Mazor , Z. W. ( 1979 ), Piloplex, 
a new long - acting pilocarpine polymer salt. A: Long - term study , Br. J. Ophthalmol. , 63 ( 1 ), 
45 – 47 . 
98. Klein , H. Z. , Lugo , M. , Shields , M. B. , Leon , J. , and Duzman , E. ( 1985 ), A dose - response 
study of piloplex for duration of action , Am. J. Ophthalmol. , 99 ( 1 ), 23 – 26 . 
99. Gurny , R. ( 1981 ), Preliminary study of prolonged acting drug delivery system for the 
treatment of glaucoma , Pharm. Acta Helv. , 56 ( 4 – 5 ), 130 – 132 . 
100. Gurny , R. , Boye , T. , and Ibrahim , H. ( 1985 ), Ocular therapy with nanoparticulate systems 
for controlled drug delivery , J. Controlled Release , 2 , 353 – 361 . 
101. Gurny , R. , Ibrahim , H. , Aebi , A. , Buri , P. , Wilson , C. G. , Washington , N. , et al. ( 1987 ), 
Design and evaluation of controlled release systems for the eye , J. Controlled Release , 
6 ( 1 ), 367 – 373 . 
102. Marchal - Heussler , L. , Orallo , F. , Vila Jato , J. L. , and Alonso , M. J. ( 1992 ), Colloidal drug 
delivery systems for the eye: A comparison of the effi cacy of three different polymers: 
Polyisobutylcyanoacrylate, polylacticcoglycolic acid, poly - epsilon - caprolactone , STP 
Pharma Sci. , 2 , 98 – 104 . 
103. Marchal - Heussler , L. , Sirbat , D. , Hoffman , M. , and Maincent , P. ( 1993 ), Poly(epsilon - 
caprolactone) nanocapsules in carteolol ophthalmic delivery , Pharm. Res. , 10 ( 3 ), 
386 – 390 . 
104. Harmia , T. , Speiser , P. , and Kreuter , J. ( 1986 ), Optimization of pilocarpine loading onto 
nanoparticles by sorption procedures , Int. J. Pharm. , 33 ( 1 – 3 ), 45 – 54 . 

760 OCULAR DRUG DELIVERY 
105. Diepold , R. , Kreuter , J. , Himber , J. , Gurny , R. , Lee , V. H. , Robinson , J. R. , et al. ( 1989 ), 
Comparison of different models for the testing of pilocarpine eyedrops using conventional 
eyedrops and a novel depot formulation (nanoparticles) , Graefes Arch. Clin. Exp. 
Ophthalmol. , 227 ( 2 ), 188 – 193 . 
106. Harmia , T. , Speiser , P. , and Kreuter , J. ( 1987 ), Nanoparticles as drug carriers in ophthalmology 
, Pharm. Acta Helv. , 62 ( 12 ), 322 – 331 . 
107. Zimmer , A. , Mutschler , E. , Lambrecht , G. , Mayer , D. , and Kreuter , J. ( 1994 ), Pharmacokinetic 
and pharmacodynamic aspects of an ophthalmic pilocarpine nanoparticle - 
delivery - system , Pharm. Res. , 11 ( 10 ), 1435 – 1442 . 
108. De Campos , A. M. , Sanchez , A. , and Alonso , M. J. ( 2001 ), Chitosan nanoparticles: A new 
vehicle for the improvement of the delivery of drugs to the ocular surface. Application 
to cyclosporin A , Int. J. Pharm. , 224(1 – 2), 159 – 168 . 
109. Calvo , P. , Vila - Jato , J. L. , and Alonso , M. J. ( 1997 ), Evaluation of cationic polymer - coated 
nanocapsules as ocular drug carriers , Int. J. Pharm. , 153 ( 1 ), 41 – 50 . 
110. De Campos , A. M. , Sanchez , A. , Gref , R. , Calvo , P. , and Alonso , M. J. ( 2003 ), The effect 
of a PEG versus a chitosan coating on the interaction of drug colloidal carriers with the 
ocular mucosa , Eur. J. Pharm. Sci. , 20 ( 1 ), 73 – 81 . 
111. Giannavola , C. , Bucolo , C. , Maltese , A. , Paolino , D. , Vandelli , M. A. , Puglisi , G. , et al. 
( 2003 ), Infl uence of preparation conditions on acyclovir - loaded poly - d,l - lactic acid nanospheres 
and effect of PEG coating on ocular drug bioavailability , Pharma. Res. , 20 ( 4 ), 
584 – 590 . 
112. Hsiue , G. - H. , Hsu , S. - h. , Yang , C. - C. , Lee , S. - H. , and Yang , I. - K. ( 2002 ), Preparation of 
controlled release ophthalmic drops, for glaucoma therapy using thermosensitive poly - 
N - isopropylacrylamide , Biomaterials , 23 ( 2 ), 457 – 462 . 
113. Pignatello , R. , Bucolo , C. , Ferrara , P. , Maltese , A. , Puleo , A. , and Puglisi , G. ( 2002 ), 
Eudragit RS100(R) nanosuspensions for the ophthalmic controlled delivery of ibuprofen 
, Eur. J. Pharm. Sci. , 16 ( 1 – 2 ), 53 – 61 . 
114. Bucolo , C. , Maltese , A. , Puglisi , G. , and Pignatello , R. ( 2002 ), Enhanced ocular anti - 
infl ammatory activity of ibuprofen carried by an eudragit RS100(R) nanoparticle suspension 
, Ophthal. Res. , 34 ( 5 ), 319 – 323 . 
115. Pignatello , R. , Bucolo , C. , Spedalieri , G. , Maltese , A. , and Puglisi , G. ( 2002 ), Flurbiprofen - 
loaded acrylate polymer nanosuspensions for ophthalmic application , Biomaterials , 
23 ( 15 ), 3247 – 3255 . 
116. Khopade , A. J. , and Jain , N. K. ( 1995 ), Self assembling nanostructures for sustained 
ophthalmic drug delivery , Pharmazie , 50 ( 12 ), 812 – 814 . 
117. Cavalli , R. , Gasco , M. R. , Chetoni , P. , Burgalassi , S. , and Saettone , M. F. ( 2002 ), Solid 
lipid nanoparticles (SLN) as ocular delivery system for tobramycin , Int. J. Pharm. , 
238 ( 1 – 2 ), 241 – 245 . 
118. Safwat , S. M. , and Al - Kassas , R. S. ( 2002 ), Evaluation of gentamicin - Eudragit microspheres 
as ophthalmic delivery systems in infl amed rabbit ’ s eyes, STP Pharma Sci. , 12 ( 6 ), 
357 – 361 . 
119. Giunchedi , P. , Conte , U. , Chetoni , P. , and Saettone , M. F. ( 1999 ), Pectin microspheres as 
ophthalmic carriers for piroxicam: Evaluation in vitro and in vivo in albino rabbits , Eur. 
J. Pharm. Sci. , 9 ( 1 ), 1 – 7 . 
120. Giunchedi , P. , Chetoni , P. , Conte , U. , and Saettone , M. F. ( 2000 ), Albumin microspheres 
for ocular delivery of piroxicam , Pharm. Pharmacol. Commun. , 6 ( 4 ), 149 – 153 . 
121. Davies , N. M. , Farr , S. J. , Hadgraft , J. , and Kellaway , I. W. ( 1992 ), Evaluation of mucoadhesive 
polymers in ocular drug delivery. II. Polymer - coated vesicles , Pharm. Res. , 9 ( 9 ), 
1137 – 1144 . 

REFERENCES 761 
122. Durrani , A. M. , Davies , N. M. , Thomas , M. , and Kellaway , I. W. ( 1992 ), Pilocarpine bioavailability 
from a mucoadhesive liposomal ophthalmic drug delivery system , Int. J. 
Pharm. , 88 ( 1 – 3 ), 409 – 415 . 
123. Henriksen , I. , Green , K. L. , Smart , J. D. , Smistad , G. , and Karlsen , J. ( 1996 ), Bioadhesion 
of hydrated chitosans: An in vitro and in vivo study , Int. J. Pharm. , 145 ( 1 – 2 ), 231 – 
240 . 
124. Bochot , A. , Fattal , E. , Gulik , A. , Couarraze , G. , and Couvreur , P. ( 1998 ), Liposomes dispersed 
within a thermosensitive gel: A new dosage form for ocular delivery of oligonucleotides 
, Pharm. Res. , 15 ( 9 ), 1364 – 1369 . 
125. Bochot , A. , Fattal , E. , Grossiord , J. L. , Puisieux , F. , and Couvreur , P. ( 1998 ), Characterization 
of a new ocular delivery system based on a dispersion of liposomes in a thermosensitive 
gel , Int. J. Pharm. , 162 ( 1 – 2 ), 119 – 127 . 
126. Bochot , A. , Mashhour , B. , Puisieux , F. , Couvreur , P. , and Fattal , E. ( 1998 ), Comparison 
of the ocular distribution of a model oligonucleotide after topical instillation in rabbits 
of conventional and new dosage forms , J. Drug Target. , 6 ( 4 ), 309 – 313 . 
127. Nagarsenker , M. S. , Londhe , V. Y. , and Nadkarni , G. D. ( 1999 ), Preparation and evaluation 
of liposomal formulations of tropicamide for ocular delivery , Int. J. Pharm. , 190 ( 1 ), 
63 – 71 . 
128. Aggarwal , D. , and Kaur , I. P. ( 2005 ), Improved pharmacodynamics of timolol maleate 
from a mucoadhesive niosomal ophthalmic drug delivery system , Int. J. Pharm. , 290 ( 1 – 
2 ), 155 – 159 . 
129. Muchtar , S. , Abdulrazik , M. , Frucht - Pery , J. , and Benita , S. ( 1997 ), Ex - vivo permeation 
study of indomethacin from a submicron emulsion through albino rabbit cornea , J. 
Controlled Release , 44 ( 1 ), 55 – 64 . 
130. Sznitowska , M. , Zurowska - Pryczkowska , K. , Dabrowska , E. , and Janicki , S. ( 2000 ), 
Increased partitioning of pilocarpine to the oily phase of submicron emulsion does not 
result in improved ocular bioavailability , Int. J. Pharm. , 202 ( 1 – 2 ), 161 – 164 . 
131. Ha . e , A. , and Keipert , S. ( 1997 ), Development and characterisation of microemulsions 
for ocular application , Eur. J. Pham. Biopharm. , 43 ( 2 ), 179 – 183 . 
132. Radomska , A. , and Dobrucki , R. ( 2000 ), The use of some ingredients for microemulsion 
preparation containing retinol and its esters , Int. J. Pharm. , 196 ( 2 ), 131 – 134 . 
133. Alany , R. G. , Rades , T. , Nicoll , J. , Tucker , I. G. , and Davies , N. M. ( 2006 ), W/O microemulsions 
for ocular delivery: Evaluation of ocular irritation and precorneal retention , 
J. Controlled Release , 111 ( 1 – 2 ), 145 – 152 . 
134. Kaback , M. B. , Podos , S. M. , Harbin , T. S. Jr , ., Mandell , A. , and Becker , B. ( 1976 ), The 
effects of dipivalyl epinephrine on the eye , Am. J. Ophthalmol. , 81 ( 6 ), 768 – 772 . 
135. Wei , C. P. , Anderson , J. A. , and Leopold , I. ( 1978 ), Ocular absorption and metabolism 
of topically applied epinephrine and a dipivalyl ester of epinephrine , Invest. Ophthalmol. 
Vis. Sci. , 17 ( 4 ), 315 – 321 . 
136. Jarvinen , T. , Suhonen , P. , Auriola , S. , Vepsalainen , J. , Peura , P. , et al. ( 1991 ), O , O. - (1,4 - 
Xylylene)bispilocarpic acid esters as new potential double prodrugs of pilocarpine for 
improved ocular delivery. Part 1. Synthesis and analysis , Int. J. Pharm. , 75 ( 2 – 3 ), 
249 – 258 . 
137. Tirucherai , G. S. , Dias , C. , and Mitra , A. K. ( 2002 ), Corneal permeation of ganciclovir: 
Mechanism of ganciclovir permeation enhancement by acyl ester prodrug design , 
J. Ocul. Pharmacol. Ther. , 18 ( 6 ), 535 – 548 . 
138. Saettone , M. F. , Chetoni , P. , Cerbai , R. , Mazzanti , G. , and Braghiroli , L. ( 1996 ), 
Evaluation of ocular permeation enhancers: In vitro effects on corneal transport of 
four beta - blockers, and in vitro/in vivo toxic activity , Int. J. Pharm. , 142 ( 1 ), 103 – 
113 . 

762 OCULAR DRUG DELIVERY 
139. Maestrelli , F. , Mura , P. , Casini , A. , Mincione , F. , Scozzafava , A. , and Supuran , C. T. ( 2002 ), 
Cyclodextrin complexes of sulfonamide carbonic anhydrase inhibitors as long - lasting 
topically acting antiglaucoma agents , J. Pharm. Sci. , 91 ( 10 ), 2211 – 2219 . 
140. Nijhawan , R. , and Agarwal , S. P. ( 2003 ), Development of an ophthalmic formulation 
containing ciprofl oxacin - hydroxypropyl - . - cyclodextrin complex , Boll. Chim. Farm. , 
142 ( 5 ), 214 – 219 . 
141. Aktas , Y. , Unlu, N. , Orhan, M. , Irkec , M. , and Hincal, A. A. (2003), Infl uence of hydroxypropyl 
beta - cyclodextrin on the corneal permeation of pilocarpine , Drug Dev. Ind. 
Pharm. , 29 ( 2 ), 223 – 230 . 
142. Harwood , R. J. , and Schwartz, J. B. (1982), Drug release from compression molded fi lms: 
Preliminary studies with pilocarpine , Drug Dev. Ind. Pharm. , 8 ( 5 ), 663 – 682 . 
143. Hsiue , G. H. , Guu , J. A. , and Cheng , C. C. ( 2001 ), Poly(2 - hydroxyethyl methacrylate) fi lm 
as a drug delivery system for pilocarpine , Biomaterials , 22 ( 13 ), 1763 – 1769 . 
144. Bharath , S. , and Hiremath , S. R. ( 1999 ), Ocular delivery systems of pefl oxacin mesylate , 
Pharmazie , 54 ( 1 ), 55 – 58 . 
145. Sultana , Y. , Aqil , M. , and Ali , A. ( 2005 ), Ocular inserts for controlled delivery of pefl oxacin 
mesylate: Preparation and evaluation , Acta Pharma. , 55 ( 3 ), 305 – 314 . 
146. Zimmermann , C. , Drewe , J. , Flammer , J. , and Shaarawy , T. ( 2004 ), In vitro release of 
mitomycin C from collagen implants , Curr. Eye Res. , 28 ( 1 ), 1 – 4 . 
147. Vasantha , V. , Sehgal , P. K. , and Rao , K. P. ( 1988 ), Collagen ophthalmic inserts for pilocarpine 
drug delivery system , Int. J. Pharm. , 47 ( 1 – 3 ), 95 – 102 . 
148. Saettone , M. F. , Giannaccini , B. , Chetoni , P. , Galli , G. , and Chiellini , E. ( 1984 ), Vehicle 
effects in ophthalmic bioavailability: An evaluation of polymeric inserts containing 
pilocarpine , J. Pharm. Pharmacol. , 36 ( 4 ), 229 – 234 . 
149. Saettone , M. F. , Torracca , M. T. , Pagano , A. , Giannaccini , B. , Rodriguez , L. , and Cini , M. 
( 1992 ), Controlled release of pilocarpine from coated polymeric ophthalmic inserts 
prepared by extrusion , Int. J. Pharm. , 86 ( 2 – 3 ), 159 – 166 . 
150. Saettone , M. F. , Chetoni , P. , Bianchi , L. M. , Giannaccini , B. , Conte , U. , and Sangalli , M. 
E. ( 1995 ), Controlled release of timolol maleate from coated ophthalmic mini - tablets 
prepared by compression , Int. J. Pharm. , 126 (1 – 2 ), 79 – 82 . 
151. Sasaki , H. , Tei , C. , Nishida , K. , and Nakamura , J. ( 1993 ), Drug release from an ophthalmic 
insert of a beta - blocker as an ocular drug delivery system , J. Controlled Release , 27 ( 2 ), 
127 – 137 . 
152. Sasaki , H. , Nagano , T. , Sakanaka , K. , Kawakami , S. , Nishida , K. , Nakamura , J. , et al. 
( 2003 ), One - side - coated insert as a unique ophthalmic drug delivery system , J. Controlled 
Release , 92 ( 3 ), 241 – 247 . 
153. Charoo , N. A. , Kohli , K. , Ali , A. , and Anwer , A. ( 2003 ), Ophthalmic delivery of cipro- 
fl oxacin hydrochloride from different polymer formulations: In vitro and in vivo studies , 
Drug Dev. Ind. Pharm. , 29 ( 2 ), 215 – 221 . 
154. Pijls , R. T. , Sonderkamp , T. , Daube , G. W. , Krebber , R. , Hanssen , H. H. L. , Nuijts , R. M. 
M. A. , et al. ( 2005 ), Studies on a new device for drug delivery to the eye , Eur. J. Pharm. 
Biopharm. , 59 ( 2 ), 283 – 288 . 
155. Lux , A. , Maier , S. , Dinslage , S. , Suverkrup , R. , and Diestelhorst , M. ( 2003 ), A comparative 
bioavailability study of three conventional eye drops versus a single lyophilisate , Br. J. 
Ophthalmol. , 87 ( 4 ), 436 – 440 . 
156. Dinslage , S. , Diestelhorst , M. , Weichselbaum , A. , and Suverkrup , R. ( 2002 ), Lyophilisates 
for drug delivery in Ophthalmology: Pharmacokinetics of fl uorescein in the human 
anterior segment , Br. J. Ophthalmol. , 86 ( 10 ), 1114 – 1117 . 
157. Ceulemans , J. , Vermeire , A. , Adriaens , E. , Remon , J. P. , and Ludwig , A. ( 2001 ), Evaluation 
of a mucoadhesive tablet for ocular use , J. Controlled Release , 77 ( 3 ), 333 – 344 . 

REFERENCES 763 
158. Weyenberg , W. , Vermeire , A. , Remon , J. P. , and Ludwig , A. ( 2003 ), Characterization and 
in vivo evaluation of ocular bioadhesive minitablets compressed at different forces , 
J. Controlled Release , 89 ( 2 ), 329 – 340 . 
159. Baeyens , V. , Kaltsatos , V. , Boisrame , B. , Fathi , M. , and Gurny , R. ( 1998 ), Evaluation of 
soluble Bioadhesive Ophthalmic Drug Inserts (BODI) for prolonged release of gentamicin: 
Lachrymal pharmacokinetics and ocular tolerance , J. Ocul. Pharmacol. Ther. , 
14 ( 3 ), 263 – 272 . 
160. Baeyens , V. , Felt - Baeyens , O. , Rougier , S. , Pheulpin , S. , Boisrame , B. , and Gurny , R. 
( 2002 ), Clinical evaluation of bioadhesive ophthalmic drug inserts (BODI(R)) for the 
treatment of external ocular infections in dogs , J. Controlled Release , 85 ( 1 – 3 ), 163 – 168 . 
161. Chetoni , P. , Di Colo , G. , Grandi , M. , Morelli , M. , Saettone , M. F. , and Darougar , S. ( 1998 ), 
Silicone rubber/hydrogel composite ophthalmic inserts: Preparation and preliminary in 
vitro/in vivo evaluation , Eur. J. Pharm. Biopharm. , 46 ( 1 ), 125 – 132 . 
162. Di Colo , G. , and Zambito , Y. ( 2002 ), A study of release mechanisms of different ophthalmic 
drugs from erodible ocular inserts based on poly(ethylene oxide) , Eur. J. Pharm. 
Biopharm. , 54 ( 2 ), 193 – 199 . 
163. Di Colo , G. , Burgalassi , S. , Chetoni , P. , Fiaschi , M. P. , Zambito , Y. , and Saettone , M. F. 
( 2001 ), Relevance of polymer molecular weight to the in vitro/in vivo performances of 
ocular inserts based on poly(ethylene oxide) , Int. J. Pharm. , 220 ( 1 – 2 ), 169 – 177 . 
164. Di Colo , G. , Burgalassi , S. , Chetoni , P. , Fiaschi , M. P. , Zambito , Y. , and Saettone , M. F. 
( 2001 ), Gel - forming erodible inserts for ocular controlled delivery of ofl oxacin , Int. J. 
Pharm. , 215 ( 1 – 2 ), 101 – 111 . 
165. Di Colo , G. , Zambito , Y. , Burgalassi , S. , Serafi ni , A. , and Saettone , M. F. ( 2002 ), Effect 
of chitosan on in vitro release and ocular delivery of ofl oxacin from erodible inserts 
based on poly(ethylene oxide) , Int. J. Pharm. , 248 ( 1 – 2 ), 115 – 122 . 
166. Hornof , M. , Weyenberg , W. , Ludwig , A. , and Bernkop - Schnurch , A. ( 2003 ), Mucoadhesive 
ocular insert based on thiolated poly(acrylic acid): Development and in vivo evaluation 
in humans , J. Controlled Release , 89 ( 3 ), 419 – 428 . 
167. Frucht - Pery , J. , Mechoulam , H. , Siganos , C. S. , Ever - Hadani , P. , Shapiro , M. , and Domb , 
A. ( 2004 ), Iontophoresis - gentamicin delivery into the rabbit cornea, using a hydrogel 
delivery probe , Exper. Eye Res. , 78 ( 3 ), 745 – 749 . 
168. Vandamme , T. F. , and Brobeck , L. ( 2005 ), Poly(amidoamine) dendrimers as ophthalmic 
vehicles for ocular delivery of pilocarpine nitrate and tropicamide , J. Controlled Release , 
102 ( 1 ), 23 – 38 . 
169. Lang , J. C. ( 1995 ), Ocular drug delivery conventional ocular formulations , Adv. Drug 
Deliv. Rev. , 16 ( 1 ), 39 – 43 . 
170. Fitzgerald , P. , and Wilson , C. G. ( 1994 ), Polymeric systems for ophthalmic drug delivery , 
in Dimitriuitra S. , Ed., Polymeric Biomaterials , Marcel Dekker , New York , pp. 
373 – 398 . 
171. Kupferman , A. , Pratt , M. V. , Suckewer , K. , and Leibowitz , H. M. ( 1974 ), Topically applied 
steroids in corneal disease. 3. The role of drug derivative in stromal absorption of dexamethasone 
, Arch. Ophthalmol. , 91 ( 5 ), 373 – 376 . 
172. Wilson , C. G. , Olejnik , O. , and Hardy , J. G. ( 1983 ), Precorneal drainage of polyvinyl 
alcohol solutions in the rabbit assessed by gamma scintigraphy , J. Pharm. Pharmacol. , 
35 ( 7 ), 451 – 454 . 
173. Olejnik , O. , and Weisbecker , C. A. ( 1990 ), Ocular bioavailability of topical prednisolone 
preparations , Clin. Ther. , 12 ( 1 ), 2 – 11 . 
174. Davies , N. M. ( 2000 ), Biopharmaceutical considerations in topical ocular drug delivery , 
Clin. Exp. Pharmacol. Physiol. , 27 ( 7 ), 558 – 562 . 

764 OCULAR DRUG DELIVERY 
175. Davies , N. M. , Wang , G. , and Tucker , I. G. ( 1997 ), Evaluation of a hydrocortisone/ 
hydroxypropyl - [beta] - cyclodextrin solution for ocular drug delivery , Int. J. Pharm. , 
156 ( 2 ), 201 – 209 . 
176. Olejnik , O. ( 1993 ), Conventional systems in ophthalmic drug delivery , in Mitra , A. K. , 
Ed., Ophthalmic Drug Delivery Systems , Marcel Dekker , New York , pp. 177 – 198 . 
177. Schoenwald , R. D. , and Stewart , P. ( 1980 ), Effect of particle size on ophthalmic bioavailability 
of dexamethasone suspensions in rabbits , J. Pharm. Sci. , 69 ( 4 ), 391 – 394 . 
178. MacKeen , D. L. ( 1980 ), Aqueous formulations and ointments , Int. Ophthalmol. Clin. , 
20 ( 3 ), 79 – 92 . 
179. Sieg , J. W. , and Robinson , J. R. ( 1977 ), Vehicle effects on ocular drug bioavailability II: 
Evaluation of pilocarpine , J. Pharm. Sci. , 66 ( 9 ), 1222 – 1228 . 
180. Greaves , J. L. , Wilson , C. G. , and Birmingham , A. T. ( 1993 ), Assessment of the precorneal 
residence of an ophthalmic ointment in healthy subjects , Br. J. Clin. Pharmacol. , 35 ( 2 ), 
188 – 192 . 
181. Lee , V. H. , and Robinson , J. R. ( 1986 ), Topical ocular drug delivery: Recent developments 
and future challenges , J. Ocul. Pharmacol. , 2 ( 1 ), 67 – 108 . 
182. Saettone , M. F. , Giannaccini , B. , Teneggi , A. , Savigni , P. , and Tellini , N. ( 1982 ), Vehicle 
effects on ophthalmic bioavailability: The infl uence of different polymers on the activity 
of pilocarpine in rabbit and man , J. Pharm. Pharmacol. , 34 ( 7 ), 464 – 466 . 
183. Trueblood , J. H. , Rossomondo , R. M. , Wilson , L. A. , and Carlton , W. H. ( 1975 ), Corneal 
contact times of ophthalmic vehicles. Evaluation by microscintigraphy , Arch. Ophthalmol. 
, 93 ( 2 ), 127 – 130 . 
184. Patton , T. F. , and Robinson , J. R. ( 1976 ), Quantitative precorneal disposition of topically 
applied pilocarpine nitrate in rabbit eyes , J. Pharm. Sci. , 65 ( 9 ), 1295 – 1301 . 
185. Patton , T. F. , and Robinson , J. R. ( 1975 ), Ocular evaluation of polyvinyl alcohol vehicle 
in rabbits , J. Pharm. Sci. , 64 ( 8 ), 1312 – 1316 . 
186. Tiffany , J. M. (1991), The viscosity of human tears , Int. Ophthalmol. , 15 ( 6 ), 371 – 376 . 
187. Van Ooteghem , M. ( 1987 ), Factors infl uencing the retention of ophthalmic solutions on 
the eye surface , in Saettone , M. F. , Bucci , M. , and Speiser , P. , Eds., Ophthalmic Drug 
Delivery: Biopharmaceutical, Technological and Clinical Aspects , Vol. 11, Livinia, Padova, 
pp. 7 – 17 
188. Ludwig , A. , van Haeringen , N. J. , Bodelier , V. M. , and Van Ooteghem , M. ( 1992 ), Relationship 
between precorneal retention of viscous eye drops and tear fl uid composition , 
Int. Ophthalmol. , 16 ( 1 ), 23 – 26 . 
189. Greaves , J. L. , Olejnik , O. , and Wilson , C. G. ( 1992 ), Polymers and the precorneal tear 
fi lm , STP Pharma Sci. , 2 , 13 – 33 . 
190. Park , H. , and Robinson , J. R. ( 1987 ), Mechanisms of mucoadhesion of poly(acrylic acid) 
hydrogels , Pharm. Res. , 4 ( 6 ), 457 – 464 . 
191. Robinson , J. R. , and Mlynek , G. M. ( 1995 ), Bioadhesive and phase - change polymers for 
ocular drug delivery , Adv. Drug Deliv. Rev. , 16 ( 1 ), 45 – 50 . 
192. Lehr , C. - M. , Bouwstra , J. A. , Schacht , E. H. , and Junginger , H. E. ( 1992 ), In vitro evaluation 
of mucoadhesive properties of chitosan and some other natural polymers , Int. J. 
Pharm. , 78 ( 1 – 3 ), 43 – 48 . 
193. Park , K. , and Robinson , J. R. ( 1984 ), Bioadhesive polymers as platforms for 
oral - controlled drug Delivery: Method to study bioadhesion , Int. J. Pharm. , 19 ( 2 ), 
107 – 127 . 
194. Meseguer , G. , Gurny , R. , Buri , P. , Rozier , A. , and Plazonnet , B. ( 1993 ), Gamma scintigraphic 
study of precorneal drainage and assessment of miotic response in rabbits 

REFERENCES 765 
of various ophthalmic formulations containing pilocarpine , Int. J. Pharm. , 95 ( 1 – 3 ), 
229 – 234 . 
195. Thermes , F. , Rozier , A. , Plazonnet , B. , and Grove , J. ( 1992 ), Bioadhesion: The effect 
of polyacrylic acid on the ocular bioavailability of timolol , Int. J. Pharm. , 81 ( 1 ), 59 – 
65 . 
196. Duchene , D. , Touchard , F. , and Peppas , N. A. ( 1988 ), Pharmaceutical and medical aspects 
of bioadhesive systems for drug administration , Drug Dev. Ind. Pharm. , 14 ( 2 – 3 ), 
283 – 318 . 
197. Calonge , M. ( 2001 ), The treatment of dry eye , Surv. Ophthalmol. , 45 ( Suppl. 2 ), 
S227 – S239 . 
198. Zignani , M. , Tabatabay , C. , and Gurny , R. ( 1995 ), Topical semi - solid drug delivery: 
Kinetics and tolerance of ophthalmic hydrogels , Adv. Drug Deliv. Rev. , 16 ( 1 ), 51 – 
60 . 
199. Krauland , A. H. , Leitner , V. M. , and Bernkop - Schnurch , A. ( 2003 ), Improvement in the 
in situ gelling properties of deacetylated gellan gum by the immobilization of thiol 
groups , J. Pharm. Sci. , 92 ( 6 ), 1234 – 1241 . 
200. Gurny , R. , Ibrahim , H. , and Buri , P. ( 1993 ), The development and use of in situ formed 
gels, triggered by pH , in Edman , P. , Ed., Biopharmaceutics of Ocular drug Delivery , CRC 
Press , Boca Raton, FL , pp. 81 – 90 . 
201. Greaves , J. L. , Wilson, C. G. , Rozier , A. , Grove , J. , and Plazonnet, B. (1990), Scintigraphic 
assessment of an ophthalmic gelling vehicle in man and rabbit , Curr. Eye Res. , 9 ( 5 ), 
415 – 420 . 
202. Furrer , P. , Plazonnet , B. , Mayer , J. M. , and Gurny , R. ( 2000 ), Application of in vivo confocal 
microscopy to the objective evaluation of ocular irritation induced by surfactants , 
Int. J. Pharm. , 207 ( 1 – 2 ), 89 – 98 . 
203. Kumar , S. , Haglund , B. O. , and Himmelstein , K. J. ( 1994 ), In situ - forming gels for ophthalmic 
drug delivery , J. Ocul. Pharmacol. , 10 ( 1 ), 47 – 56 . 
204. Calvo , P. , Alonso , M. J. , Vila - Jato , J. L. , and Robinson , J. R. ( 1996 ), Improved ocular bioavailability 
of indomethacin by novel ocular drug carriers , J. Pharm. Pharmacol. , 48 ( 11 ), 
1147 – 1152 . 
205. Lallemand , F. , Felt - Baeyens , O. , Besseghir , K. , Behar - Cohen , F. , and Gurny , R. ( 2003 ), 
Cyclosporine A delivery to the eye: A pharmaceutical challenge , Eur. J. Pharm. Biopharm. 
, 56 ( 3 ), 307 – 318 . 
206. Kreuter , J. ( 1990 ), Nanoparticles as bioadhesive ocular drug delivery systems , in Lenaerts 
, V. , and Gurny , R. , Eds., Bioadhesive Drug Delivery Systems , CRC Press , Boca 
Raton, FL , pp. 203 – 212 . 
207. Mainardes , R. M. , and Silva , L. P. ( 2004 ), Drug delivery systems: Past, present, and future , 
Curr. Drug Targets , 5 ( 5 ), 449 – 455 . 
208. Ding , S. ( 1998 ), Recent developments in ophthalmic drug delivery , Pharm. Sci. Technol. 
Today , 1 ( 8 ), 328 – 335 . 
209. Kaur , I. P. , and Kanwar , M. ( 2002 ), Ocular preparations: The formulation approach , Drug 
Dev. Ind. Pharm. , 28 ( 5 ), 473 – 493 . 
210. Meisner , D. , and Mezei , M. ( 1995 ), Liposome ocular delivery systems , Adv. Drug Deliv. 
Rev. , 16 ( 1 ), 75 – 93 . 
211. El - Gazayerly , O. N. , and Hikal , A. H. ( 1997 ), Preparation and evaluation of acetazolamide 
liposomes as an ocular delivery system , Int. J. Pharm. , 158 ( 2 ), 121 – 127 . 
212. Fresta , M. , Panico , A. M. , Bucolo , C. , Giannavola , C. , and Puglisi , G. ( 1999 ), Characterization 
and in - vivo ocular absorption of liposome - encapsulated acyclovir , J. Pharm. Pharmacol. 
, 51 ( 5 ), 565 – 576 . 

766 OCULAR DRUG DELIVERY 
213. Law , S. L. , Huang , K. J. , and Chiang , C. H. ( 2000 ), Acyclovir - containing liposomes for 
potential ocular delivery: Corneal penetration and absorption , J. Controlled Release , 
63 ( 1 – 2 ), 135 – 140 . 
214. Alonso , M. J. , and Sanchez , A. ( 2003 ), The potential of chitosan in ocular drug delivery , 
J. Pharm. Pharmacol. , 55 ( 11 ), 1451 – 1463 . 
215. Carafa , M. , Santucci , E. , and Lucania , G. ( 2002 ), Lidocaine - loaded non - ionic surfactant 
vesicles: Characterization and in vitro permeation studies , Int. J. Pharm. , 231 ( 1 ), 21 – 32 . 
216. Vyas , S. P. , Mysore , N. , Jaitely , V. , and Venkatesan , N. ( 1998 ), Discoidal niosome based 
controlled ocular delivery of timolol maleate , Pharmazie , 53 ( 7 ), 466 – 469 . 
217. Schmalfuss , U. , Neubert , R. , and Wohlrab , W. ( 1997 ), Modifi cation of drug penetration 
into human skin using microemulsions , J. Controlled Release , 46 ( 3 ), 279 – 285 . 
218. Bagwe , R. P. , Kanicky , J. R. , Palla , B. J. , Patanjali , P. K. , and Shah , D. O. ( 2001 ), Improved 
drug delivery using microemulsions: Rationale, recent progress, and new horizons , Crit. 
Rev. Ther. Drug Carrier Syst. , 18 ( 1 ), 77 – 140 . 
219. Attwood , D. ( 1994 ), Microemulsions , in Kreuter , J. , Ed., Colloidal Drug Delivery Systems , 
Marcel Dekker , New York , pp. 31 – 71 . 
220. Benita , S. , and Levy , M. Y. ( 1993 ), Submicron emulsions as colloidal drug carriers for 
intravenous administration: Comprehensive physicochemical characterization , J. Pharm. 
Sci. , 82 ( 11 ), 1069 – 1079 . 
221. Gasco , M. R. , Gallarate , M. , Trotta , M. , Bauchiero , L. , Chiappero , O. , et al. ( 1989 ), Microemulsions 
as topical delivery vehicles: Ocular administration of timolol , J. Pharm. 
Biomed. Anal. , 7 ( 4 ), 433 – 439 . 
222. Beilin , M. , Bar - Ilan , A. , Amselem , S. , Schwarz , J. , Yogev , A. , and Neumann , R. ( 1995 ), 
Ocular retention time of submicron emulsion (SME) and the miotic response to pilocarpine 
delivered in SME , Invest. Ophthalmol. Vis. Sci. , 36 ( 4 ), 166 . 
223. Esfand , R. , and Tomalia , D. A. ( 2001 ), Poly(amidoamine) (PAMAM) dendrimers: From 
biomimicry to drug delivery and biomedical applications , Drug Discov. Today , 6 ( 8 ), 
427 – 436 . 
224. Cloninger , M. J. ( 2002 ), Biological applications of dendrimers , Curr. Opin. Chem. Biol. , 
6 ( 6 ), 742 – 748 . 
225. Patri , A. K. , Majoros , I. J. , Baker , J. , and James R. ( 2002 ), Dendritic polymer macromolecular 
carriers for drug delivery , Curr. Opin. Chem. Biol. , 6 ( 4 ), 466 – 471 . 
226. Stella , V. J. , and Himmelstein , K. J. ( 1980 ), Prodrugs and site - specifi c drug delivery , 
J. Med. Chem. , 23 ( 12 ), 1275 – 1282 . 
227. Lee , V. H. ( 1993 ), Improved ocular drug delivery by use of chemical modifi cations (prodrugs 
), in Edman , P. , Ed., Biopharmaceutics of Ocular Drug Delivery , CRC Press , Boca 
Raton, FL , pp. 121 – 143 . 
228. Lee , V. H. ( 1983 ), Esterase activities in adult rabbit eyes , J. Pharm. Sci. , 72 ( 3 ), 239 – 244 . 
229. Redell , M. A. , Yang , D. C. , and Lee , V. H. L. ( 1983 ), The role of esterase activity in 
the ocular disposition of dipivalyl epinephrine in rabbits , Int. J. Pharm. , 17 ( 2 – 3 ), 
299 – 312 . 
230. Jarvinen , T. , and Jarvinen , K. ( 1996 ), Prodrugs for improved ocular drug delivery , Adv. 
Drug Deliv. Rev. , 19 ( 2 ), 203 – 224 . 
231. Sasaki , H. , Igarashi , Y. , Nagano , T. , Nishida , K. , and Nakamura , J. ( 1995 ), Different effects 
of absorption promoters on corneal and conjunctival penetration of ophthalmic beta - 
blockers , Pharm. Res. , 12 ( 8 ), 1146 – 1150 . 
232. Hochman , J. , and Artursson , P. ( 1994 ), Mechanisms of absorption enhancement and tight 
junction regulation , J. Controlled Release , 29 (Mar.), 253 – 267 . 

REFERENCES 767 
233. Grass , G. M. , and Robinson , J. R. ( 1984 ), Relationship of chemical structure to corneal 
penetration and infl uence of low - viscosity solution on ocular bioavailability , J. Pharm. 
Sci. , 73 ( 8 ), 1021 – 1027 . 
234. Szejtli , J. ( 1994 ), Medicinal applications of cyclodextrins , Med. Res. Rev. , 14 ( 3 ), 
353 – 386 . 
235. Sasaki , H. , Yamamura , K. , Nishida , K. , Nakamura , J. , and Ichikawa , M. ( 1996 ), Delivery 
of drugs to the eye by topical application , Prog. Retin. Eye Res. , 15 ( 2 ), 583 – 620 . 
236. Jarvinen , K. , Jarvinen , T. , Thompson , D. O. , and Stella , V. J. ( 1994 ), The effect of a modi- 
fi ed beta - cyclodextrin, SBE4 - beta - CD, on the aqueous stability and ocular absorption 
of pilocarpine , Curr. Eye Res. , 13 ( 12 ), 897 – 905 . 
237. Jarho , P. , Jarvinen , K. , Urtti , A. , Stella , V. J. , and Jarvinen , T. ( 1996 ), Modifi ed beta - 
cyclodextrin (SBE7 - beta - CyD) with viscous vehicle improves the ocular delivery and 
tolerability of pilocarpine prodrug in rabbits , J. Pharm. Pharmacol. , 48 ( 3 ), 263 – 269 . 
238. Sultana , Y. , Jain , R. , Aqil , M. , and Ali , A. ( 2006 ), Review of ocular drug delivery , Curr. 
Drug Deliv. , 3 ( 2 ), 207 – 217 . 
239. Ranade , V. V. , and Hollinger , M. A. ( 2003 ), Intranasal and ocular drug delivery. in Drug 
Delivery Systems , 2nd ed., CRC Press , Boca Raton, FL , pp. 249 – 287 . 
240. Lee , V. H. ( 1990 ), New directions in the optimization of ocular drug delivery , J. Ocul. 
Pharmacol. , 6 ( 2 ), 157 – 164 . 


769 
5.10 
MICROEMULSIONS AS DRUG 
DELIVERY SYSTEMS 
Raid G. Alany and Jingyuan Wen 
The University of Auckland Auckland, New Zealand 
Contents 
5.10.1 Historical Background, Terminology, and Defi nition 
5.10.2 Structure and Formation of Microemulsion Systems 
5.10.3 Role of Cosurfactants/Cosolvents in Formation and Stabilization of Microemulsion 
Systems 
5.10.4 Pharmaceutical Formulation of Microemulsions 
5.10.4.1 Selection of Microemulsion Ingredients 
5.10.4.2 Phase Behavior Studies 
5.10.5 Techniques Used to Characterize Microemulsions and Related Systems 
5.10.5.1 Polarized Light Microscopy 
5.10.5.2 Transmission Electron Microscopy 
5.10.5.3 Electrical Conductivity Measurements 
5.10.5.4 Viscosity Measurements 
5.10.5.5 Other Characterization Techniques 
5.10.6 Microemulsions as Drug Delivery Systems 
5.10.6.1 Oral Drug Delivery 
5.10.6.2 Transdermal Drug Delivery 
5.10.6.3 Parenteral Drug Delivery 
5.10.6.4 Ocular Drug Delivery 
5.10.7 Concluding Remarks 
References 
5.10.1 HISTORICAL BACKGROUND, TERMINOLOGY, 
AND DEFINITION 
Hoar and Schulman coined the term microemulsion (ME) in 1943 to defi ne a transparent 
system obtained by titrating a turbid oil - in - water (o/w) emulsion with a 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

770 MICROEMULSIONS AS DRUG DELIVERY SYSTEMS 
medium - chain alcohol, namely hexanol. Since then the term has been used to 
describe systems comprising a nonpolar component, an aqueous component, a surfactant, 
and a cosurfactant. While a cosurfactant is usually present, a ME can be 
formulated without a cosurfactant, that is, using a single surfactant. It is important 
to point out that the term ME was (and occasionally is) used in the literature to 
describe various liquid crystalline systems (lamellar, hexagonal, and cubic), surfactant 
systems (micelles and reverse micelles), and even coarse emulsions that are 
micronized using external energy (submicrometer emulsions). To avoid such confusion, 
Danielsson and Lindmann [1] proposed the following defi nition: “ Microemulsion 
is defi ned as a system of water, oil and amphiphile which is optically isotropic 
and a thermodynamically stable liquid solution. ” By this defi nition the following 
systems were excluded: 
• Aqueous solutions of surfactants (micellar and nonmicellar) without additives 
or with water soluble nonelectrolytes as additives 
• Liquid crystalline phases (mesophases) 
• Coarse emulsions, including micronized coarse emulsions 
• Systems that are surfactant free 
The term ME is often incorrectly used in the literature to describe oil and water 
dispersions of small droplet size produced by prolonged ultrasound mixing, high - 
shear homogenization, and microfl uidisation, that is, submicrometer emulsions. The 
major differences between a microemulsion and a coarse emulsion are shown in 
Table 1 . 
5.10.2 STRUCTURE AND FORMATION OF 
MICROEMULSION SYSTEMS 
A ME system can be one of three types depending on the composition: oil in water 
(o/w ME), in which water is the continuous medium; water in oil (w/o ME), in which 
oil is the continuous medium, and water - and - oil bicontinuous ME, in which almost 
equal amounts of water and oil exist [3] . While the three types are quite different 
in terms of microstructure, they all have an interfacial amphiphile monolayer separating 
the oil and water domains. 
The formation of a ME system can be explained using a simplifi ed thermodynamic 
approach and with reference to the equation 
TABLE 1 Comparison of Emulsions and Microemulsions 
Property Emulsion Microemulsion 
Disperse - phase droplet size 0.2 – 10 . m Less than 0.2 . m 
Visual appearance Turbid to milky Transparent to translucent 
Stability Thermodynamically unstable Thermodynamically stable 
Formation Requires energy input Spontaneous 
Source : From ref. 2. 

. . . G A T S = . . (1) 
where . G - free energy of ME formation 
. - interfacial tension at oil – water interface 
. A - change in interfacial area (associated with reducing droplet size) 
S - system entropy 
T - absolute temperature 
The process of ME formation is associated with a reduction in droplet size, which 
results in an increase in the value of . A due to an overall increase in surface area 
that is associated with droplet size reduction. This is compensated by a very low 
interfacial tension that is normally achieved by using relatively high amphiphile 
concentrations. Furthermore, the process of ME formation is accompanied by a 
favorable entropy contribution (increased value of . S ) that is due to the mixing of 
the two immiscible phases, surfactant molecules partitioning in favor of the interface 
rather than the bulk and monomer – micelle surfactant exchange. The net outcome 
is a negative value for . G which translates into a spontaneous ME formation [4] . 
The simplest representation of the ME microstructure is with reference to the 
droplet model in which an interfacial fi lm consisting of amphiphile (surfactant/ 
cosurfactant) molecules surrounds the dispersed droplets (Figures 1 a and b ). The 
orientation of the amphiphile at the interface differs depending on the type of the 
ME. Whether an o/w or w/o ME forms is dependent to a great extent on the volume 
fraction of oil and water as well as the nature of the interfacial fi lm as refl ected 
by the geometry of the amphiphile molecules forming the fi lm. It follows that the 
FIGURE 1 Diagrammatic representation of different types of ME systems: ( a ) w/o ME; 
( b ) o/w ME; ( c ) water - and - oil bicontinuous ME. Droplet diameter for ( a ) and ( b ) is typically 
less than 140 nm. 
(a) (b) 
(c) 
STRUCTURE AND FORMATION OF MICROEMULSION SYSTEMS 771

772 MICROEMULSIONS AS DRUG DELIVERY SYSTEMS 
presence of o/w ME droplets is more likely to happen in systems where the oil 
volume fraction is low, whereas w/o ME droplets form when the water volume fraction 
is low and oil is present in abundance. Interestingly, in systems containing 
comparable amounts of water and oil, a bicontinuous ME may exist (Figure 1 c ). In 
such systems both oil and water exist as microdomains that are separated by an 
amphiphile - stabilized interface with a zero net curvature. 
Mitchell and Ninham [5] extended the theory of self - assembly of surfactant molecules 
forming micelles and bilayers [6] to ME systems. Accordingly, if the volume 
of the surfactant is v , its head group surface area a , and its length l , it follows that 
when the critical packing parameter (CPP = v/al ) has values between 0 and 1, o/w 
MEs are likely to be formed. On the other hand, when CPP is greater than 1, w/o 
MEs are favored. When using surfactants with critical packing parameters close to 
unity (CPP . 1) and at approximately equal volumes of water and oil, the mean 
curvature of the interfacial fi lm approaches zero and droplets may merge into a 
bicontinuous structure (Figure 1 c ). It should be noted, however, that this approach 
is based solely on geometric considerations and does not account for penetration 
of oil and cosurfactant molecules into the interface and the hydration of surfactant 
head groups. 
The ratio of the hydrophilic and the hydrophobic groups of the surfactant 
molecules, that is, their hydrophile – lipophile balance (HLB), is also important in 
determining interfacial fi lm curvature and consequently the structure of the ME. 
The HLB system has been used for the selection of surfactants to formulate MEs 
and accordingly the HLB of the candidate surfactant blend should match the 
required HLB of the oily component for a particular system; furthermore a 
match in the lipophilic part of the surfactant used with the oily component is favorable 
[7] . 
Shinoda and Kuineda [8] highlighted the effect of temperature on the phase 
behavior of systems formulated with two surfactants and introduced the concept of 
the phase inversion temperature (PIT) or the so - called HLB temperature. They 
described the recommended formulation conditions to produce MEs with surfactant 
concentration of about 5 – 10% w/w being (a) the optimum HLB or PIT of a surfactant; 
(b) the optimum mixing ratio of surfactants, that is, the HLB or PIT of the 
mixture; and (c) the optimum temperature for a given nonionic surfactant. They 
concluded that (a) the closer the HLBs of the two surfactants, the larger the cosolubilization 
of the two immiscible phases; (b) the larger the size of the solubilizer, the 
more effi cient the solubilisation process; and (c) mixtures of ionic and nonionic 
surfactants are more resistant to temperature changes than nonionic surfactants 
alone. 
5.10.3 ROLE OF COSURFACTANTS/COSOLVENTS IN FORMATION 
AND STABILIZATION OF MICROEMULSIONS 
Cosurfactants are molecules with weak amphiphilic properties that are mixed with 
the surfactant(s) to enhance their ability to reduce the interfacial tension of a system 
and promote the formation of a ME [3] . Cosolvents have also been described as 
weak amphiphilic molecules that tend to distribute between the aqueous phase, the 

oily phase, and the interfacial layer and act by making the aqueous phase less hydrophilic, 
the oily phase less hydrophobic, and the interfacial fi lm more fl exible and less 
condensed [9, 10] . 
Most single - chain surfactants do not suffi ciently lower the oil – water interfacial 
tension to form MEs, nor are they of the right molecular structure (i.e., HLB) to 
act as cosolvents. To overcome such a barrier, cosurfactant/cosolvent molecules are 
added to further lower the interfacial tension between oil and water, fl uidize the 
hydrocarbon region of the interfacial fi lm, and infl uence the curvature of the fi lm. 
Typically small molecules (C3 – C8) with a polar head (hydroxyl or amine) group 
that can diffuse between the bulk oil and water phase and the interfacial fi lm are 
suitable candidates [11] . 
All the abovementioned mechanisms are expected to facilitate the formation and 
stabilization of ME systems. 
5.10.4 PHARMACEUTICAL FORMULATION OF MICROEMULSIONS 
5.10.4.1 Selection of Microemulsion Ingredients 
Pharmaceutically acceptable ME systems are formulated using at least GRAS - 
(generally regarded as safe) and preferably pharmaceutical - grade ingredients, that 
is, ones already in use in pharmaceutical formulation and devoid of serious side 
effects and toxicity in humans [12] . Nonionic and zwitterionic surfactants are among 
the most commonly used ingredients to formulate pharmaceutical MEs while vegetable 
oils, medium - and long - chain triglycerides, and esters of fatty acids are the 
most commonly used oils [2] . Among the range of nonionic surfactants used are 
sucrose esters [13] , polyoxyethylene alkyl ethers [14] , polyglycerol fatty acid esters 
[15] , polyoxyethylene hydrogenated castor oil [16] , and sorbitan esters [17] . Furthermore, 
systems formulated with zwitterionic phospholipids, particularly lecithin, have 
been widely investigated because of their biocompatible nature [9, 18 – 22] . 
The effect of the oily component on the phase behavior of o/w ME - forming 
systems formulated with nonionic surfactants was reported [23] . The authors showed 
that it is possible to formulate cosurfactant - free o/w ME systems suitable for use as 
drug delivery vehicles using either polyoxyethylene surfactants or amine - N - oxide 
surfactants. The major advantage of these ME systems is their ability to be diluted 
without destroying their integrity; however both classes of surfactants were shown 
to be sensitive to electrolytes. 
The choice of cosurfactants to formulate pharmaceutically acceptable MEs is 
challenging as most of the cosurfactants investigated in fundamental ME research 
cannot be used for the development of pharmaceutically acceptable systems due to 
biocompatibility considerations. Among the pharmaceutically acceptable cosurfactants 
are ethanol [24] , medium - chain mono - and diglycerides [25 – 28] , 1,2 - alkanediols 
[29, 30] and sucrose – ethanol combinations [31] , alkyl monoglucosides, and 
geraniol [32] . Kahlweit et al. [29, 30] reported on the usefulness of certain 1,2 - 
alkanediol cosurfactants as nontoxic substitutes to the physiologically incompatible 
short - and medium - chain alcohols. They suggested the possible use of these components 
for the formulation of nontoxic ME systems. 
PHARMACEUTICAL FORMULATION OF MICROEMULSIONS 773

774 MICROEMULSIONS AS DRUG DELIVERY SYSTEMS 
5.10.4.2 Phase Behavior Studies 
Before a ME can be used as a drug delivery vehicle, the phase behavior of the 
particular combination of the candidate ingredients should be established. This is 
necessary due to the diverse range of colloidal and coarse dispersions that could 
be obtained when oil, water, and an amphiphile blend are mixed. Coarse emulsions, 
vesicles, lyotropic liquid crystals, and micellar systems are some examples. 
A variety of multiphase systems may coexist and the demarcations of the regional 
boundaries become important. One of the most suitable methods to study the 
phase behavior of such systems is to construct a ternary phase diagram using a 
Gibbs triangle (Figure 2 ). A ternary phase diagram can be constructed by two 
methods [33] : 
• Titrating a mixture of two components with the third component 
• Preparing a large number of samples of different composition 
If all mixtures reach equilibrium rapidly, both methods give identical results. For 
mixtures that do not reach equilibrium quickly, the second method is recommended, 
as with the titration method the change in the ratio of components during titration 
may occur too fast, not allowing suffi cient time to visually recognize phase changes 
[33] . 
As the formulation may contain more than three components, the complete 
phase behavior cannot be fully represented using a triangular diagram. However, 
FIGURE 2 Ternary phase diagram used to elucidate ME formation regions. Each of the 
three corners represents 100% of the individual components. Apex S = 100% w/w surfactant 
(0% oil and water), apex W = 100% w/w water (0% oil and surfactant), and apex O = 100% w/ 
w oil (0% water and surfactant). The three lines joining the corner points represent two - 
component systems. The area within the triangle represents all possible combinations of the 
three components. 
W 0 
70 
10 
20 
50 
60 
40 
30 
80
90
100 0 
10 
20 
30
40
50 
60
70 
80
90
100 
0 10 20 30 40 50 60 70 80 90 100 
S 
O 
Increasing 
S 
Increasing 
W 
Increasing O

the phase behavior of a four - component mixture at fi xed pressure and temperature 
can be represented using a tetrahedron. Full characterization of such systems is a 
tedious task requiring a large number of experiments [34] . One acceptable approach 
for representing such systems is by fi xing the mass ratio of two components 
(such as the two amphiphiles) and as such considered a single component. Such an 
approach, although regarded by many as an oversimplifi cation of the systems, is yet 
acceptable for the purpose of phase behavior studies. Such systems are described 
as “ pseudoternary ” as they comprise more than three components (four or possibly 
fi ve) yet are represented using a Gibbs triangle, which is a used to describe the phase 
behavior of a three - component system. 
A novel approach to reduce the experimental effort associated with constructing 
pseudoternary phase diagrams is by using expert systems to predict the phase 
behavior of multicomponent ME - forming systems. Artifi cial neural networks have 
been investigated and were shown to be promising in phase behavior studies 
[17, 35, 36] as well as in the process of ingredient selection [37] . 
5.10.5 TECHNIQUES USED TO CHARACTERIZE MICROEMULSIONS 
AND RELATED SYSTEMS 
The physicochemical and analytical techniques used to characterize MEs and related 
systems could be categorized into those used to: 
• Elucidate the microstructure and monitor phase behavior changes 
• Determine the droplet size of the disperse phase 
The choice of a particular technique is limited by factors such as availability, 
feasibility, and the nature of the information sought. Pharmaceutical scientists are 
more focused on the usefulness of a particular ME system for a drug delivery application 
and the infl uence of the microstructure on that, rather than on the fundamental 
understanding of aspects such as microstructure and phase behavior. 
Polarized light microscopy is a readily available technique that could be used at the 
early formulation development stage to differentiate between isotropic and anisotropic 
systems. Transmission electron microscopy (TEM) is another available 
technique that has been shown to provide microstructural as well as size - related 
information on droplet and bicontinuous ME systems. 
The main disadvantages of TEM applications (such as freeze fracture and cryo - 
TEM) are the lengthy and sophisticated experimental procedures associated with 
sample preparation and the possibility of creating artefacts during sample preparation. 
Other readily available and more user friendly techniques are electrical conductivity 
and viscosity measurements. Electrical conductivity measurements are 
widely used for their simplicity, feasibility, and sensitivity to structural changes in 
systems with increasing water content, particularly systems undergoing percolation 
transitions. Viscosity measurements, on the other hand, require more sophisticated 
instrumentation yet provide useful information on changes in the fl ow properties 
associated with structural changes of the systems. A brief overview of the key characterization 
techniques will follow. 
CHARACTERIZE MICROEMULSIONS AND RELATED SYSTEMS 775

776 MICROEMULSIONS AS DRUG DELIVERY SYSTEMS 
5.10.5.1 Polarized Light Microscopy 
When a mixture of oil, water, and surfactant(s) is examined under a polarized light 
microscope, the textures observed depend on the nature of the surfactant aggregate 
formed and the relative ratio of the comprising constituents. If the resulting aggregates 
are anisotropic, they tend to show strong birefringence and characteristic 
textures could be viewed when examined using a polarized light microscope. On the 
other hand, if the resulting aggregates are isotropic (as with ME systems or coarse 
emulsions), then polarized light microscopy would be of less value in disclosing 
structural information. Thermotropic and lyotropic liquid crystalline (LC) systems 
such as lamellar, hexagonal, and reverse hexagonal mesophases are anisotropic and 
exhibit characteristic textures when viewed under a polarizing light microscope. The 
only type of LC systems that is isotropic and would not display birefringence when 
viewed under a cross - polarizer is the cubic mesophase. 
Polarized light microscopy is a simple technique to learn and use, readily available, 
and of great value to differentiate between various anisotropic LC systems. It 
is also of value to formulation scientists investigating amphiphile – oil – water mixtures 
with emphasis on colloidal systems in general and MEs in particular. This is 
mainly due to the fact that many LC systems may appear transparent to the naked 
eye and can be easily misinterpreted as isotropic ME systems. Thus it becomes 
essential when investigating systems of amphiphile – oil – water to confi rm fi ndings 
based on visual appearance with polarized light microscopic examination. 
5.10.5.2 Transmission Electron Microscopy 
Transmission electron microscopy was one of the earliest techniques used to investigate 
MEs [38, 39] . Freeze fracture along with replication is a sophisticated sample 
preparation method for TEM that requires careful attention to a variety of details 
to avoid formation of sample artefacts. The technique involves rapidly freezing the 
sample by immersing it in a cryogen (slush nitrogen, propane, ethane, and freons). 
For systems with volatile ingredients the freezing must be achieved rapidly to avoid 
phase separation or crystallization. Thus, high cooling rate and adequate environmental 
control of the samples before freezing are critical to prevent loss of volatile 
components. The frozen sample is then transferred to a vacuum chamber and split 
under vacuum by a fracturing device. The fractured surface is then shadowed with 
metal (usually platinum) deposited from one side. The shadowed surface is then 
coated with a layer of carbon that is directly deposited from above the specimen. 
The carbon layer is transparent when examined and forms a supportive backing for 
the shadowing metal deposited on the fractured surface. The specimen is then 
removed from the vacuum and treated with solvents of different polarities, leaving 
the metal carbon fi lm as a replica of the fracture surface. 
Although freeze - fracture TEM provides direct visualization of ME structures, it 
is not currently in wide use probably due to the experimental diffi culties associated 
with the technique. The points to consider when preparing conventional TEM replicas 
are the physical and chemical sample properties, freezing, cleaving, etching, 
replication, cleaning, and mounting steps of the procedure. 
Jahn and Strey [40] investigated systems with varying water - to - oil ratios at constant 
amphiphile concentration. The TEM images support the notion of a bicontinu

ous network for systems containing comparable amounts of the aqueous and oil 
components. The author also showed images of w/o droplet ME systems which 
showed a reduction in number densities of the dispersed phase droplets upon dilution 
with the organic phase. 
Freeze - fracture TEM combined with nuclear magnetic resonance and quasi - 
elastic light scattering was used to study the microstructure of surfactant – water 
systems and dynamics of o/w and bicontinuous ME systems [41] . The authors 
reported a rather abrupt transition from a discontinuous droplet (o/w) to bicontinuous 
(oil - and - water) microstructure occurring at low surfactant concentration, close 
to a three - phase region in the constructed phase diagram of pentaethylene glycol 
dodecyl ether, water, and octane [41] . 
Direct imaging of the ME microstructure using cryo - TEM involves directly investigating 
a thin proportion of the specimen in the frozen hydrated stage by using a 
cryo stage in the transmission electron microscope. Cryo - TEM was used in combination 
with nuclear magnetic resonance (NMR), small - angle X - ray diffraction, and 
small - angle neutron scattering to investigate four - component nonionic systems 
composed of 1 - dodecane, octa - ethylene glycol mono dodecyl ether, n - pentanol, and 
water [42] . These authors reported on the existence of at least two different colloidal 
microstructures, swollen spherical micelles with a diameter of around 8 nm and 
lamellar structures. Both o/w and w/o MEs were also visualized using cryo - TEM 
[43] . 
In the pharmaceutical fi eld, very little has been done to elucidate the microstructure 
of ME systems using electron microscopy. Bolzinger et al. [44] reported on 
bicontinuous sucrose ester - based ME systems for transdermal drug delivery. The 
microstructure of the investigated ME systems was viewed by freeze - fracture TEM. 
The authors showed images of a bicontinuous structure and reported that incorporating 
the anti - infl ammatory drug nifl umic acid into the system did not alter the ME 
microstructure. Alany et al. (2001) reported on the microstructure of ME systems 
formulated using a blend of two nonionic surfactants, ethyl oleate and water with 
and without 1 - butanol. They described two distinct microstructures, namely droplet 
w/o and bicontinuous MEs [35] . Their TEM observations were complemented by 
electrical conductivity and viscosity measurements. 
5.10.5.3 Electrical Conductivity Measurements 
Electrical conductivity measurements can provide valuable information concerning 
the structure and phase behavior of ME systems [45] . Schulman et al. [38] measured 
the conductivity of ME systems, but only in a qualitative way, as they did not monitor 
changes in conductivity that are associated with phase changes. 
Shah and Hamlin [46] studied the changes in electrical resistance associated with 
change in the water - to - oil ratio in an ME system during inversion into various LC 
systems and coarse dispersions. In such systems the electrical conductivity may 
exhibit both maxima and/or minima, refl ecting changes in ion mobility caused 
by variation in viscosity. The most important feature of systems undergoing ME - to - 
LC transition is the gradual change in electrical conductivity with changing 
composition. 
On the other hand, a large electrical conductivity transition has been observed 
in several w/o ME systems [47] . A well - known feature of w/o ME systems is the 
CHARACTERIZE MICROEMULSIONS AND RELATED SYSTEMS 777

778 MICROEMULSIONS AS DRUG DELIVERY SYSTEMS 
steep rise in electrical conductivity as the water concentration increases [48] . This 
sudden change in electrical conductivity has been attributed to the percolation of 
spherical droplets (water droplets surrounded by an amphiphile shell, that is, water - 
swollen reverse micelles) in the oil phase [49] . The conductivity remains low up to 
a certain water volume fraction due to the nonconducting nature of the continuous 
phase of the w/o system. However, as the volume fraction of water reaches and 
exceeds the percolation threshold ( . p ), some of these conductive droplets begin to 
contact each other and form clusters which are suffi ciently close to each other. This 
causes an effi cient transfer of charge carriers between the dispersed droplets by 
charge hopping or transient merging of connected droplets resulting in an exponential 
increase of conductance from an almost zero value to much higher levels. It is 
reasonable to imagine a continuous pathway of water extending through the ME 
system, which by some authors is recognized as a sign of emergence of a bicontinuous 
structure [50] . Different methods were used to estimate the percolation threshold 
( . p ) from the conductivity data by various investigators [35, 48, 51 – 54] . 
5.10.5.4 Viscosity Measurements 
The viscosity of ME systems is also sensitive to structural changes and Newtonian 
fl ow is usually observed. The low viscosity of ME refl ects the fl uid character of the 
overall structure, which is a favorable feature for most ME applications. The pioneering 
work of Attwood et al. [55] on ME systems formulated using liquid paraffi n, 
water, Span 60, and Tween 80 made reference to the equation 
. . .. 
rel = 1 . ( ) a (2) 
where . rel - relative viscosity 
a - viscosity constant with theoretical value of 2.5 for solid spheres 
. - volume fraction of disperse phase 
. - hydrodynamic interaction coeffi cient 
In the same study Attwood et al. [55] investigated the effect of increasing the surfactant 
concentration on the overall viscosity of an o/w ME system and obtained 
values for the viscosity constant a of 3.19 – 4.17. The authors concluded that allowance 
for the hydration of the polyoxyethylene chain of the used surfactant reduced 
the value of the viscosity constant a toward the theoretical value of 2.5 for a solid 
sphere. They also concluded that changing the ratio of the nonionic surfactants did 
not signifi cantly affect the viscosity of the system. 
Baker et al. [56] studied the viscosity of w/o ME systems containing water, xylene, 
sodium alkylbenzenesulfonate, and hexanol using Equation (1) . They reported 
values of the viscosity constant a of 3.3 – 6.0, which is above the theoretical value of 
2.5 for a sphere with an increase in the surfactant concentration. This fi nding was 
attributed to the increase in the ratio of surfactant layer thickness to droplet core 
radius as the surfactant concentration was increased. However, deviation in values 
of the viscosity constant a from the solid - sphere theoretical value of 2.5 could also 
be attributed to changes in the droplet shape or symmetry [57] . 
Viscosity studies have also been carried out to investigate the effect of the surfactant 
and cosurfactant concentrations as well as the surfactant – cosurfactant mass 
ratio on the hydration of the disperse - phase droplets for o/w ME systems [58] . A 

study was conducted on systems composed of isopropyl myristate, water, polysorbate 
80, and sorbitol. The results showed an increase in the viscosity constant and 
a decrease in the hydrodynamic interaction coeffi cient with decreasing surfactant – 
cosurfactant mass ratio. The increase in the viscosity constant resulted from greater 
hydrodynamic volume of droplets as well as the associated increase in the bound 
solvent layer of the droplet core radius from 7 to 22%. 
Kaler et al. [50] reported on the viscosity changes in association with a percolative 
phenomenon for systems containing the commercial surfactant TRS 10 – 80, octane, 
tertiary amyl alcohol, and various brines. Their viscosity results were interpreted as 
evidence for a smooth transition from an oil - continuous to a bicontinuous one in 
which both oil and water span the sample. A second transition was observed and was 
attributed to a transition from a bicontinuous to a water - continuous system. 
Borkovec et al. [59] also reported on a two - stage percolation process for the ME 
AOT (Aerosol OT, bis(2 - ethylhexyl)sodium sulfosuccinate) system AOT – decane – 
water. The structural inversions were investigated using viscosity, conductivity, and 
electro - optical effect measurements. The viscosity results showed a characteristic 
profi le with two maxima, which was interpreted as evidence for two symmetrical 
percolation processes: an oil percolation on the water - rich side of the phase diagram 
and a water percolation process on the oil - rich side. 
Alany et al. [11, 35] reported on the phase behavior of two pharmaceutical ME 
systems showing interesting viscosity changes. The viscosity of both systems increased 
with increasing volume fraction of the dispersed phase to 0.15 and fl ow was Newtonian. 
However, formation of LC in one of the two systems, namely the cosurfactant 
- free system, resulted in a dramatic increase in viscosity that was dependent on 
the volume fraction of the internal phase and a change to pseudoplastic fl ow. In 
contrast, the viscosity of the bicontinuous ME was independent of water volume 
fraction. The authors used two different mathematical models to explain the viscosity 
results and related those to the different colloidal microstructures described. 
5.10.5.5 Other Characterization Techniques 
Among the other techniques used to characterize ME systems with emphasis on 
droplet size determination are optical techniques such as static and dynamic light 
scattering and nonoptical techniques such as small - angle X - ray scattering and small - 
angle neutron scattering [22, 45, 60 – 67] , pulsed fi eld gradient NMR [21, 42, 61, 68 – 
71] , and dielectric measurements [72 – 74] . One limitation of dynamic light scattering 
or so - called photon correlation spectroscopy is the need to either dilute or heat the 
ME system to overcome droplet – droplet interactions as well as microstructure 
changes that are associated with increased viscosity. Such techniques are valuable 
to obtain useful information regarding such interactions, but the downside would 
be the inevitable phase behavior changes that would render these techniques of 
some limited value in determining the original droplet size [75] . 
5.10.6 MICROEMULSIONS AS DRUG DELIVERY SYSTEMS 
ME systems have been attracting increasing interest as vehicles for drug delivery 
via the various routes. Particular emphasis has been put on the oral, transdermal, 
ocular, and parenteral routes. Moreover, these systems have been investigated for 
MICROEMULSIONS AS DRUG DELIVERY SYSTEMS 779

780 MICROEMULSIONS AS DRUG DELIVERY SYSTEMS 
other applications that are relevant to the pharmaceutical, chemical, and biological 
sciences. The most signifi cant development from a pharmaceutical perspective to 
this date would be undoubtedly the launch of the fi rst oral cyclosporin A ME formulation, 
namely Sandimmnue Neoral. Other breakthroughs are likely to follow. 
Some of the key advantages related to ME systems include their thermodynamic 
stability, transparency, ease of preparation, low viscosity, and ultralow interfacial 
tension, to mention a few. 
Furthermore, the presence of nanodomains of different polarity along with an 
interfacial surfactant/cosurfactnat fi lm within the same systems allow for hydrophilic, 
lipophilic, and amphiphilic drugs to be accommodated together if needed. 
For o/w ME systems, their main advantage would be to improve the oral bioavailability 
of class two (II) drugs. These are drugs that display a dissolution - 
dependent bioabsorption [76] . The rationale would be to dissolve the poorly 
water soluble drug in the oil phase, thereby rendering it available in a molecular 
form ready for dissolution. However, it is important to remember that most drugs 
are insoluble in hydrocarbon and to some extent in vegetable oils, rather they 
would require more polar oils such as mixtures of mono - and diglycerides along 
the lines of Miglyol 840. Using such oils results in systems where the poorly 
soluble drug is dispersed as molecules in nanometer - sized oil droplets and as 
such are readily available for dissolution. Moreover, it is worth noting that the 
use of o/w MEs for delivery routes where extensive aqueous dilution is likely is 
less problematic than using w/o ME systems. This is because the external phase 
of an o/w ME will be diluted by water (being the main constituent of biological 
fl uids) and will therefore retain its microstructure. Conversely, the potential use 
of w/o ME systems in the area of drug delivery where there is extensive aqueous 
dilution is rather complicated by the fact that such systems will be diluted by the 
aqueous biological fl uids. This will result in droplet growth and subsequent phase 
changes with potential dose dumping. Despite the aforementioned disadvantages 
w/o MEs offer an option for the formulation of class III drugs. These drugs show 
permeability - limited bioabsorption in vivo [76] . Peptides/proteins, antisense oligonucleotides, 
deoxyribozymes, and small interfering ribonucleic acids (siRNAs) 
are good examples on class II drugs. Such molecules display little or no activity 
when delivered orally and are highly susceptible to the harsh gastrointestinal 
(GI) tract conditions and to their degrading enzymes in vivo. Water - in - oil MEs 
offer an exciting opportunity for optimizing the delivery of such molecules as 
they can be successfully incorporated in the internal (aqueous) phase and therefore 
are denied access to the harsh external conditions. Moreover the internal 
water droplets are likely to act as a nanoreservoir to control the mass transfer 
process of the loaded drug, that is, offer a mechanism for controlled release. In 
addition, the presence of surfactants/cosurfactants as constituents of the formulation 
can serve to increase membrane permeability, thereby improving drug 
absorption and possibly bioavailability. Bicontinuous MEs are the least investigated 
as drug delivery vehicles. They are highly fl uid with low viscosity and 
possess ultralow interfacial tension. These properties render them potential candidates 
for topical and ocular drug delivery where their wetting and spreading 
properties come as an advantage. 
The following section highlights some of the main drug delivery areas where ME 
systems have been researched as potential drug carriers. 

5.10.6.1 Oral Drug Delivery 
The most common method for drug delivery is through the oral route as it offers 
convenience and high patient compliance. However, recent advances in combinatorial 
chemistry is resulting in new molecules with very low water solubility. This leads 
to poor dissolution in the GI tract and subsequent erratic and unpredictable bioabsorption 
post – oral administration. Self - emulsifying drug delivery systems 
(SEDDSs) and self - micro - emulsifying drug delivery systems (SMEDDSs) offer an 
interesting option to optimize the delivery of such problematic drug molecules. 
SMEDDSs can be defi ned as isotropic, anhydrous systems comprising oil and surfactant 
that form o/w MEs upon mild agitation in the presence of water [77, 78] . 
The usefulness of SEDDSs in the area of oral drug delivery has been previously 
reported by Charman and co - workers, who showed improved pharmacodynamic 
properties of the investigational lipophilic drug WIN 54954 [79] . Systems comprising 
the medium - chain triglycerides Captex 355 and 800 in combination with Capmul 
MCM (medium chain mono - and di - glyceride mixture) and polyoxyethylene 20 
sorbitan mono - oleate (Tween 80) were formulated by Constantinides et al. [26, 28, 
80] . The authors reported on the improved bioavailability of calcein, RGD peptide, 
and a water - soluble marker when incorporated using a ME preconcentrate and w/o 
ME in comparison with an aqueous solution serving as a control. The same group 
also reported on the use of ME systems to increase oral absorption of poorly water 
soluble drugs [25] . The authors reported on the ability of the investigated systems 
to increase drug aqueous solubility and improve dissolution and oral bioavailability. 
Surprisingly, the formation of lamellar LC systems upon aqueous dilution of SEDDSs 
was reported to be a characteristic feature of the most effective SEDDSs [81] . Tenjarla 
summarized the factors that are likely to have an effect on the in vivo absorption 
of drugs from ME systems as being phase volume ratios, in vivo droplet size, 
partition coeffi cient of the drug between the two immiscible phases, the presence 
of a drug in an emulsifi ed form or dispersed in oil, site or path of absorption, metabolism 
of the oil in the formulation, excipients that may act as absorption promoters, 
gastric emptying, and drug solubility in the ME excipients [2] . It was also reported 
that peptide uptake from MEs in the GI tract is dependent on particle size, type of 
ME lipid phase, digestability of lipid used, presence of bile slats, lipase, type of surfactants 
in ME, pH, and shedding of enterocytes [82] . 
The most remarkable story of success with ME research and development has 
to do with the oral delivery of cyclosporin A, marketed nowadays under the commercial 
name Sandimmune Neoral. Cyclosporin A is a cyclic peptide used posttransplantation 
surgery as an immune - suppressing agent. Unlike most peptides, 
cyclosporin A is hydrophobic and possesses very limited water solubility. The conventional 
cyclosporin oral formulation (Sandimmune) is in the form of a drug solution 
in olive oil along with ethanol and polyethoxylated oleic acid glycerides. Once 
given orally, the oily solution forms a coarse emulsion and as such behaves as a 
SEDDS with a bioabsorption process that is slow and incomplete. The net outcome 
is fl uctuating drug plasma levels, poor and variable bioavailability, and pronounced 
inter - and intrapatient variability [83] . In an attempt to overcome some of these 
problems, Tarr and Yalkowsky [84] demonstrated that particle size reduction using 
high - shear homogenization can enhance absorption in rats. This improvement in 
oral absorption was attributed to the increased dosage form surface area that is 
MICROEMULSIONS AS DRUG DELIVERY SYSTEMS 781

782 MICROEMULSIONS AS DRUG DELIVERY SYSTEMS 
associated with droplet size reduction. The Neoral formulation is composed of a 
concentrated blend of two surfactants based on medium - chain partial glycerides 
along with an equivalent chain length triglyceride serving as oil, a cosolvent, and 
the drug and is described by the manufacturer as “ microemulsion preconcentrate. ” 
Exposure of this “ preconcentrate ” to water results in the formation of a w/o ME 
that upon further dilution undergoes a phase change into an o/w ME. The improved 
in vivo performance of Neoral over Sandimmune has been demonstrated on multiple 
occasions [83, 85, 86] . Furthermore, the available pharmacokinetic data have 
been reviewed and it was concluded that Neoral offers better predictable and more 
extensive drug absorption than Sandimmune. Other poorly water soluble molecules 
that have been recently formulated in the form of SMEDDSs with the aim of 
improving bioavailability include simvastatin [87] and paclitaxel [88] . 
Water - in - oil microemulsions, on the other hand, offer an exciting opportunity to 
enhance the oral bioavailability of water - soluble peptide drugs. Because of their low 
oral bioavailability, peptide drugs are mostly available as parenteral formulations. 
However, parenteral peptides have an extremely short biological half - life and would 
therefore require multiple daily injections. This is likely to be problematic in chronic 
conditions (insulin for diabetes management is a good example) where patient 
compliance is likely to be an issue. Hydrophilic peptide drugs of this nature can be 
successfully accommodated into the internal aqueous phase of w/o ME systems 
where they are provided with protection from enzymatic degradation post – oral 
administration [89] . Furthermore, the presence of surfactant and some cosurfactants 
(such as medium - chain glycerides) can act to increase GI membrane permeability 
through interacting with the cell membrane bilayer and as such improve oral bioavaialibility 
[25, 26, 80, 89 – 92] . 
One major concern regarding the safety profi le of ME systems intended for oral 
administration is the comparatively high amphiphile content. Both o/w and w/o 
ME systems are amphiphile - rich systems compared to conventional emulsions and 
would contain in the most conservative case up to 15 – 20% w/w surfactant – cosurfactant. 
This is further complicated by the limited models available to evaluate chronic 
toxicology in comparison to conventional oral dosage forms such as tablets [91] . 
5.10.6.2 Transdermal Drug Delivery 
Transdermal drug delivery to the systemic circulation is one of the oldest routes 
that have been exploited using ME systems. This route offers distinct advantages 
compared to traditional routes by avoidance of fi rst - pass metabolism, potential of 
controlled release, ease of administration, and possibility of immediate withdrawal 
of treatment when necessary [93] . Transdermal drug delivery aims at maximizing 
drug fl ux into the systemic circulation through the skin, whereas dermal drug delivery 
aims at targeting either the epidermis or the dermis of the skin. The key challenge 
in both cases is to provide suffi cient increase in drug fl ux with minimal or no 
signifi cant irreversible alteration to the skin barrier function [93] . 
Several studies have reported on the enhanced bioavailability of cutaneous drugs 
using o/w and w/o MEs compared to conventional emulsions, gels or solutions, 
mesophases, micellar and inverse micellar systems, and vesicles [93] . Moreover, a 
diverse range of drug molecules such as ketoprofen, apomorphine, estradiol, lidocaine 
[94 – 97] , indomethacin and diclofenac [98] , prostaglandin E 1 [99] , aceclofenac 

[100] , vinpocetine [101] , azelaic acid [102] , methotrexate [103] , piroxicam [104] , 
triptolide [105] , fl uconazol [106] , and ascorbyl palmitate [107] were incorporated 
into different ME systems. 
Kantaria et al. [108, 109] reported on gelatin ME - based organogels (MBGs) as 
potential iontophoretic systems for the transdermal delivery of drugs. The microstructure 
of the proposed MBG was elucidated using small - angle neutron scattering 
where w/o ME droplets were entrapped in an extensive network of gelatin – water 
percolative channels. Theses MBGs were found to be electrically conducting and 
were shown to successfully deliver a model drug (sodium salicylate). Theses systems 
were formulated using pharmaceutically acceptable ingredients, including Tween 80 
as a surfactant and isopropyl myristate as the oily component [108] . 
Transdermal delivery of proteins and/or DNA vaccines for needle - free immunization 
has been attracting increasing interest. Cui et al. [110] reported on ethanol - 
in - fl uorocarbone (E/F) MEs for topical immunisation. The authors showed that 
plasmid DNA incorporated into E/F MEs was found to be stable. Furthermore, after 
topical application to the skin, signifi cant enhancements in luciferase expression, 
antibody production, and T - helper type 1 based immune response compared to an 
aqueous or ethanolic solutions of DNA were observed [110] . 
One major concern with the topical application of ME systems is their biocompatibility 
and toxicity potential, mostly due to their high surfactant – cosurfactant content. 
Fundamental ME research utilizes ionic surfactants and medium - chain alcohols. 
While these ingredients are interesting from a physicochemical perspective, they pose 
serious biocompatibility and toxicity concerns [75] . Nonionic and zwitter ion – based 
surfactants (such as certain phospholipids) offer a more pharmaceutically acceptable 
alternative. Several research groups have been focusing on formulating ME systems 
using a single surfactant such as lecithin [98, 111, 112] or n - alkyl POE (polyoxyethylene 
ethers) [14, 112, 113] . This approach tends to compromise the phase behavior 
of ME - forming systems. This is usually seen when the ME region in the constructed 
ternary phase diagrams tends to become smaller in size. This translates to less choice 
in terms of ME composition and possibly stability. Alternative cosurfactants with 
improved biocompatibility and lower skin irritation potential have been recently 
introduced. Plurol isostearique has been shown to be compatible with a range of 
surfactants and oils and was capable of providing sizable ME regions. 
In conclusion, topically applied MEs have been shown to signifi cantly increase 
the cutaneous uptake of both lipophilic and hydrophilic drugs. The favorable properties 
of ME systems include the large concentration gradient (between vehicle and 
skin) due to the high drug solubilization power of ME systems without increasing 
drug affi nity to the vehicle compared to conventional topical delivery systems 
[93] . Moreover, the penetration - enhancing properties of the individual surfactant/ 
cosurfactant ingredient, ease of preparation and “ infi nite ” physical stability, and 
good wetting and spreading properties make ME promising for future topical 
applications. 
5.10.6.3 Parenteral Drug Delivery 
Flubiprofen o/w ME systems were prepared and evaluated as vehicles for parenteral 
drug delivery [114] . These systems were formulated using POE 20 sorbitan monolaurate 
(Tween 20) as the surfactant and ethyl oleate as the oil phase. Flubiprofen 
MICROEMULSIONS AS DRUG DELIVERY SYSTEMS 783

784 MICROEMULSIONS AS DRUG DELIVERY SYSTEMS 
solubility in the o/w ME systems was eight times higher than that in an isotonic 
buffer; however, there was no signifi cant differences in the pharmacokinetic parameters 
in rats between the ME formulation and the buffer [114] . Bicontinuous MEs 
designed for intravenous (i.v.) administration have been prepared and characterized 
[71] . The bicontinuous ME system underwent a phase change into an o/w emulsion 
upon aqueous dilution. In vitro investigations revealed small droplets with mean 
size radii of 60 – 200 nm. While solubilization studies were conducted using two drugs, 
namely felodipine and an antioxidant experimental drug (H 290/58), in vivo evaluations 
were conducted using the drug - free formulation. Doses of up to 0.5 mL/kg 
given i.v. to rats did not show any undesirable effects and had no signifi cant effects 
on acid – base balance, blood gases, plasma electrolytes, arterial blood pressure, or 
heart rate [71] . 
Lecithin - based o/w MEs for parenteral use were formulated using polysorbate 
80, IPM (Isopropyl myristate) , lecithin, and water at different lecithin – polysorbate 
80 weight ratios [115] . The formulated systems were shown to be highly stable and 
of minimal toxicity when evaluated in vitro. Phospholipid - based ME formulations 
of all - trans retinoic acid (ATRA) for parenteral administration were prepared and 
tested in vitro [116] . ATRA is effective against acute promyelocytic leukemia with 
highly variable oral bioavailability. Parenteral ME of ATRA was prepared using 
pharmaceutically acceptable ingredients, namely phospholipids and soybean oil. The 
inhibitory effect of ATRA on two human cancer cell lines (HL - 60 and MCF - 7) was 
not affected by incorporation into a ME formulation. 
ME systems intended for parenteral application have to be formulated using 
nontoxic and biocompatible ingredients. The o/w ME systems would be suitable to 
improve the solubility of poorly water soluble drug molecules whereas w/o ME 
systems would be best suited for optimizing the delivery of hydrophilic drug molecules 
that are susceptible to the harsh GI conditions. Moreover, w/o systems can 
serve to prolong the release and mask any potential tissue irritation and site toxicity 
that are caused by intramuscular (i.m.) administration of hydrophilic drug 
molecules. 
5.10.6.4 Ocular Drug Delivery 
Aqueous solutions account for around 90% of the available ophthalmic formulations, 
mainly due to their simplicity and convenience [117] . However, extensive loss 
caused by rapid precorneal drainage and high tear turnover are among the main 
drawbacks associated with topical ocular drug delivery. Only 1 – 5% of the topically 
applied drug reaches the intraocular tissue with the remainder of the instilled dose 
undergoing nonproductive absorption via the conjunctiva or drainage via the nasolacrimal 
duct. This results in drug loss into the systemic circulation and provides 
undesirable systemic side effects [118] . Many strategies have been implemented to 
overcome such delivery challenges. These include the use of thermosetting in situ 
gelling polymer - based systems [119] , nanoparticles, liposomes, and niosomes [120 – 
123] . However, MEs offer a promising alternative as they comprise aqueous and 
oily components and can therefore accommodate both hydrophilic and lipophilic 
drugs. Moreover, they are transparent and thermodynamically stable and possess 
ultralow interfacial tension and therefore offer excellent wetting and spreading 
properties. Further advantages result form possible improvement of solubility and 

REFERENCES 785 
stability of incorporated drugs with potential increase in bioavailability; hence these 
systems could be a suitable alternative to conventional ocular formulations. So far, 
only few investigators [124 – 127] have considered the use of MEs for ocular drug 
delivery. Their work was solely focused on o/w MEs as ocular delivery carriers. 
Recently Alany et al. [128] reported on w/o MEs formulated using a blend of two 
nonionic surfactants (Crillet 4 and Crill 1), an oily component (Crodamol EO), and 
water. These systems were shown to be capable of undergoing a phase change to 
lamellar liquid crystals upon aqueous dilution. The ocular irritation potential of the 
individual components and fi nal formulations was assessed using a modifi ed hen ’ s 
egg chorioallantoic membrane test (HET - CAM), and the preocular retention was 
investigated in the rabbit eye using gamma scintigraphy. The authors demonstrated 
that the retention of ME systems was signifi cantly greater than an aqueous solution. 
The rapid clearance of the w/o ME formulated with 10% water compared to the 
LC system indicated that phase change is less likely to take place in the rabbit eye 
[128] . It was also concluded that w/o MEs may be of value as vehicles for the ocular 
drug delivery of irritant hydrophilic drugs as they appear to have a protective effect 
when evaluated using a modifi ed HET - CAM test [128] . The potential of bicontinuous 
ME systems as vehicles for ocular drug delivery is yet to be investigated. A 
recent review reported on the potential of MEs as ocular drug delivery systems; 
however, submicrometer emulsions and systems requiring energy input to prepare 
were also covered and classifi ed as MEs [129] . 
5.10.7 CONCLUDING REMARKS 
Microemulsions represent an exciting opportunity for pharmaceutical formulators 
and drug delivery scientists. They are easy to prepare and thermodynamically stable. 
Moreover, they can accommodate drugs of different physicochemical properties and 
protect those that are labile. They have the potential to increase the solubility of 
poorly water soluble drugs, enhance the bioavailability of problematic drugs, reduce 
patient variability, and offer an option for controlled drug release. A critical look at 
the current literature shows that exciting and promising research is taking place. It 
is only a matter of time before new ME - based products will fi nd their way to the 
market following the successful introduction of Sandimmune Neoral. 
REFERENCES 
1. Danielsson , I. , and Lindman , B. ( 1981 ), The defi nition of microemulsion , Colloid Surfaces 
, 3 , 391 . 
2. Tenjarla , S. ( 1999 ), Microemulsions: An overview and pharmaceutical applications 
[Review] , Crit. Rev. Ther. Drug Carrier Syst. , 16 ( 5 ), 461 – 521 . 
3. Attwood , D. ( 1994 ), Microemulsions , in Kreuter , J. , Ed., Colloidal Drug Delivery Systems , 
Marcel Dekker , New York , pp 31 – 71 . 
4. Lawrence , M. J. , and Rees , G. D. ( 2000 ), Microemulsion - based media as novel drug 
delivery systems , Adv. Drug Del. Rev. , 45 ( 1 ), 89 – 121 . 
5. Mitchell, D. , and Ninham, W. (1981), Micelles vesicles and microemulsions , J. Chem. Soc. 
Faraday Trans. , 77 ( 2 ), 601 – 629 . 

786 MICROEMULSIONS AS DRUG DELIVERY SYSTEMS 
6. Israelachvili , J. N. , Mitchell , D. , and Ninham , W. ( 1976 ), Theory of self - assembly of hydrocarbon 
amphiphiles into micelles and bilayers , J. Chem. Soc. Faraday Trans. II , 72 , 
1525 – 1568 . 
7. Prince , L. M. ( 1977 ), Formulation , in Prince , L. M. , Ed., Microemulsions: Theory and 
Practice , Academic , New York , pp 33 – 49 . 
8. Shinoda , K. , and Kuineda , H. ( 1973 ), Condition to produce so - called microemulsions: 
Factors to increase mutual solubility of oil and water solubilisers , J. Colloid Interface 
Sci. , 42 , 381 – 387 . 
9. Aboofazeli , R. , Lawrence , C. B. , Wicks , S. R. , and Lawrence , M. J. ( 1994 ), Investigations 
into the formation and characterization of phospholipid microemulsions. Part 3. Pseudo - 
ternary phase diagrams of systems containing water - lecithin - isopropyl myristate and 
either an alkanoic acid, amine, alkanediol, polyethylene glycol alkyl ether or alcohol as 
cosurfactant , Int. J. Pharm. , 111 , 63 – 72 . 
10. Kahlweit , M. , Busse , G. , and Faulhaber , B. ( 1995 ), Preparing microemulsions with alkyl 
monoglucosides and the role of N - alkanols , Langmuir , 11 ( 9 ), 3382 – 3387 . 
11. Alany , R. G. , Rades , T. , Agatonovic - Kustrin , S. , Davies , N. M. , and Tucker , I. G. ( 2000 ), 
Effects of alcohols and diols on the phase behaviour of quaternary systems , Int. J. 
Pharm. , 196 ( 2 ), 141 – 145 . 
12. FDA (rev. Apr. 1, 2001 ), Food and drugs, Chapter I — Food and Drug Administration, in 
Department of Health and Human Services , Ed., Code of Federal Regulations, Title 21 
(Vol. 3), U.S. Government Printing Offi ce , Washington, DC . 
13. Thevenin , M. A. , Grossiord , J. L. , and Poelman , M. C. ( 1996 ), Sucrose esters cosurfactant 
microemulsion systems for transdermal delivery — assessment of bicontinuous structures 
, Int. J. Pharm. , 137 ( 2 ), 177 – 186 . 
14. Malcolmson , C. , Satra , C. , Kantaria , S. , Sidhu , A. , and Lawrence , M. J. ( 1998 ), Effect of 
oil on the level of solubilization of testosterone propionate into nonionic oil - in - water 
microemulsions , J. Pharm. Sci. , 87 ( 1 ), 109 – 116 . 
15. Ho , H. O. , Hsiao , C. C. , and Sheu , M. T. ( 1996 ), Preparation of microemulsions using 
polyglycerol fatty acid esters as surfactant for the delivery of protein drugs , J. Pharm. 
Sci. , 85 ( 2 ), 138 – 143 . 
16. Kuineda , H. , Hasegawa , Y. , John , A. C. , Naito , M. , and Muto , M. ( 1996 ), Phase behaviour 
of polyoxyethylene hydrogenated caster oil in oil / water systems , Colloid Surfaces , 
209 – 216 . 
17. Agatonovic - Kustrin , S. , Glass , B. D. , Wisch , M. H. , and Alany , R. G. ( 2003 ), Prediction of 
a stable microemulsion formulation for the oral delivery of a combination of antitubercular 
drugs using ANN methodology , Pharm. Res. , 20 ( 11 ), 1760 – 1765 . 
18. Aboofazeli , R. , and Lawrence , M. J. ( 1993 ), Investigations into the formation and characterization 
of phospholipid microemulsions. Part 1. Pseudo - ternary phase diagrams of 
systems containing water - lecithin - alcohol - isopropyl myristate , Int. J. Pharm. , 93 (May 
31), 161 – 175 . 
19. Aboofazeli , R. , and Lawrence , M. J. ( 1994 ), Investigations into the formation and characterization 
of phospholipid microemulsions. Part 2. Pseudo - ternary phase diagrams of 
systems containing water - lecithin - isopropyl myristate and alcohol: Infl uence of purity 
of lecithin , Int. J. Pharm. , 106 , 51 – 61 . 
20. Aboofazeli , R. , Patel , N. , Thomas , M. , and Lawrence , M. J. ( 1995 ), Investigations into the 
formation and characterization of phospholipid microemulsions. 4. Pseudo - ternary 
phase diagrams of systems containing water - lecithin - alcohol and oil — the infl uence of 
oil , Int. J. Pharm. , 125 ( 1 ), 107 – 116 . 
21. Shinoda , K. , Araki , M. , Sadaghiani , A. , Khan , A. , and Lindman , B. ( 1991 ), Lecithin - based 
microemulsions; phase behaviour and microstructure , J. Phys. Chem. , 95 , 989 – 993 . 

REFERENCES 787 
22. Saint - Ruth , H. , Attwood , D. , Ktistis , G. , and Taylor , C. J. ( 1995 ), Phase studies and 
particle size analysis of oil - in - water phospholipid microemulsions , Int. J. Pharm. , 116 , 
253 – 261 . 
23. Warisnoicharoen , W. , Lansley , A. B. , and Lawrence , M. J. ( 2000 ), Nonionic oil - in - water 
microemulsions: The effect of oil type on phase behaviour , Int. J. Pharm. , 198 ( 1 ), 7 – 27 . 
24. Park , K. M. , Lee , M. K. , Hwang , K. J. , and Kim , C. K. ( 1999 ), Phospholipid - based microemulsions 
of fl urbiprofen by the spontaneous emulsifi cation process , Int. J. Pharm. , 
183 ( 2 ), 145 – 154 . 
25. Constantinides , P. P. ( 1995 ), Lipid microemulsions for improving drug dissolution 
and oral absorption: physical and biopharmaceutical aspects , Pharm. Res. , 12 (Nov.), 1561 – 
1572 . 
26. Constantinides , P. P. , Lancaster , C. M. , Marcello , J. , Chiossone , D. C. , Orner , D. , Hidalgo , 
I. , et al. ( 1995 ), Enhanced intestinal absorption of an RGD peptide from water - in - oil 
microemulsions of different composition and particle size , J. Controlled Release , 34 , 
109 – 116 . 
27. Constantinides , P. P. , and Yiv , S. H. ( 1995 ), Particle size determination of phase - inverted 
water - in - oil microemulsions under different dilution and storage conditions , Int. J. 
Pharm. , 115 (Mar. 7), 225 – 234 . 
28. Constantinides , P. P. , Welzel , G. , Ellens , H. , Smith , P. L. , Sturgis , S. , Yiv , S. H. , et al. ( 1996 ), 
Water - in - oil microemulsions containing medium - chain fatty acids/salts: Formulation and 
intestinal absorption enhancement evaluation , Pharm. Res. , 13 ( 2 ), 210 – 215 . 
29. Kahlweit , M. , Busse , G. , Faulhaber , B. , and Eibl , H. ( 1995 ), Preparing nontoxic microemulsions 
, Langmuir , 11 ( 11 ), 4185 – 4187 . 
30. Kahlweit , M. , Busse , G. , and Faulhaber , B. ( 1996 ), Preparing microemulsions with alkyl 
monoglucosides and the role of alkanediols as cosolvents , Langmuir , 12 , 861 – 862 . 
31. Joubran , R. , Parris , N. , Lu , D. , and Trevino , S. ( 1994 ), Synergetic effect of sucrose and 
ethanol on formation of triglyceride microemulsions , J. Disper. Sci. Technol. , 15 ( 6 ), 
687 – 704 . 
32. Stubenrauch , C. , Paeplow , B. , and Findenegg , G. H. ( 1997 ), Microemulsions supported 
by octyl monoglucoside and geraniol. 1. The role of alcohol in the interfacial layer , 
Langmuir , 13 , 3652 – 3658 . 
33. Bhargava , H. N. , Narurkar , A. , and Lieb , L. M. ( 1987 ), Using microemulsions for drug 
delivery , Pharm. Technol. , 11 (Mar.), 46 . 
34. Bourrel , M. , and Schechter , R. S. ( 1988 ), The R - ratio , in Bourrel , M. , and Schechter , R. 
S. Eds., Microemulsions and Related Systems , Marcel Dekker , New York , pp 1 – 30 . 
35. Alany , R. G. , Davies , N. M. , Tucker , I. G. , and Rades , T. ( 2001 ), Characterising colloidal 
structures of pseudoternary phase diagrams formed by oil/water/amphiphile systems , 
Drug Dev. Ind. Pharm. , 27 ( 1 ), 33 – 41 . 
36. Alany , R. G. , Agatonovic - Kustrin , S. , Rades , T. , and Tucker , I. G. ( 1999 ), Use of artifi cial 
neural networks to predict quaternery phase systems from limited experimental data , J. 
Pharm. Biomed. Anal. , 19 ( 3 – 4 ), 443 – 452 . 
37. Richardson , C. J. , Mbanefo , A. , Aboofazeli , R. , Lawrence , M. J. , and Barlow , D. J. ( 1997 ), 
Prediction of phase behavior in microemulsion systems using artifi cial neural networks , 
J. Colloid Interface Sci. , 187 ( 2 ), 296 – 303 . 
38. Schulman , J. H. , Stoeckenius ,W. , and Prince , L. M. ( 1959 ), Mechanism of formaution and 
structure of microemulsions by electron microscopy , J. Phys. Chem. , 63 , 1677 – 1685 . 
39. Schulman , J. H. , Stoeckenius , W. , and Prince , L. M. ( 1960 ), The structure of meylin fi gures 
and microemulsions as observed with the electron microscope , Kolloid - Z , 169 , 170 – 
179 . 

788 MICROEMULSIONS AS DRUG DELIVERY SYSTEMS 
40. Jahn , W. , and Strey , R. ( 1988 ), Microstructure of microemulsions by freeze fracture 
electron microscopy , J. Phys. Chem. , 92 , 2294 – 2301 . 
41. Bodet , J. F. , Bellare , J. R. , Davis , H. T. , Scriven , L. E. , and Miller , W. G. ( 1988 ), Fluid 
microstructure transition from globular to bicontinuous midrange microemulsion , 
J. Phys. Chem. , 92 , 1898 – 1902 . 
42. Regev , O. , Ezrahi , S. , Aserin , A. , Garti , N. , Wachtel , E. , and Kaler , E. W. , et al. ( 1996 ), A 
study of microstructure of a four component nonionic microemulsion by cryo - TEM, 
NMR, SAXS and SANS , Langmuir , 12 , 668 – 674 . 
43. Krauel , K. , Davies , N. M. , Hook , S. , and Rades , T. ( 2005 ), Using different structure types 
of microemulsions for the preparation of poly(alkylcyanoacrylate) nanoparticles by 
interfacial polymerization , J. Controlled Release , 106 ( 1 – 2 ), 76 – 87 . 
44. Bolzinger , M. A. , Thevenin , Carduner , T. C. , and Poelman , M. C. ( 1998 ), Bicontinuous 
sucrose ester microemulsion: A new vehicle for topical delivery of nifl umic acid , Int. J. 
Pharm. , 176 ( 1 ), 39 – 45 . 
45. Kahlweit , M. , Strey , R. , Hasse , D. , Kuineda , H. , Schmeling , T. , and Faulhaber , B. , et al. 
( 1987 ), How to study microemulsions , J. Colloid Interface Sci. , 118 , 436 – 451 . 
46. Shah , D. O. , and Hamlin , R. M. J. ( 1971 ), Structure of water in microemulsions: Electrical 
birefringence and nuclear magnetic resonance studies , Sciences , 171 , 483 – 485 . 
47. Clausse , M. , Nicholas - Morgantini , L. , Zarbda , A. , and Touraud , D. ( 1987 ), Water/sodium 
dodecylsulfate/1 - pentanol/ n - dodecanemicroemulsions realms of existence and transport 
properties , in Rosano , H. L. , and Clausse , M. , Eds., Microemulsion Systems , Marcel 
Dekker , New York , pp 387 – 425 . 
48. Lagues , M. , and Sauterey , C. ( 1980 ), Percolation transition in water in oil microemulsions. 
Electrical conductivity measurements , J. Phys. Chem. , 84 , 3503 – 3508 . 
49. Lagourette , B. , Peyrelasse , J. , Boned , C. , and Clausse , M. ( 1979 ), Percolative conduction 
in microemulsion type system , Nature , 281 , 60 – 62 . 
50. Kaler , E. W. , Bennett , K. E. , Davis , H. T. , and Scriven , L. V. ( 1983 ), Toward understanding 
microemulsion microstructure I: A small - angle x - ray scattering study , J. Phys. Chem. , 79 , 
5673 – 5684 . 
51. Fang , J. , and Venable , R. L. ( 1987 ), Conductivity study of the microemulsions system 
sodium dodecyl sulfate - hexylamine - heptane - water , J. Colloid Interface Sci. , 116 , 269 – 
277 . 
52. Gu , G. , Wang , W. , and Yan , H. ( 1996 ), Electric percolation of water - in - oil microemulsions: 
The application of effective medium theory to system sodium dodecylbenzenesulfonate 
(DDBS)/ n - pentanol/ n - heptane/water , J. Colloid Interface Sci. , 178 ( 1 ), 358 – 360 . 
53. Mehta , S. K. , and Bala , K. ( 1995 ), Volumetric and transport properties in microemulsions 
and the point of view of percolation theory , Phys. Rev. E , 51 ( 6 ), 5732 – 5737 . 
54. Ray , S. , Paul , S. , and Moulik , S. P. ( 1996 ), Physicochemical studies on microemulsions: V. 
Additive effects on the performance of scaling equations and activation energy for 
percolation of conductance of water/AOT/heptane microemulsion , J. Colloid Interface 
Sci. , 183 ( 1 ), 6 – 12 . 
55. Attwood , D. , Currie , L. R. J. , and Elworthy , P. H. ( 1974 ), Studies of solubilised micellar 
solutions , J. Colloid Interface Sci. , 46 , 261 – 265 . 
56. Baker , R. C. , Florence , A. T. , Ottewill , R. H. , and Tadros , T. F. ( 1984 ), Investigations 
into the formation and characterization of microemulsions. II. Light scattering conductivity 
and viscosity studies of microemulsions , J. Colloid Interface Sci. , 100 ( 2 ), 332 – 
349 . 
57. Florence , A. T. , and Attwood , D. ( 1998 ), Physicochemical Principles of Pharmacy , 3rd 
ed. , Macmillan , Basingstoke .

REFERENCES 789 
58. Ktistis , G. ( 1990 ), Viscosity study on oil - in - water microemulsions , Int. J. Pharm. , 61 (June 
30), 213 – 218 . 
59. Borkovec , M. , Eicke , H. F. , Hammerich , H. , and Das Gupta , B. ( 1988 ), Two percolation 
process in microemulsions , J. Phys. Chem. , 92 , 206 – 211 . 
60. Patel , N. , Marlow , M. , and Lawrence , M. J. ( 1998 ), Microemulsions: A novel pMD1 
formulation , in Drug Delivery to the Lungs IX , AerosolSociety , London , pp 160 – 
163 . 
61. Giustini , M. , Palazzo , G. , Colafemmina , G. , Dealla Monica , M. , Giomini , M. , and Ceglie , 
A. ( 1996 ), Microstructure and dynamics of the water - in - oil CTAB/ n - pentanol/ m - hexane/ 
water microemulsions: spectroscopic and conductivity study , J. Phys. Chem. , 100 , 
3190 – 3198 . 
62. Schurtenberger , P. , Peng , Q. , Leser , M. E. , and Luisi , P. L. ( 1993 ), Structure and phase 
behaviour of lecithin - based microemulsions: A study of chanin length dependence , 
J. Colloid Interface Sci. , 156 , 43 – 51 . 
63. Shioi , A. , Harada , M. , and Tanabe , M. ( 1995 ), Static light scattering from oil - rich microemulsions 
containing polydispersed cylindrical aggregates in sodium bis(2 - ethylhexyl) 
phosphate system , J. Phys. Chem. , 99 , 4750 – 4756 . 
64. Ktistis , G. ( 1997 ), Effect of polysorbate 80 and sorbitol concentration on in vitro release 
of indomethacin from microemulsions , J. Disper. Sci. Technol. , 18 ( 1 ), 49 – 61 . 
65. Eugster , C. , Rivara , G. , Forni , G. , and Vai , S. ( 1996 ), Marigenol - concentrates comprising 
Taxol and/or Taxan esters as active substances , Panminerva Med. , 38 ( 4 ), 234 – 242 . 
66. Constantinides , P. P. , and Scalart , J. P. ( 1997 ), Formulation and physical characterisation 
of water - in - oil microemulsions containing long versus medium chain length glycerides , 
Int. J. Pharm. , 58 , 57 – 68 . 
67. Hantzschel , D. , Enders , S. , Kahl , H. , and Quitzsch , K. ( 1999 ), Phase behaviour of quaternary 
systems containing carbohydrate surfactants - water - oil - cosurfactant , Phys. Chem. 
Chem. Phys. , 1 , 5703 – 5701 . 
68. Olla , M. , Monduzzi , M. , and Ambrosone , L. ( 1999 ), Microemulsions and emulsions 
in DDAB/water/oil systems , Colloids Surfaces A: Physicochem. Eng. Aspects , 160 , 
392 – 401 . 
69. Carlfors , J. , Blunte , I. , and Schmidt , V. ( 1991 ), Lidocaine in microemulsion — a dermaldelivery 
system , J. Disper. Sci. Technol. , 12 , 467 – 482 . 
70. Angelico , R. , Palazzo , G. , Colafemmina , G. , Cirkel , P. A. , Giustini , M. , and Ceglie , A. 
( 1998 ), Water diffusion and head group mobility in polymer - like reverse micelle: Evidence 
of a sphere - to - rod - sphere transition , J. Phys. Chem. B , 102 , 2883 – 2889 . 
71. von Corswant ,C. , Thoren , P. , and Engstrom , S. ( 1998 ), Triglyceride - based microemulsion 
for intravenous administration of sparingly soluble substances , J. Pharm. Sci. , 87 ( 2 ), 
200 – 208 . 
72. D ’ Angelo , M. , Fioretto , D. , Onori , G. , Palmieri , L. , and Santucci , A. ( 1996 ), Dynamics of 
water - containing sodium bis(2 - ethylhexyl)sulfosuccinate (AOT) reverse micelles: A 
high - frequency dielectric study , Phys. Rev. E , 54 ( 1 ), 993 – 996 . 
73. Feldman , Y. , Kozlovich , N. , Nir , I. , and Garti , N. ( 1997 ), Dielectric spectroscopy of microemulsions 
, Colloid Surfaces , 128 , 47 – 61 . 
74. Cirkel , P. A. , van der Ploeg , J. P. M. , and Koper , G. J. M. ( 1998 ), Branching and percolation 
in lecithin wormlike micelles studied by dielectric spectroscopy , Phys. Rev. E , 57 , 
6875 – 6883 . 
75. Alany , R. G. ( 2001 ), Microemulsions as vehicles for ocular drug delivery: Formulation, 
Physical - chemical characterisation and biological evaluation , Ph. D. thesis, University of 
Otago , Otago . 

790 MICROEMULSIONS AS DRUG DELIVERY SYSTEMS 
76. Amidon , G. L. , Lennernas , H. , Shah , V. P. , and Crison , J. R. ( 1995 ), A theoretical basis 
for a biopharmaceutic drug classifi cation: The correlation of in vitro drug product dissolution 
and in vivo bioavailability , Pharm. Res. , 12 ( 3 ), 413 – 420 . 
77. Greiner , R. W. , and Evans , D. F. ( 1990 ), Spontaneous formation of a water - continuous 
emulsion from water - in - oil microemulsion , Langmuir , 6 , 1793 – 1796 . 
78. Shah , N. H. , Carvajal , M. T. , Patel , C. I. , Infeld , M. H. , and Malick , A. W. ( 1994 ), Self - 
emulsifying drug delivery systems with poly - glycolyzed glycerides for improving in vitro 
dissolution and oral absorption of lipophilic drugs , Int. J. Pharm. , 106 , 15 – 23 . 
79. Charman , S. A. , Charman , W. N. , Rogge , M. C. , Wilson , T. D. , Dutko , F. J. , and Pouton , 
C. W. ( 1992 ), Self - emulsifying drug delivery systems: Formulation and biopharmaceutic 
evaluation of an investigational lipophilic compound , Pharm. Res. , 9 ( 1 ), 87 – 93 . 
80. Constantinides , P. P. , Scalart , J. P. , Lancaster , C. , Marcello , J. , and Smith , P. L. , et al. ( 1994 ), 
Formulation and intestinal absorption enhancement evaluation of water - in - oil microemulsions 
incorporating medium - chain glycerides , Pharm. Res. , 11 , 1385 – 1390 . 
81. Craig , D. Q. M. , Barker , S. A. , Banning , D. , and Booth , S. W. ( 1995 ), An investigation into 
the mechanisms of self - emulsifi cation using particle size analysis and low frequency 
dielectric spectroscopy , Int. J. Pharm. , 114 ( 1 ), 103 – 110 . 
82. Ritschel , W. A. ( 1991 ), Microemulsions for improved peptide absorption from the gastrointestinal 
tract , Methods Findings Exp. Clin. Pharm. , 13 ( 3 ), 205 – 220 . 
83. Holt , D. W. , Mueller , E. A. , Kovarik , J. M. , van Bree , J. B. , and Kutz , K. ( 1994 ), The pharmacokinetics 
of Sandimmun Neoral: a new oral formulation of cyclosporine , Transplant. 
Proc. , 26 ( 5 ), 2935 – 2939 . 
84. Tarr , B. D. , and Yalkowsky , S. H. ( 1989 ), Enhanced intestinal absorption of cyclosporine 
in rats through the reduction of emulsion droplet size , Pharm. Res. , 6 ( 1 ), 40 – 43 . 
85. Kovarik , J. M. , Mueller , E. A. , van Bree , J. B. , Tetzloff , W. , and Kutz , K. ( 1994 ), Reduced 
inter - and intraindividual variability in cyclosporine pharmacokinetics from a microemulsion 
formulation , J. Pharm. Sci. , 83 ( 3 ), 444 – 446 . 
86. Mueller , E. A. , Kovarik , J. M. , van Bree , J. B. , Tetzloff , W. , Grevel , J. , and Kutz , K. ( 1994 ), 
Improved dose linearity of cyclosporine pharmacokinetics from a microemulsion formulation 
, Pharm. Res. , 11 ( 2 ), 301 – 304 . 
87. Kang , B. K. , Lee , J. S. , Chon , S. K. , Jeong , S. Y. , Yuk , S. H. , and Khang , G. , et al. ( 2004 ), 
Development of self - microemulsifying drug delivery systems (SMEDDS) for oral 
bioavailability enhancement of simvastatin in beagle dogs , Int. J. Pharm. , 274 ( 1 – 2 ), 
65 – 73 . 
88. Yang , S. , Gursoy , R. N. , Lambert , G. , and Benita , S. ( 2004 ), Enhanced oral absorption of 
paclitaxel in a novel self - microemulsifying drug delivery system with or without concomitant 
use of P - glycoprotein inhibitors , Pharm. Res. , 21 ( 2 ), 261 – 270 . 
89. Sarciaux , J. M. , Acar , L. , and Sado , P. A. ( 1995 ), Using microemulsion formulations for 
drug delivery of therapeutic peptides , Int. J. Pharm. , 120 , 127 – 136 . 
90. Swenson , E. S. , and Curatolo , W. ( 1992 ), Intestinal permeability enhancement for proteins, 
peptides and other polar drugs: Mechanism and potential toxicity , Adv. Drug Del. 
Rev. , 8 , 39 – 92 . 
91. Swenson , E. S. , Milisen , W. B. , and Curatolo , W. ( 1994 ), Intestinal permeability enhancement: 
Effi cacy, acute local toxicity, and reversibility , Pharm. Res. , 11 ( 8 ), 1132 – 1142 . 
92. Pouton , C. W. ( 1997 ), Formulation of self - emulsifying drug delivery systems for peptides: 
reduced plasma testosterone levels in male rats after a single injection , Int. J. Pharm. , 
25 , 47 – 58 . 
93. Kreilgaard , M. ( 2002 ), Infl uence of microemulsions on cutaneous drug delivery , Adv. 
Drug Del. Rev. , 54 Suppl 1, S77 – S98 . 

REFERENCES 791 
94. Rhee , Y. S. , Choi , J. G. , Park , E. S. , and Chi , S. C. ( 2001 ), Transdermal delivery of ketoprofen 
using microemulsions , Int. J. Pharm. , 228 ( 1 – 2 ), 161 – 170 . 
95. Peira , E. , Scolari , P. , and Gasco , M. R. ( 2001 ), Transdermal permeation of apomorphine 
through hairless mouse skin from microemulsions , Int. J. Pharm. , 226 ( 1 – 2 ), 47 – 51 . 
96. Peltola , S. , Saarinen - Savolainen , P. , Kiesvaara , J. , Suhonen , T. M. , and Urtti , A. ( 2003 ), 
Microemulsions for topical delivery of estradiol , Int. J. Pharm. , 254 ( 2 ), 99 – 107 . 
97. Sintov , A. C. , and Shapiro , L. ( 2004 ), New microemulsion vehicle facilitates percutaneous 
penetration in vitro and cutaneous drug bioavailability in vivo , J. Controlled Release , 
95 ( 2 ), 173 – 183 . 
98. Dreher , F. , Walde , P. , Walther , P. , and Wehrli , E. ( 1997 ), Interaction of a lecithin microemulsion 
gel with human stratum corneum and its effect on transdermal transport , 
J. Controlled Release , 45 ( 2 ), 131 – 140 . 
99. Ho , H. O. , Huang , M. C. , Chen , L. C. , Hsia , A. , Chen , K. T. , and Chiang , H. S. , et al. ( 1998 ), 
The percutaneous delivery of prostaglandin E1 and its alkyl esters by microemulsions , 
Clin. Pharm. J. , 50 , 257 – 266 . 
100. Yang , J. H. , Kim , Y. I. , and Kim , K. M. ( 2002 ), Preparation and evaluation of aceclofenac 
microemulsion for transdermal delivery system , Arch. Pharm. Res. , 25 ( 4 ), 534 – 540 . 
101. Hua , L. , Weisan , P. , Jiayu , L. , and Hongfei , L. ( 2004 ), Preparation and evaluation of 
microemulsion of vinpocetine for transdermal delivery , Pharmazie , 59 ( 4 ), 274 – 278 . 
102. Gasco , M. R. , Gallarate , M. , and Pattarino , F. ( 1991 ), In vitro permeation of azelaic acid 
from viscosized microemulsions , Int. J. Pharm. , 69 ( 3 ), 193 – 196 . 
103. Alvarez - Figueroa , M. J. , and Blanco - Mendez , J. ( 2001 ), Transdermal delivery of 
methotrexate: Iontophoretic delivery from hydrogels and passive delivery from 
microemulsions , Int. J. Pharm. , 215 ( 1 – 2 ), 57 – 65 . 
104. Park , E. S. , Cui , Y. , Yun , B. J. , Ko , I. J. , and Chi , S. C. ( 2005 ), Transdermal delivery of 
piroxicam using microemulsions , Arch. Pharm. Res. , 28 ( 2 ), 243 – 248 . 
105. Mei , Z. , Chen , H. , Weng , T. , Yang , Y. , and Yang , X. ( 2003 ), Solid lipid nanoparticle and 
microemulsion for topical delivery of triptolide , Eur. J. Pharm. Biopharm. , 56 ( 2 ), 
189 – 196 . 
106. El Laithy , H. M. , and El - Shaboury , K. M. ( 2002 ), The development of Cutina lipogels 
and gel microemulsion for topical administration of fl uconazole , AAPS Pharm. Sci. 
Tech. , 3 ( 4 ), E35 . 
107. Jurkovic , P. , Sentjurc , M. , Gasperlin , M. , Kristl , J. , and Pecar , S. ( 2003 ), Skin protection 
against ultraviolet induced free radicals with ascorbyl palmitate in microemulsions , Eur. 
J. Pharm. Biopharm. , 56 ( 1 ), 59 – 66 . 
108. Kantaria , S. , Rees , G. D. , and Lawrence , M. J. ( 1999 ), Gelatin - stabilised microemulsion - 
based organogels: Rheology and application in iontophoretic transdermal drug delivery , 
J. Controlled Release , 60 ( 2 – 3 ), 355 – 365 . 
109. Kantaria , S. , Rees , G. D. , and Lawrence , M. J. ( 2003 ), Formulation of electrically conducting 
microemulsion - based organogels , Int. J. Pharm. , 250 ( 1 ), 65 – 83 . 
110. Cui , Z. , Fountain , W. , Clark , M. , Jay , M. , and Mumper , R. J. ( 2003 ), Novel ethanol- in - 
fl uorocarbon microemulsions for topical genetic immunization , Pharm. Res. , 20 ( 1 ), 
16 – 23 . 
111. Bonina , F. P. , Montenegro , L. , Scrofani , N. , Esposito , E. , Cortesi , R. , and Menegatti , E. , 
et al. ( 1995 ), Effects of phospholipid based formulations on in vitro and in vivo percutaneous 
absorption of methyl nicotinate , J. Controlled Release , 34 ( 1 ), 53 – 63 . 
112. Malcolmson , C. , and Lawrence , M. J. ( 1990 ), A comparison between nonionic micelles 
and microemulsions as a means of incorporating the poorly water soluble drug diazepam 
, J. Pharm. Pharmcol. , Suppl. 42, 6P . 

792 MICROEMULSIONS AS DRUG DELIVERY SYSTEMS 
113. Muller , M. , Mascher , H. , Kikuta , C. , Schafer , S. , Brunner , M. , and Dorner , G. , et al. ( 1997 ), 
Diclofenac concentrations in defi ned tissue layers after topical administration , Clin. 
Pharmacol. Therap. , 62 ( 3 ), 293 – 299 . 
114. Park , K. - M. , and Kim , C. - K. ( 1999 ), Preparation and evaluation of fl urbiprofen - loaded 
microemulsion for parenteral delivery , Int. J. Pharm. , 181 ( 2 ), 173 – 179 . 
115. Moreno , M. A. , Ballesteros , M. P. , and Frutos , P. ( 2003 ), Lecithin - based oil - in - water 
microemulsions for parenteral use: pseudoternary phase diagrams, characterization and 
toxicity studies , J. Pharm. Sci. , 92 ( 7 ), 1428 – 1437 . 
116. Hwang , S. R. , Lim , S. - J. , Park , J. - S. , and Kim , C. - K. ( 2004 ), Phospholipid - based microemulsion 
formulation of all - trans - retinoic acid for parenteral administration , Int. J. 
Pharm. , 276 ( 1 – 2 ), 175 – 183 . 
117. Bourlais , C. L. , Acar , L. , Zia , H. , Sado , P. A. , Needham , T. , and Leverge , R. ( 1998 ), Ophthalmic 
drug delivery systems — recent advances , Prog. Retinal Eye Res. , 17 ( 1 ), 33 – 58 . 
118. Lang , J. C. ( 1995 ), Ocular drug delivery conventional ocular formulations , Adv. Drug 
Del. Rev. Ocular Drug Del. , 16 ( 1 ), 39 – 43 . 
119. Miller , S. C. , and Donovan , M. D. ( 1982 ), Effect of poloxamer 407 gel on the miotic 
activity of pilocarpine nitrate in rabbits , Int. J. Pharm. , 12 ( 2 – 3 ), 147 – 152 . 
120. Fitzgerald , P. , Hadgraft , J. , Kreuter , J. , and Wilson , C. G. ( 1987 ), A . - scintigraphic evaluation 
of microparticulate ophthalmic delivery systems: Liposomes and nanoparticles , Int. 
J. Pharm. , 40 ( 1 – 2 ), 81 – 84 . 
121. Calvo , P. , Vila - Jato , J. L. , and Alonso , M. J. ( 1997 ), Evaluation of cationic polymer - coated 
nanocapsules as ocular drug carriers , Int. J. Pharm. , 153 ( 1 ), 41 – 50 . 
122. Pignatello , R. , Bucolo , C. , Ferrara , P. , Maltese , A. , Puleo , A. , and Puglisi , G. ( 2002 ), 
Eudragit RS100 nanosuspensions for the ophthalmic controlled delivery of ibuprofen , 
Eur. J. Pharm. Sci. , 16 ( 1 – 2 ), 53 – 61 . 
123. Vyas , S. P. , Mysore , N. , Jaitely , V. , and Venkatesan , N. ( 1998 ), Discoidal niosome based 
controlled ocular delivery of timolol maleate , Pharmazie , 53 ( 7 ), 466 – 469 . 
124. Gasco , M. R. , Gallarate , M. , Trotta , M. , Bauchiero , L. , Gremmo , E. , and Chiappero , O. 
( 1989 ), Microemulsions as topical delivery vehicles: Ocular administration of timolol , 
J. Pharm. Biomed. Anal. , 7 ( 4 ), 433 – 439 . 
125. Gallarate , M. , Gasco , M. R. , Trotta , M. , Chetoni , P. , and Saettone , M. F. ( 1993 ), Preparation 
and evaluation in vitro of solutions and o/w microemulsions containing levobunolol 
as ion - pair , Int. J. Pharm. , 100 ( 1 – 3 ), 219 – 225 . 
126. Hasse , A. , and Keipert , S. ( 1997 ), Development and characterization of microemulsions 
for ocular application , Eur. J. Biopharm. , 43 , 179 – 183 . 
127. Siebenbrodt , I. , and Keipert , S. ( 1993 ), Poloxamer - Systems as potential ophthalmics II. 
Microemulsions , Eur. J. Biopharm. , 39 , 25 – 30 . 
128. Alany , R. G. , Rades , T. , Nicoll , J. , Tucker , I. G. , and Davies , N. M. ( 2006 ), W/O microemulsions 
for ocular delivery: Evaluation of ocular irritation and precorneal retention , 
J. Controlled Release , 111 ( 1 – 2 ), 145 – 152 . 
129. Vandamme , T. F. ( 2002 ), Microemulsions as ocular drug delivery systems: Recent developments 
and future challenges , Prog. Retinal Eye Res. , 21 ( 1 ), 15 – 34 . 

793 
5.11 
TRANSDERMAL DRUG DELIVERY 
C. Scott Asbill and Gary W. Bumgarner 
Samford Unversity, Birmingham, Alabama 
Contents 
5.11.1 Introduction 
5.11.2 Physiology and Characteristics of Human Skin 
5.11.2.1 Transepidermal Water Loss and Occlusion 
5.11.2.2 Skin Lipids 
5.11.3 Diffusion 
5.11.4 Drug Candidates for Transdermal Drug Delivery 
5.11.5 In Vitro Testing of Transdermal Devices and Drug Candidates 
5.11.6 Transdermal Patch Design 
5.11.6.1 Membrane - Moderated Patches 
5.11.6.2 Adhesive Matrix 
5.11.7 Commercially Available Patches 
5.11.7.1 Transderm Scop 
5.11.7.2 Catapres TTS 
5.11.7.3 Androderm 
5.11.7.4 Estradiol Transdermal Systems 
5.11.7.5 CombiPatch 
5.11.7.6 Duragesic 
5.11.7.7 Ortho Evra 
5.11.7.8 Oxytrol 
5.11.7.9 Emsam 
5.11.7.10 Daytrana 
5.11.8 Chemical and Physical Approaches to Transdermal Delivery 
5.11.8.1 Chemical Penetration Enhancers 
5.11.8.2 Physical Enhancement Methods 
5.11.9 The Future of Transdermal Drug Delivery 
References 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

794 TRANSDERMAL DRUG DELIVERY 
5.11.1 INTRODUCTION 
During the last decade there has been an explosion of drug candidates in clinical 
trials and a high number of dosage forms approved by the Food and Drug Administration 
(FDA) that utilize the transdermal route of drug delivery (Table 1 ). The 
interest in transdermals can be attributed to the many advantages offered by this 
route of delivery [1] . These advantages include maintaining steady - state drug concentrations 
(Figure 1 ), bypassing fi rst - pass metabolism, mimicking an intravenous 
(IV) infusion, less frequent dosing, and increased patient compliance (Table 2 ). 
The disadvantages of transdermal patches are patient allergies to adhesives found 
in transdermal patches, patch excipients that sometimes produce local irritation, 
potential for abuse or misuse, temperature that can affect the delivery of drugs from 
certain patches, and the inability of most drugs to be delivered transdermally 
(Table 3 ) [2] . 
In the 1960s a proposal was made that suggested the transdermal exposure of 
nitroglycerin to munitions workers resulted in tolerance of the workers to nitroglycerin. 
Long - term exposure of nitroglycerin in high doses can lead to tolerance. 
Workers who were abruptly removed from exposure to the nitroglycerin often 
experienced cardiac arrest and cases of sudden death were sometimes reported [3] . 
Soon after, a correlation was made between the physical and chemical properties 
of nitroglycerin and its signifi cant transdermal penetration [4] . 
TABLE 1 Selected FDA - Approved Transdermal Patches 
Trade Name Generic Name Indication Company 
Androderm Testosterone Testosterone defi ciency Watson Pharma 
Catapres TTS Clonidine Hypertension Boehringer 
Ingelheim 
Climara Estradiol Hormone replacement Berlex 
CombiPatch Estradiol/norethindrone 
acetate 
Hormone replacment Novartis 
Daytrana Methylphenidate Attention - defi cit 
hyperactivity disorder 
Shire 
Pharmaceuticals 
Duragesic Fentanyl Pain managment Janssen 
Emsam Selegiline Depression Somerset 
Pharmaceuticals 
Estraderm Estradiol Hormone replacement Novartis 
Nicoderm CQ Nicotine Smoking cessation GlaxoSmithKline 
Nicotrol Nicotine Smoking cessation Pfi zer 
Ortho Evra Ethinyl 
estradiol/norelgestromin 
Contraception Ortho - Mcneil 
Oxytrol Oxybutynin Urinary incontinence Watson Pharma 
Transderm 
Nitro 
Nitroglycerin Angina Novartis 
Transdrerm 
Scop 
Scopolamine Motion sickness Novartis 
Vivelle Estradiol Hormone replacement Noven 
Vivelle Dot Estradiol Hormone replacement Noven 

Another historical perspective on transdermal drug delivery involved tobacco 
farmers receiving transdermal doses of nicotine from handling tobacco leaves. A 
paper published by Gehlback et al. in 1974 introduced the concept of transdermal 
nicotine [5] . It was common for farm workers who had direct contact with tobacco 
plants during rainy and humid conditions to exhibit symptoms often associated with 
nicotine poisoning. This condition is called green tobacco sickness and has affected 
many farm workers, particularly in states such as Kentucky and North Carolina, 
where the farming of tobacco is signifi cant. Moisture on tobacco leaves from rain 
or dew may contain signifi cant amounts of nicotine due to its high water solubility. 
Farm workers directly handling tobacco leaves often retained the moisture - 
FIGURE 1 Steady - state drug levels achieved by transdermal delivery. 
Steady-state levels of transdermal dose 
Time (h) 
0 10 20 30 40 
Drug 
plasma 
concentration 
Minimum toxic 
concentration 
Minimum therapeutic 
concentration 
TABLE 2 Advantages of Transdermal Drug Delivery 
Excellent for drugs with short half - lives 
Analogous to IV infusion 
Route bypasses fi rst - pass metabolism 
Reduced side effects 
Decreased dosing 
Zero - order kinetics 
Self - administration 
Increased patient compliance 
TABLE 3 Disadvantages of Transdermal Drug Delivery 
Allergies to adhesives 
Product of local irritation 
Potential for abuse or misuse 
Temperature affects delivery 
Drug needs to be potent and have desired physical and 
chemical properties 
INTRODUCTION 795

796 TRANSDERMAL DRUG DELIVERY 
containing nicotine on their clothes or skin. Nicotine, because of its physical and 
chemical properties, is low molecular weight and has excellent transdermal penetration, 
and signifi cant blood levels can lead to many adverse reactions, such as nausea 
and vomiting [6] . 
5.11.2 PHYSIOLOGY AND CHARACTERISTICS OF HUMAN SKIN 
The skin is the largest organ in the human body and has many physiological functions. 
The skin serves to regulate overall body homeostasis, protect the body from 
external pathogens and chemicals, as well as control water loss from the body. The 
skin has three main layers. The epidermis, which is the outermost layer, is the thinnest 
layer of the skin and provides the most signifi cant barrier function [7] . Beneath 
the epidermis, the dermis provides mechanical support to the skin and the third 
layer, immediately under the dermis, is a layer of subcutaneous fat called the 
hypodermis. 
The epidermis consists of fi ve principal layers and is an area of both intense biochemical 
activity and differentiation. These layers are the stratum corneum, stratum 
lucidum, stratum granulosum, stratum spinosum, and stratum basale. The stratum 
corneum (horny layer) is the uppermost layer of the epidermis and the skin. The 
stratum corneum is composed of dead keratinocytes, which are called corneocytes, 
and has an abundance of keratin and lipid structures [8] . The stratum corneum is 
considered the rate - limiting barrier for the diffusion of chemical compounds across 
the skin. The stratum lucidum (clear layer) is composed of two to three layers of 
dead fl attened keratinocytes which appear translucent under a microscope and are 
present only in thick glabrous skin. 
Stratum granulosum (granular layer) is an epidermal layer than consists of three 
to fi ve layers of keratinocytes which have started to fl atten out, and their nuclei and 
organelles have begun to disintegrate. Also, lipid and keratin granules have started 
to form inside the keratinocytes. Eventually the granules will give rise to the brick - 
and - mortar structure of the stratum corneum. 
The stratum spinosum (prickly layer) is composed of several layers of keratinocytes 
which are starting to exhibit histological and biochemical changes that mark 
the beginning of the differentiation process. The shape of the keratinocytes has 
become irregular and enzymes responsible for lipid synthesis are present. 
The stratum basale is the deepest layer of the epidermis and is composed mainly 
of keratinocytes with melanocytes making up approximately 10% of the cell population. 
The stratum basale is one cell layer thick and is a layer of rapid cell division 
where keratinocytes are rapidly dividing and giving rise to the uppermost layers of 
the epidermis. 
The dermis is the largest layer of the skin. It is a region of strong and fl exible 
connective tissue. The dermis consists of two primary layers, the papillary layer and 
the reticular layer. The papillary layer is the smallest layer of the dermis and is 
composed mainly of collagen and elastin fi bers. The reticular layer is the largest 
layer of the dermis and is composed of mainly dense connective tissue. The layer of 
subcutaneous fat found directly beneath the dermis provides insulation and additional 
mechanical support to the skin. 

5.11.2.1 Transepidermal Water Loss and Occlusion 
Transepidermal water loss (TEWL) is a natural occurrence that takes place in the 
skin layers. TEWL is the result of movement of water from the deep skin layers 
across the epidermis into the outside atmosphere. It is a tightly regulated process 
that is controlled by the stratum corneum [9] . Occlusive topical bases and devices, 
such as transdermal patches, block TEWL and cause increased hydration of the skin. 
Hydration of the skin increases the permeation rates of compounds transdermally. 
The occluding effect of transdermal patches is an important mechanism that 
promotes increased diffusion of the compound across the skin into the systemic 
circulation [10] . 
5.11.2.2 Skin Lipids 
As keratinocytes differentiate and move toward the stratum corneum, they synthesize 
an abundance of lipids, much of which are packaged into small organelles called 
lamellar granules [11] . When reaching the stratum corneum, the corneocytes eject 
these lipid granules forming a principal component of the brick - and - mortar structure 
of the stratum corneum. Lipids synthesized in the skin layers are thought to 
arrive from carbon sources derived from acetate obtained from the systemic circulation. 
The role of epidermal lipids in the brick - and - mortar structure of the stratum 
corneum and the barrier function of the skin is well established in the literature 
[12] . Many studies have suggested that when organic solvents such as dimethyl 
sulfoxide (DMSO) are used to dissolve epidermal lipids, an increase in skin permeability 
is found [13] . 
The major lipids found in the stratum corneum are ceramides, fatty acids, and 
cholesterol. Free fatty acids make up 10 – 15% of the lipid mass of the stratum 
corneum and predominantly consist of straight - chain saturated species ranging from 
14 to 28 carbons in length. Cholesterol, a major lipid found in the stratum corneum, 
represents approximately 25% of the total stratum corneum lipid while cholesterol 
sulfate accounts for another 5%. 
Ceramides are considered to be the largest group of lipids found in the stratum 
corneum, representing 50% of the total lipid weight. Six distinct ceramide fractions 
have been isolated and characterized [14] . Ceramide 1 is derived from the linoleate 
- rich acylglucosylceramide. Ceramide 2 consists of straight - chain saturated fatty 
acids amide linked to sphingosine and dihydrosphingosine bases. Ceramide 3 consists 
of saturated fatty acids amide linked to phytosphingosine, which has an additional 
hydroxyl group on carbon 4. Ceramides 4 and 5 both consist of . - hydroxyacids 
amide linked to sphingosines and dihydrosphingosines. Ceramide 6 contains . - 
hydroxyacids amide linked to phytosphingosines. 
It has been suggested that these ceramides form a gel - phase membrane domain 
within the skin. Straight fatty acid chains as well as the small polar head groups on 
the ceramides are thought to produce a tightly packed domain which is less fl uid 
and thereby less permeable than other liquid crystalline domains which are also 
present. Recent evidence using differential scanning calorimetery (DSC) and infrared 
absorption spectroscopy analyses verifi es the presence of gel phases within the 
stratum corneum. 
PHYSIOLOGY AND CHARACTERISTICS OF HUMAN SKIN 797

798 TRANSDERMAL DRUG DELIVERY 
5.11.3 DIFFUSION 
The diffusion of an active ingredient from a transdermal patch through the skin 
layers and into the systemic circulation can be attributed to Fick ’ s law. Fick ’ s law is 
best defi ned as a linear relationship between the fl ux of a chemical and the chemical 
’ s concentration gradient. The concentration gradient is the engine which triggers 
drug diffusion in all directions [15] . The chemicals move from a region of higher 
concentration to a region of lower concentration. Transdermal patches are typically 
loaded with large amounts of active ingredients in order to maximize diffusion. 
Once the drug is absorbed systemically the concentration gradient is maintained 
with “ sink conditions ” existing between the dosage form and the systemic 
circulation. 
5.11.4 DRUG CANDIDATES FOR TRANSDERMAL DELIVERY 
Several characteristics are important in determining which drugs are candidates for 
transdermal drug delivery. These are half - life, molecular weight, lipophilicity, and 
potency. Some pharmaceutical active ingredients have short biological half - lives 
whereas other have very long half - lives. For drugs such as estradiol with half - lives 
of less than 2 h, for the drug to be delivered orally, a frequent dosing schedule must 
be followed due to the rapid clearance of the drug from the systemic circulation. 
This high dosing frequency may be inconvenient and may lead to poor patient 
compliance. However, drugs with short half - lives are excellent candidates for transdermal 
drug delivery because the rate of drug delivery is constant and mimics the 
kinetics offered by an IV infusion. The steady - state drug delivery allows for less 
frequent dosing, which may result in better patient compliance. 
Molecular weight is another property that must be considered when selecting 
potential candidates for transdermal drug delivery. Molecular weight is a major 
determinant in whether a molecule may pass through the restrictive barrier of the 
stratum corneum. Drugs that have low molecular weights have a better chance of 
penetrating the stratum corneum compared to high - molecular - weight compounds 
such as proteins and oligonucleotides, which are too large to passively diffuse across 
the stratum corneum. It has been proposed in the literature that compounds with a 
molecular weight of less than 400 daltons are potential candidates for transdermal 
drug delivery (Table 4 ) [16] . Lipophilicity of the drug is also an important factor 
concerning the chemical ’ s ability to undergo transdermal delivery. Pharmaceutical 
active ingredients must have suffi cient solubility in both the lipid portion and hydrophilic 
regions of the skin in order to have signifi cant permeation. Drugs with a log P 
between . 1.0 and 4.0 can potentially traverse the brick - and - mortar structure of the 
stratum corneum. 
TABLE 4 Physico Chemical Factors That Affect 
Permeation of Compounds 
Small molecular weight 
Melting point < 200 ° C 
Suitable partition coeffi cient > 0.5 . 10 . 3 cm/h 

Another important parameter when selecting transdermal candidates is the 
drug ’ s potency. Only drugs that provide therapeutic effects at low steady - state 
plasma levels are viable candidates. The input rate needed for a transdermal patch 
fl ux can be determined from multiplying the clearance of the drug with the desired 
plasma concentration of the drug at steady state. This is a useful predictor when 
selecting the feasibility of using an active ingredient in a transdermal patch. 
5.11.5 IN VITRO TESTING OF TRANSDERMAL DEVICES AND 
DRUG CANDIDATES 
The ability to test the transdermal penetration of compounds in vitro has been 
investigated extensively over the last decade. Hundreds of studies have shown that 
the permeation rate of compounds can be accurately measured using several available 
in vitro diffusion cells and various types of skin models [17] . 
A typical in vitro permeation cell is composed of both a donor compartment and 
a receptor compartment (Figure 2 ). The model skin or membrane for permeation 
testing is placed between the donor and receptor compartments. The donor compartment 
is where the drug solution, ointment, or transdermal patch is applied. The 
compound then permeates across the membrane into the receptor compartment. 
The receptor compartment typically contains a buffered solution maintained at 
37 ° C and is continuously stirred [18] . Often antibiotics and preservatives are added 
to the receptor compartment to restrict growth of microorganisms and to maintain 
the integrity of the skin model that is being utilized for the permeation study [19] . 
Samples are withdrawn from the receptor compartment at predetermined intervals 
for analysis. Following the withdrawal of samples from the receptor compartment, 
an equal amount of diffusion buffer is added to the volume. 
5.11.6 TRANSDERMAL PATCH DESIGN 
5.11.6.1 Membrane - Moderated Patches 
Membrane - moderated patches have been utilized in many FDA - approved transdermal 
patches such as Duragesic, Transderm Scop, and Catapress TTS. This type 
of patch utilizes a rate - controlling membrane to precisely control the release of 
FIGURE 2 Typical in vitro permeation cell. 
Sampling port 
Donor compartment 
Receptor compartment 
IN VITRO TRANSDERMAL PATCH DESIGN 799

800 TRANSDERMAL DRUG DELIVERY 
active ingredient from the transdermal patch and provides release profi les of active 
ingredients that exhibit zero - order kinetics [20] . The layers of a membrane - 
moderated patch are as follows: The uppermost layer is an occlusive backing that 
is impermeable; next is the reservoir layer, which consists of active ingredient dissolved 
in an appropriate solvent such as mineral oil or ethyl alcohol. The next layer 
is a rate - controlling membrane that is typically composed of ethylene vinyl acetate 
copolymer. Underlying the rate - controlling membrane is a layer of adhesive and 
then fi nally the release liner (Figure 3 ). Commonly burst doses of the active ingredient 
are placed in the adhesive layer to saturate the skin layers and reduce the lag 
time to steady - state blood levels [21] (Figure 1 ). 
5.11.6.2 Adhesive Matrix 
Adhesive matrix patches are increasingly common, and most of the recently 
approved transdermal drug delivery devices utilize this type of technology [22 – 24] . 
Advantages of this type of system are easier to manufacture than membrane - 
moderated patches, smaller in size, and resistant to manufacturing defects which 
could lead to dose dumping. The layers of an adhesive matrix patch consists of the 
backing followed by a layer containing the active ingredient dissolved and mixed 
with adhesive and then the release liner [25] (Figure 3 ). 
5.11.7 COMMERCIALLY AVAILABLE PATCHES 
5.11.7.1 Transderm Scop 
In 1978 Tranderm Scop was the fi rst transdermal patch to receive FDA approval. 
Scopolamine, the active ingredient, is a belladonna alkaloid that is frequently 
used to treat motion sickness and nausea resulting from anesthetics and analgesics. 
Transderm Scop is a membrane - moderated patch that has a three - day life span. It 
has a circular shape with an area (2.5 cm 2 ) approximately the size of a quarter 
[26] . 
FIGURE 3 Diagram of membrane - moderated and adhesive matrix transdermal patches. 
Membrane moderated patch 
Backing 
Reservoir 
Rate-controlling 
membrane 
Adhesive layer 
Adhesive matrix patch 
Adhesive layer 
Backing 
Drug adhesive

5.11.7.2 Catapres TTS 
Catapres TTS is the fi rst and only transdermal patch approved by the FDA for the 
treatment of hypertension. Catapres TTS contains the active ingredient clondidine 
and was approved in 1985 [27] . Catapres TTS is a seven - day patch and is a membrane 
- moderated transdermal patch. The patch comes in three sizes, 0.1, 0.2, and 
0.3 mg/day. In addition there is also a burst dose of clonidine in the adhesive layer. 
The presence of the burst doses provides an immediate - release dose of clonidine 
which promotes rapid systemic levels of the drug. 
5.11.7.3 Androderm 
Androderm is an FDA - approved membrane - moderated transdermal patch that 
delivers testosterone. Androderm is manufactured in two sizes and both are 24 - h 
patches. The round Androderm patch contains 12.2 mg of testosterone and delivers 
a 2.5 - mg dose over a 24 - h period. The larger oval patch contains 24.3 mg of testosterone 
and delivers 5 mg over a 24 - h period. The reservoir of Androderm contains 
testosterone gelled with alcohol and glycerin. 
5.11.7.4 Estradiol Transdermal Systems 
There are several FDA - approved estradiol patches currently on the market. These 
patches are three - to four - day patches used to treat symptoms associated with 
menopause. Estraderm is the only membrane - moderated estradiol system that is on 
the market. Estraderm is available in two sizes: 0.05 and 0.1 - mg/day patches. Vivelle, 
Vivelle Dots, Alora, and Climara are examples of commercially available estradiol 
patches that utilize adhesive matrix patch design. 
5.11.7.5 CombiPatch 
CombiPatch is a three - or four - day patch that delivers both estradiol and norethindrone 
acetate. CombiPatch is available in two sizes: a 9 - cm 2 patch that delivers 
0.05 mg of estradiol per day and 0.14 mg of norethindrone acetate per day and a 
16 - cm 2 patch that delivers 0.05 mg of estradiol per day and 0.25 mg of norethindrone 
acetate per day. Estradiol is a lipophilic compound with a molecular weight of 272. 
The molecular weight of norethindrone acetate is 340. The design of the patch is 
considered an adhesive - matrix - type patch that consists of three layers. The backing 
is comprised of polyolefi n and the adhesive layer contains a silicone adhesive, acrylate 
adhesive, estradiol, norethindrone acetate, oleic acid, and oleyl alcohol. 
5.11.7.6 Duragesic 
Duragesic is a transdermal patch that delivers the potent opioid analgesic fentanyl. 
The life span of the patch is three days and it is manufactured in fi ve sizes: 12, 25, 
50, 75, and 100 . g/h. Duragesic is a membraned - moderated patch and consists of 
four patch layers: a backing layer of polyester fi lm, a drug reservoir that contains 
fentanyl, and U.S. Pharmacopeia (USP) alcohol gelled with hydroxyethyl cellulose, 
COMMERCIALLY AVAILABLE PATCHES 801

802 TRANSDERMAL DRUG DELIVERY 
a rate - controlling membrane made of ethylene - vinyl acetate copolymer, and a layer 
of silicone adhesive with a burst dose of fentanyl. Fentanyl has a molecular weight 
of 336.5 and is a lipophilic drug with signifi cant transdermal permeation. 
5.11.7.7 Ortho Evra 
Ortho Evra was approved by the FDA in 2002 as the world ’ s fi rst transdermal contraceptive 
patch. Ortho Evra contains both ethinyl estradiol and norelgestromin as 
active ingredient. Ortho Evra is a three - layered patch that is a matrix - type design. 
The molecular weight of ethinyl estradiol is 296.41 and the molecular weight of 
norelgestromin is 327.47. The adhesives used in this system are polyisobutylene and 
polybutene. Ortho Evra is a seven - day patch which is cycled three weeks on and 
one week patch free. 
5.11.7.8 Oxytrol 
Oxytrol is a three - or four - day patch used for the treatment of urinary incontinence 
that was recently approved by the FDA. It delivers the active ingredient oxybutynin. 
Oxybutynin is an antispasmotic and anticholinergic agent with a molecular weight 
of 357. Oxybutynin is considered to be a lipophilic drug. Oxytrol is a matrix - type 
patch with a surface area of 39 cm 2 and contains 36 mg of oxybutynin. Oxytrol has 
an in vivo delivery rate of approximately 3.9 mg/day. 
5.11.7.9 Emsam 
Emsam is a once - a - day patch that delivers an active ingredient selegiline that is used 
to treat depression. Selegiline is an irreversible monoamine oxidase inhibitor and 
has a molecular weight of 187.30. Emsam is a matrix - type patch that has three layers 
and is available in three size. The 6 - mg/day patch has a surface area of 20 mg/20 cm 2 , 
the 9 - mg/day patch has a surface area of 30 mg/30 cm 2 , and the 12 - mg/day patch has 
a surface area of 40 mg/40 cm 2 . 
5.11.7.10 Daytrana 
Daytrana is a newly approved methylphenidate transdermal system. It is a 9 - h 
adhesive matrix patch and comes in four sizes: a 12.5 - cm 2 patch that has a delivery 
rate of 1.1 mg/h, an 18.75 - cm 2 patch that has a delivery rate of 1.6 mg/h, a 25 - cm 2 
patch that delivers 2.2 mg/h, and a 37.5 - cm 2 patch that delivers 3.3 mg/h. 
5.11.8 CHEMICAL AND PHYSICAL APPROACHES TO 
TRANSERMAL DELIVERY 
Due to the brick - and - mortar structure of the stratum corneum, the skin is a diffi cult 
layer to permeate across for most active pharmaceutical ingredients. Because of this 
diffusional barrier, new strategies have been developed to allow compounds to 
better penetrate the stratum corneum [28] . These strategies can be defi ned as either 
chemical or physical approaches to disrupting the barrier function of the skin. 

5.11.8.1 Chemical Penetration Enhancers 
Many approaches that have been investigated over the last several decades enhance 
the permeation of compounds across the skin using novel chemical compounds [29] . 
These permeation enhancers are compounds which partition into the stratum 
corneum and promote the passage of topically applied compounds across the skin 
layers by using three possible mechanisms of action (Table 5 ). The mechanisms by 
which these compounds enhance permeability of the skin have been previously 
described by Williams and Barry and are often referred to as the lipid – protein partitioning 
theory [30] . One proposed mechanism of action of permeation enhancers 
and the most common method by which chemical enhancers increase permeation 
is by fl uidizing the intercellular lipid structures that are found within the stratum 
corneum. By interacting with and disorganizing these lipid structures, channels can 
be formed which allow the compound to better diffuse across the rate - limiting 
barrier of the stratum corneum. 
Another method is based on the ability of some permeation enhancers to interact 
with intracellular proteins such as keratins inside corneocytes. The disassembly of 
these proteins structures within the corneocytes allows some compounds to transcellularly 
penetrate the stratum corneum. Also, some enhancers act as vehicles and 
cosolvents, increasing and promoting the partitioning of compounds into the stratum 
corneum (Table 6 ). 
5.11.8.2 Physical Enhancement Methods 
Microneedles Microneedles are a new type of transdermal device that is receiving 
tremendous attention by pharmaceutical companies because of the potential for the 
transdermal delivery of high - molecular - weight compounds. This type of transdermal 
device contains microscopic needles or projections that can be loaded with active 
ingredient [31] . The projections when placed on the skin penetrate beyond the 
stratum corneum into the living epidermis. This allows the compound to bypass 
passage directly through the stratum corneum, which is the rate - limiting barrier for 
TABLE 5 Desired Properties of Chemical Permeation 
Enhancers 
Provide reversible effects in skin layers 
Low systemic bioavailability 
High stability and compatibility with formulations 
Possess no pharmacological activity 
Should be nontoxic and nonirritating 
TABLE 6 Investigated Chemical Permeation Enhancers 
Azone Oleic acid 
Ethano Oxazolidnesl 
Dimethyl sulfoxide Propylene glycol 
Fatty acids Sodium lauryl sulfate 
Lecithin Terpenes 
CHEMICAL AND PHYSICAL APPROACHES TO TRANSERMAL DELIVERY 803

804 TRANSDERMAL DRUG DELIVERY 
passive diffusion. The drug can be loaded into the microneedles and then released 
into the lower skin levels or the microneedle patch may contain a reservoir and the 
drug will penetrate through channels in the stratum corneum that have been created 
by the microneedles. This allows compounds that normally would not passively 
diffuse across the stratum corneum, such as therapeutic proteins, to be delivered 
transdermally. Currently various formulations of microneedle patches are being 
investigated in clinical trials in the United States [32 – 34] . 
Iontophoresis and Sonophoresis In the early 1900s it was discovered that some 
chemical compounds could be delivered into the systemic circulation across the skin 
using an electric current. This phenomenon was later described as iontophoresis. 
Iontophoresis occurs when an electric potential difference is created across the skin 
layers by an electric current and this gradient drives the penetration of both charged 
and uncharged drugs across the skin [35] . 
One of the earliest FDA - approved products that utilized iontophoresis was the 
iontophoretic delivery of pilocaripne as a method for diagnosing cystic fi brosis [36] . 
The sweat of individuals with cystic fi brosis contains large amounts of both sodium 
and chloride ions. The pilocarpine that is delivered into the skin promotes increased 
sweating, which can be easily collected and analyzed. 
Another product that uses iontophoresis has been recently approved by the FDA. 
This device is called Ionsys and is an iontophoretic system that delivers fentanyl 
hydrochloride transdermally [37, 38] . This is a patient - controlled device that provides 
on - demand delivery of fentanyl for up to 24 h or 80 doses. This device contains 
10.8 mg of fentanyl hydrochloride and is designed to deliver a 40 - . g dose of fentanyl 
over a 10 - min period upon activation of the dose button by the patient. 
Another popular physical enhancement method that has routes in physical 
therapy and sports rehabilitation clinics is sonophoresis. Sonophoresis involves the 
use of ultrasound as a source of disrupting intercellular lipid structures in the 
stratum corneum [39, 40] . The sound waves produced by the device induce cavitation 
of the lipids found within the stratum corneum, which then opens channels and 
allows the chemical compound to easily penetrate the skin. This is a safe and reversible 
process that has received much attention in the literature and by pharmaceutical 
companies. 
5.11.9 FUTURE OF TRANSDERMAL DRUG DELIVERY DEVICES 
Through practical application of recombinant deoxyribonucleic acid (DNA) technology 
there has been a recent explosion of biotech drugs that are in clinical trials, 
and the FDA has approved many. Unfortunately, due to the high molecular weights 
and other physical and chemical characteristics of these macromolecules, many 
delivery obstacles exist that prevent these compounds from being delivered transdermally. 
Many physical and chemical approaches are being investigated that 
enhance the delivery of the biotech agents across the skin. 
TransPharma - Medical, an Israeli - based pharmaceutical company, is investigating 
the transdermal delivery of human parathyroid hormone fragment for the treatment 
of osteoporosis in addition to the delivery of human growth hormone [41] . This 
technology utilizes a 1 - cm 2 patch that creates small channels or holes in the stratum 

corneum much like the microneedle technology that was previously discussed. The 
delivery of insulin has been studied for over a decade using transdermal systems. 
Unfortunately, due to the high molecular weight and dosing considerations with 
insulin, it has been diffi cult. However, insulin delivery transdermally has recent successes 
clinically using sonophoresis, permeation enhancers, iontophoresis, and 
microneedles [42, 43] . 
REFERENCES 
1. Robinson , D. H. , and Mauger , J. W. ( 1991 ), Drug delivery systems , Am. J. Hosp. Pharm ., 
48 ( 10 Suppl. 1 ), S14 – 23 . 
2. Murphy , M. , and Carmichael , A. J. ( 2000 ), Transdermal drug delivery systems and skin 
sensitivity reactions. Incidence and management , Am. J. Clin. Dermatol ., 1 ( 6 ), 361 – 368 . 
3. Forman , S. A. , Helmkamp , J. C. , and Bone , C. M. ( 1987 ), Cardiac morbidity and mortality 
associated with occupational exposure to 1,2 propylene glycol dinitrate , J. Occup. Med ., 
29 ( 5 ), 445 – 450 . 
4. Ben - David , A. ( 1989 ), Cardiac arrest in an explosives factory worker due to withdrawal 
from nitroglycerin exposure , Am. J. Ind. Med ., 15 ( 6 ), 719 – 722 . 
5. Gehlbach , S. H. , et al . ( 1974 ), Green - tobacco sickness. An illness of tobacco harvesters , 
JAMA , 229 ( 14 ), 1880 – 1883 . 
6. Curwin , B. D. , et al . ( 2005 ), Nicotine exposure and decontamination on tobacco harvesters 
’ hands , Ann. Occup. Hyg ., 49 ( 5 ), 407 – 413 . 
7. Xhaufl aire - Uhoda , E. , et al . ( 2006 ), Dynamics of skin barrier repair following topical 
applications of miconazole nitrate , Skin Pharmacol. Physiol ., 19 ( 5 ), 290 – 294 . 
8. Norlen , L. , and Al - Amoudi , A. ( 2004 ), Stratum corneum keratin structure, function, and 
formation: The cubic rod - packing and membrane templating model , J. Invest. Dermatol ., 
123 ( 4 ), 715 – 732 . 
9. Fluhr , J. W. , Feingold , K. R. , and Elias , P. M. ( 2006 ), Transepidermal water loss refl ects 
permeability barrier status: Validation in human and rodent in vivo and ex vivo models , 
Exp. Dermatol ., 15 ( 7 ), 483 – 492 . 
10. Casiraghi , A. , et al . ( 2002 ), Occlusive properties of monolayer patches: In vitro and in 
vivo evaluation , Pharm. Res ., 19 ( 4 ), 423 – 426 . 
11. Wertz , P. W. ( 2000 ), Lipids and barrier function of the skin , Acta Derm. Venereol. Suppl. 
(Stockh.) , 208 , 7 – 11 . 
12. Wertz, P. W. (1997), Integral lipids of hair and stratum corneum, Exs , 78 , 227 – 237 . 
13. Astley , J. P. , and Levine , M. ( 1976 ), Effect of dimethyl sulfoxide on permeability of human 
skin in vitro , J. Pharm. Sci ., 65 ( 2 ), 210 – 215 . 
14. Wertz , P. W. , et al . ( 1985 ), The composition of the ceramides from human stratum corneum 
and from comedones , J. Invest. Dermatol ., 84 ( 5 ), 410 – 412 . 
15. Barry , B. W. ( 2002 ), Drug delivery routes in skin: A novel approach , Adv. Drug Deliv. Rev ., 
54 ( Suppl 1 ), S31 – 40 . 
16. Buchwald , P. , and Bodor , N. ( 2001 ), A simple, predictive, structure - based skin permeability 
model , J. Pharm. Pharmacol ., 53 ( 8 ), 1087 – 1098 . 
17. El - Kattan , A. , Asbill , C. S. , and Haidar , S. ( 2000 ), Transdermal testing: Practical aspects 
and methods , Pharm. Sci. Technol. Today , 3 ( 12 ), 426 – 430 . 
18. Asbill , C. , et al . ( 2000 ), Evaluation of a human bio - engineered skin equivalent for drug 
permeation studies , Pharm. Res ., 17 ( 9 ), 1092 – 1097 . 
REFERENCES 805

806 TRANSDERMAL DRUG DELIVERY 
19. El - Kattan , A. F. , Asbill , C. S. , and Michniak , B. B. ( 2000 ), The effect of terpene enhancer 
lipophilicity on the percutaneous permeation of hydrocortisone formulated in HPMC gel 
systems , Int. J. Pharm ., 198 ( 2 ), 179 – 189 . 
20. Krishnaiah , Y. S. , and Bhaskar , P. ( 2004 ), Studies on the transdermal delivery of 
nimodipine from a menthol - based TTS in human volunteers , Curr. Drug Deliv ., 1 ( 2 ), 
93 – 102 . 
21. Misra , A. , et al . ( 1997 ), Biphasic testosterone delivery profi le observed with two different 
transdermal formulations , Pharm. Res ., 14 ( 9 ), 1264 – 1268 . 
22. Burkman , R. T. ( 2004 ), The transdermal contraceptive system , Am. J. Obstet. Gynecol ., 
190 ( 4 , Suppl), S49 – 53 . 
23. Sane , N. , and McGough , J. J. ( 2002 ), MethyPatch Noven , Curr. Opin. Invest. Drugs , 3 ( 8 ), 
1222 – 1224 . 
24. Amsterdam , J. D. ( 2003 ), A double - blind, placebo - controlled trial of the safety and effi - 
cacy of selegiline transdermal system without dietary restrictions in patients with major 
depressive disorder , J. Clin. Psychiatry , 64 ( 2 ), 208 – 214 . 
25. Chien , Y. W. ( 1985 ), Microsealed drug delivery systems: Fabrications and performance , 
Methods Enzymol ., 112 , 461 – 470 . 
26. Parrott , A. C. ( 1989 ), Transdermal scopolamine: A review of its effects upon motion sickness, 
psychological performance, and physiological functioning , Aviat. Space Environ. 
Med ., 60 ( 1 ), 1 – 9 . 
27. Burris , J. F. , and Mroczek , W. J. ( 1986 ), Transdermal administration of clonidine: A new 
approach to antihypertensive therapy , Pharmacotherapy , 6 ( 1 ), 30 – 34 . 
28. Trommer , H. , and Neubert , R. H. ( 2006 ), Overcoming the stratum corneum: The modulation 
of skin penetration. A review , Skin Pharmacol. Physiol ., 19 ( 2 ), 106 – 121 . 
29. Xu , P. , and Chien , Y. W. ( 1991 ), Enhanced skin permeability for transdermal drug delivery: 
Physiopathological and physicochemical considerations , Crit. Rev. Ther. Drug Carrier 
Syst ., 8 ( 3 ), 211 – 236 . 
30. Williams , A. C. , and Barry , B. W. ( 1991 ), Terpenes and the lipid - protein - partitioning theory 
of skin penetration enhancement , Pharm. Res ., 8 ( 1 ), 17 – 24 . 
31. Henry , S. , et al . ( 1998 ), Microfabricated microneedles: A novel approach to transdermal 
drug delivery , J. Pharm. Sci ., 87 ( 8 ), 922 – 925 . 
32. Giudice , E. L. , and Campbell , J. D. ( 2006 ), Needle - free vaccine delivery , Adv. Drug Del. 
Rev ., 58 ( 1 ), 68 – 89 . 
33. Aqil , M. , Sultana , Y. , and Ali , A. ( 2006 ), Transdermal delivery of beta - blockers , Expert 
Opin. Drug Deliv , 3 ( 3 ), 405 – 418 . 
34. Cormier , M. , et al . ( 2004 ), Transdermal delivery of desmopressin using a coated microneedle 
array patch system , J. Controlled Release , 97 ( 3 ), 503 – 511 . 
35. Fischer , G. A. ( 2005 ), Iontophoretic drug delivery using the IOMED Phoresor system , 
Expert Opin. Drug Deliv ., 2 ( 2 ), 391 – 403 . 
36. Parad , R. B. , et al . ( 2005 ), Sweat testing infants detected by cystic fi brosis newborn screening 
, J. Pediatr ., 147 ( 3, Suppl ), S69 – 72 . 
37. Miaskowski , C. ( 2005 ), Patient - controlled modalities for acute postoperative pain management 
, J. Perianesth. Nurs ., 20 ( 4 ), 255 – 267 . 
38. Sinatra , R. ( 2005 ), The fentanyl HCl patient - controlled transdermal system (PCTS): An 
alternative to intravenous patient - controlled analgesia in the postoperative setting , Clin. 
Pharmacokinet ., 44 ( Suppl 1 ), 1 – 6 . 
39. Mitragotri , S. , and Kost , J. ( 2000 ), Low - frequency sonophoresis: A noninvasive method 
of drug delivery and diagnostics , Biotechnol. Prog ., 16 ( 3 ), 488 – 492 . 

40. Mitragotri , S. , Blankschtein , D. , and Langer , R. ( 1996 ), Transdermal drug delivery using 
low - frequency sonophoresis , Pharm. Res , 13 ( 3 ), 411 – 420 . 
41. Levin , G. , et al . ( 2005 ), Transdermal delivery of human growth hormone through RF - 
microchannels , Pharm. Res ., 22 ( 4 ), 550 – 555 . 
42. Rastogi , S. K. , and Singh , J. ( 2005 ), Effect of chemical penetration enhancer and iontophoresis 
on the in vitro percutaneous absorption enhancement of insulin through porcine 
epidermis , Pharm. Dev. Technol ., 10 ( 1 ), 97 – 104 . 
43. King , M. J. , et al . ( 2002 ), Transdermal delivery of insulin from a novel biphasic lipid system 
in diabetic rats , Diabetes Technol. Ther ., 4 ( 4 ), 479 – 488 . 
REFERENCES 807


809 
5.12 
VAGINAL DRUG DELIVERY 
Jose das Neves , Maria Helena Amaral , and 
Maria Fernanda Bahia 
University of Porto, Porto, Portugal 
Contents 
5.12.1 Introduction 
5.12.2 The Human Vagina 
5.12.2.1 Anatomy 
5.12.2.2 Histology 
5.12.2.3 Physiology 
5.12.2.4 Childhood, Pregnancy, and Menopause 
5.12.3 General Features of Vaginal Drug Delivery 
5.12.3.1 Advantages and Disadvantages of Vaginal Drug Delivery 
5.12.3.2 Permeability and Drug Absorption 
5.12.3.3 First - Uterine - Pass Effect 
5.12.4 Vaginal Drug Delivery Systems 
5.12.4.1 Overview 
5.12.4.2 Excipients 
5.12.4.3 Solid Systems 
5.12.4.4 Semisolid Systems 
5.12.4.5 Liquid Systems 
5.12.4.6 Vaginal Rings 
5.12.4.7 Vaginal Films 
5.12.4.8 Medicated Vaginal Tampons 
5.12.4.9 Vaginal Foams 
5.12.4.10 Vaginal Sponges 
5.12.4.11 Other Strategies and Vaginal Drug Delivery Systems 
5.12.4.12 Packaging and Vaginal Applicators 
5.12.5 Pharmaceutical Evaluation of Vaginal Drug Delivery Systems 
5.12.5.1 Legal and Offi cial Compendia Requirements 
5.12.5.2 Drug Release and Permeability 
5.12.5.3 pH and Acid - Buffering Capacity 
5.12.5.4 Rheological Studies 
5.12.5.5 Textural Studies 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

810 VAGINAL DRUG DELIVERY 
5.12.5.6 Mucoadhesion 
5.12.5.7 Vaginal Distribution and Retention 
5.12.5.8 Safety and Toxicology 
5.12.5.9 Other Characteristics 
5.12.6 Clinical Usage and Potential of Vaginal Drug Delivery 
5.12.6.1 Microbicides 
5.12.6.2 Antimicrobials 
5.12.6.3 Hormonal Contraceptives and Hormonal Replacement Therapy 
5.12.6.4 Spermicides 
5.12.6.5 Labor Inducers and Abortifacients 
5.12.6.6 Proteins and Peptides 
5.12.6.7 Vaccines 
5.12.6.8 Other Uses 
5.12.7 Acceptability and Preferences of Women Related to Vaginal Drug Delivery 
5.12.8 Veterinary Vaginal Drug Delivery 
5.12.9 Conclusions and Future Trends 
References 
5.12.1 INTRODUCTION 
The vagina has been used for a long time as a route for drug administration, being 
as old as medicine and pharmacy themselves. Throughout the history of human 
civilization, vaginal administration of drugs has been practiced and recorded until 
the modern era. Some of the fi rst records were found in Egypt, where the Kahun 
Papyrus, the oldest of the surviving medical papyri (ca. 1850 b . c .), included references 
to vaginal “ preparations ” containing substances such as mud, frankincense, 
oil, malachite, ass urine, myrrh, crocodile dung, honey, and sour milk, normally used 
in female genitalia – related conditions and contraception [1] . Latter papyri such as 
the Ramesseum Papyrus (ca. 1700 b . c .), the Ebers Papyrus (ca. 1550 b . c .), and the 
Greater Berlin Papyrus (ca. 1300 b . c .) also contained drug formulations to be 
administered in the vagina. Vaginal administration of drugs continued to be carried 
out by other civilizations from ancient Greece and Rome to the Middle Ages, 
in the Arabic and Oriental cultures, passing through the Renaissance, until our 
days [1, 2] . 
Although traditionally used for local action, some drugs can permeate the vaginal 
mucosa and reach the bloodstream in suffi cient concentrations to have systemic 
effects. Current understanding of the vaginal anatomy, physiology, and pathophysiology 
is very considerable, contrasting with the still limited knowledge of the possibilities 
of vaginal drug delivery. Nonetheless, interest and contribution of 
pharmaceutical scientists toward vaginal drug delivery development have increased 
in the last years in response to the specifi c needs of this route of drug administration. 
Indeed, vaginal drug delivery has seen recent advances that make it very 
promising, in particular therapeutic fi elds such as the prevention of human immunodefi 
ciency virus (HIV) and other sexually transmitted infections, contraception, 
hormone replacement therapy in menopausal women, and labor induction. 

Therefore, this chapter discusses the main features of vaginal anatomy, physiology, 
and histology related to drug delivery as well as vaginal drug delivery systems 
and their evaluation. Also, past and current usage of the vagina as a route of drug 
administration, ongoing investigation, and promising strategies in this fi eld are 
addressed. 
5.12.2 THE HUMAN VAGINA 
The knowledge of the human vagina ’ s anatomy, histology, and physiology is fundamental 
in the design of drug delivery systems, helping researchers optimize vaginal 
products. Pharmaceutical scientists must comprehend the particularities of this 
organ in order to overcome its natural limitations and enhance its advantages over 
other drug delivery routes. 
5.12.2.1 Anatomy 
The organs of the female reproductive tract are classically divided into the internal 
and external genitalia [3] . The internal genital organs include the vagina, cervix, 
uterus, oviducts, ovaries, and surrounding supporting structures, as seen in Figure 1 . 
The external genital organs, also known as the vulva or pudendum, are composed 
by the structures that surround the vaginal entrance, which are visible in the perineal 
area. These include the mons pubis, labia majora, labia minora, hymen, clitoris, 
vestibule, urethra, vestibular glands (Skene glands and Bartholin glands), and vestibular 
bulbs. 
THE HUMAN VAGINA 811 
FIGURE 1 Female reproductive system and related structures: vagina (1), cervix (2), uterus 
(3), ovary (4), fallopian tube (5), urinary bladder (6), urethra (7), anus (8), rectum (9), colon 
(10), vestibule (11), and pubic symphysis (12). (Courtesy of Lu i s Paup e rio.) 

812 VAGINAL DRUG DELIVERY 
The vagina is a tubular, fi bromuscular organ approximately 9 cm long, extending 
from the cervix to the vestibule, being positioned between the urinary bladder and 
the rectum (Figure 1 ). Because of the protrusion of the cervix into the upper part 
of the vagina, two deep recessions (anterior and posterior) are formed, called fornices, 
the posterior fornix being considerably larger than the anterior one. Also, two 
lateral fornices can be considered [3, 4] . In the adult woman, the anterior wall of 
the vagina is approximately 7.5 cm long, while the posterior wall extends by approximately 
9 cm. The width of the vagina varies throughout its length, the transverse 
diameter being higher at the level of the fornices, decreasing progressively toward 
the vaginal entrance. The vagina is commonly misunderstood as a straight tube, 
descending from the cervix to the introitus. The axes of the upper and lower vagina 
are different, forming a slight S - shaped curve: The axis of the lower vagina in relation 
to a standing woman is vertical and posterior, while the upper part (from the 
pelvic diaphragm to the cervix) becomes more horizontal, with the fi nal portion of 
the vagina curving toward the hollow of the sacrum [3, 5] . The angle between these 
two portions is approximately 130 ° C. This fact is important in drug delivery, taking 
into consideration that pharmaceutical formulations must be retained in the vagina, 
resisting to gravity forces. The walls of the vagina are covered by a mucosal tissue 
that forms a series of transverse folds denominated rugae (more prominent in the 
lower third of the vagina), thus increasing the available surface for absorption and 
allowing considerable distension of this organ during penile penetration and childbirth 
[4, 6] . Indeed, the surface area of the vaginal mucosa is an important parameter 
concerning its coverage by vaginal products or drug absorption. Results by Pendergrass 
et al. varied through a wide range of values (65.73 – 107.07 mm 2 ), with a mean 
surface area of 87.46 mm 2 , although these results can be an understatement because 
of limitations in the determination method [7] . In a more recent study, Barnhart 
et al. found higher surface values which ranged from 103.9 to 165.0 mm 2 . These variable 
results suggest that one volume of vaginal product may not be appropriate for 
all women [5] . The walls of the vagina are normally in apposition and fl attened in 
the anteroposterior diameter. Thus the vagina has the appearance of the letter H in 
cross section, although a W shape may also be observed [8] . 
The vagina has a high blood supply, which is primarily provided by the vaginal 
branches of the internal iliac artery. To a minor extension, blood is also supplied 
through branches of the uterine, middle rectal, and internal pudendal arteries. Blood 
drainage from the vagina returns through vaginal veins (the perineum venous 
plexus), running parallel to the course of the arteries, which fl ow into the pudendal 
vein, internal iliac vein, and then the vena cava, thus bypassing the portal circulation. 
Blood supply increases with sexual excitement, particularly to the clitoris and vestibular 
bulbs, causing the erection of these structures along with the expansion and 
elongation of the vagina, allowing the accommodation of the male penis [4] . The 
lymphatic drainage is characterized by its wide distribution and frequent crossovers 
between both sides of the pelvis. Generally, the upper third of the vagina drains to 
the external iliac nodes, the middle third to the common and internal iliac nodes, 
and the lower third to the common iliac, superfi cial inguinal, and perirectal nodes 
[3] . 
Sensory innervation of the vagina is provided by the pudendal plexus and is 
particularly developed near the vaginal orifi ce. The nervous sympathetic innervation 
of the vagina is provided by the hypogastric plexus, while the nervous parasympa

thetic innervation derives from the second and third sacral nerves. There is a lack 
of free nerve endings in the upper two - thirds of the vagina [3, 4] . Thus, objects placed 
in this area, such as vaginal drug delivery systems, are likely to be unperceived by 
women. 
5.12.2.2 Histology 
The vaginal tissue is composed of four distinct layers: stratifi ed squamous epithelium, 
lamina propria, muscular layer, and tunica adventitia, the fi rst two usually 
referred to as vaginal mucosa (Figure 2 ). The epithelial layer is about 150 – 200 . m 
thick, corresponding approximately to 30 – 45 layers of cells, being nonkeratinized. 
The epithelium presents different types of cells, making it possible to identify fi ve 
different layers, namely the basal, parabasal, intermediate, transitional, and superfi - 
cial layers. Epithelial cells are closely joined by numerous desmosomes and occasionally 
tight junctions, the last being particularly abundant in the basal layer. 
Deposits of a lipid material are found near the surface of the epithelium, acting as 
a permeability barrier to large water - soluble molecules. The lamina propria, or 
tunica, is composed of fi brous connective tissue richly supplied by small blood and 
lymphatic vessels. Although being a mucosal surface, the vaginal mucosa does not 
FIGURE 2 Detail of human vaginal mucosa of fertile adult (H & E, . 40). Image shows 
stratifi ed squamous epithelium (E), which is nonkeratinized, and lamina propria (LP), richly 
supplied by small blood vessels and deprived of glands (Courtesy of Manuel Dias.) 
THE HUMAN VAGINA 813

814 VAGINAL DRUG DELIVERY 
have mucus - secreting glands (Figure 2 ). The muscular layer has a large number of 
interlacing fi bers, making it possible to distinguish an inner circular layer and an 
outer longitudinal layer. These muscle fi bers allow substantial elongation during 
childbirth, increasing approximately fi ve - fold. The tunica adventitia consists of connective 
tissue containing a large plexus of blood vessels, lymphatic vessels, and 
nerves [9] . 
The characteristics of the vaginal mucosa depend on sexual hormones undergoing 
changes throughout the menstrual cycle. During the proliferative phase, estrogens 
increase vaginal blood fl ow and the integrity of the mucosa, inducing the 
proliferation of the vaginal epithelium, peaking at approximately midcycle. Nonetheless, 
this change in epithelial thickness is small and probably clinically insignifi - 
cant [10, 11] . In the secretory phase progesterone opposes those actions, although 
without signifi cantly infl uencing the epithelium thickness [12] . 
5.12.2.3 Physiology 
Functions of the vagina include receiving the erect penis and semen during coitus 
and ejaculation and serving as a passageway for fetus and menses to the outside of 
the body [4] . 
The vaginal milieu is typically acidic in healthy women during their fertile years, 
playing an important role in preventing the proliferation of pathogenic microorganisms. 
Vaginal pH is around 3.5 – 4.5, changing during the menstrual cycle being lower 
in the middle cycle and higher around menses [13] . Also, vaginal pH may be altered 
by several conditions, including the presence of semen (pH 7.2 – 8.0) or bacterial 
infections. In this last case vaginal pH usually increases to values of 5.0 – 6.5 [14] . 
These and other variations of vaginal pH may alter the effi cacy of administered 
drugs through quite a few mechanisms, including variable release from the delivery 
system, alteration in drug absorption and/or metabolism, and modulation of drug 
activity in the cervicovaginal milieu [15] . Lactobacillus species are present in the 
vagina of healthy women, being responsible for its acidity. These bacteria use glycogen, 
which is synthesized by epithelial cells under estrogen ’ s infl uence in order to 
produce lactic acid, thus reducing vaginal pH [16] . Besides their contribution to 
vaginal pH, lactobacilli also have the ability to regulate the proliferation of microorganisms, 
namely pathogenic species, by other mechanisms: adherence to the 
mucus, forming a barrier which prevents colonization by pathogens or competition 
for the receptors of the epithelial cells, and the production of antimicrobial compounds 
such as hydrogen peroxide, lactic acid, bacteriocin - like substances, and possibly 
biosurfactants [17] . Although lactobacilli are the main organisms responsible 
for vaginal acidity, recent research suggests that other acid - producing microorganisms, 
such as Atopobium sp., Megasphaera sp., and Leptotrichia sp., may also contribute 
to the acidity of the vaginal tract [18] . The composition of vaginal fl ora is 
complex, being infl uenced by hormonal changes, although lactobacilli levels of colonization 
seem to remain relatively constant during the menstrual cycle [19] . Also, 
other factors such as glycogen content, pH, sexual intercourse, medication, and 
immunity status are known to infl uence the vaginal ecosystem [20] . 
Although deprived of secreting glandules, the vaginal mucosa is covered with a 
watery acidic fl uid. This fl uid includes contributions from vaginal transudation, 
Bartholin and Skene glands, exfoliated epithelial cells, residual urine, and fl uids from 

the upper reproductive tract such as cervical mucus and endometrial and tube fl uids. 
Vaginal fl uid consists of 90 – 95% water, inorganic and organic salts, urea, carbohydrates, 
glycerol, mucins, fatty acids, albumins, immunoglobulins, enzymes, leukocytes, 
and epithelial debris. Although quite variable, normal daily production of vaginal 
fl uid is estimated at around 6 mL, increasing at midcycle and decreasing around the 
menstruation period [21] . In fact, the menstrual cycle plays an important role in 
vaginal fl uid characteristics, particularly on pH values, rheological properties, color 
(from milky white to transparent), and antimicrobial activity. Estrogens induce the 
production of vaginal fl uid, leading to the lubrication of the mucosa. Decline of 
serum estrogen levels, as during prepubertal or postmenopausal periods, results in 
a reduction of vaginal moisture [22, 23] . This variability may infl uence drug release, 
dissolution, absorption, and removal and thus its activity. Also, vaginal fl uid is selectively 
antimicrobial, lactic acid and to a lesser extent antimicrobial peptides and 
proteins (calprotectin, lysozyme, histones, and others) being partially responsible by 
the resistance of the normal vagina to colonization by exogenous microorganisms 
[24] . 
Cervical mucus is an important component of the vaginal fl uid, although it presents 
substantial differences when compared with whole vaginal fl uid, mainly in pH 
value (approximately 7.0). It is produced in the cervix, leaking down into the vagina, 
particularly during the three - to fi ve - day interval that precedes ovulation. Its properties 
are also infl uenced by the menstrual cycle, particularly pH (range of 5.4 – 8.0) 
and antimicrobial activity [25] . Also, estrogens stimulate the secretion of abundant 
and fl uid cervical mucus, while progesterone induces the formation of thick cervical 
mucus. These changes in viscosity infl uence the capacity of sperm and other substances, 
such as drugs, to pass the cervix and migrate to the uterus [22, 23] . 
Interaction between vaginal fl uids and drug delivery systems is an important 
aspect that has to be managed during drug design, as it may infl uence the fl ow, 
retention, drug delivery kinetics, and bioactivity of vaginal formulations [21] . Since 
fl uids present in the vaginal environment are diffi cult to obtain, simulated fl uids 
have been developed in order to emulate their physical and chemical properties. 
Recently, a vaginal fl uid simulant was proposed by Owen and Katz [21] , whether 
Burruano et al. [26] developed a synthetic cervical mucus formulation. These fl uids 
have been successfully used to evaluate vaginal formulations in vitro, being able to 
mimic with considerable accuracy the physiological fl uids. Also, the standardization 
of a single composition provides the possibility of comparing results obtained by 
different investigators. Table 1 presents the main features of these two simulants. 
Enzymes present in the vagina may infl uence drug delivery, as they can degrade 
and infl uence the permeability of the administered drugs. Although the enzymatic 
activity in the vagina is not as high as in other drug delivery sites, several enzymes 
can be found in the vaginal fl uid and in different vaginal epithelium cells, namely 
succinic and lactic dehydrogenase, acid and alkaline phosphatases, . - glucuronidase, 
phosphoamidase, lactate dehydrogenase, aminopeptidase, and esterases [27, 28] . 
This enzymatic activity can limit drug bioavailability and decrease the stability of 
prolonged delivery formulations. 
Understanding the immune mechanisms responsible for the defense of the female 
genital tract is of extreme importance concerning the development of vaccines that 
are effective against pathogens. The female genital tract has several mechanisms of 
defense against infectious agents, which appear complementary, additive, and even 
THE HUMAN VAGINA 815

816 VAGINAL DRUG DELIVERY 
synergistic. The immune response can be classifi ed in three levels: nonimmune, 
preimmune, and acquired or specifi c (Table 2 ) [29] . Nonimmune response is very 
effective in limiting the infectious inoculum, while preimmune mechanisms hold up 
the infection long enough so that the immune response can be activated. These 
defense strategies are largely infl uenced by pathogenic agents presented in the 
vagina and the hormonal milieu [29, 30] . 
The genital tract is part of the common mucosal immune system, which comprises 
all mucosal tissues of the body, sharing similarities and common immunization 
mechanisms, being able to disseminate acquired immunity between them. Nonetheless, 
it has become clear that the genital tract presents several unique features that 
differentiate it from other mucosal sites [31] . One of the most important is the ability 
to only induce local immune responses. In fact, most of the antibodies are produced 
locally at the mucosa, while those derived from the circulation represent only a small 
fraction. Also, contrasting with other mucosal tissues, vaginal secretions contain 
more immunoglobulin (Ig) G than secretory IgA [30, 32] . Hence, immunity of the 
genital tract is conferred by local production of IgA and, to a less extent, by transudation 
of serum IgG, although cellular immune response should also be considered 
[33] . Local immune cell population in the vaginal mucosa includes Langerhans 
cells, macrophages, T cells, and neutrophils. Systemic immune response is also impor- 
TABLE 1 Main Features of Vaginal Fluid Simulant a and Synthetic Cervical Mucus b 
Vaginal Fluid Simulant Synthetic Cervical Mucus 
Composition NaCl (3.51 g), KOH (1.40 g), Ca(OH) 2 
(0.222 g), bovine serum albumin 
(0.018 g), lactic acid (2.00 g), acetic 
acid (1.00 g), glycerol (0.16 g), urea 
(0.4 g), glucose (5.0 g), HCl (to 
adjust pH), and water (e.q. 1 L) 
Guar gum (1.00%), dried porcine 
gastric mucine (type III) 
(0.50%), imidurea, (0.30%), 
methylparaben (0.15%), 
propylparaben (0.02%), dibasic 
potassium phosphate (0.26%), 
monobasic potassium 
phosphate (1.57%), and water 
(96.20%) 
pH 4.2 7.4 
a From ref. 21 . 
b From ref. 26 . 
TABLE 2 Immune Response of Female Genital Tract 
Nonimmune Passive 
Synthesis of protective mucus 
pH 
Epithelial barrier 
Active 
Infl ammatory reaction 
Secretion of humoral soluble factors 
Preimmune Humoral response, cellular response 
Immune Humoral response, cellular response 
Source : From ref. 29 . 

tant in the reinforcement of the acquired mucosal immunity or to take over when 
this one has faded, although they are independent from one another [29, 30] . 
Sexual hormones, particularly estrogens, also infl uence the regulation of local 
immunity, as IgA levels in genital secretions, antigen presentation by vaginal cells, 
and lymphocyte proliferation vary throughout the menstrual cycle [34, 35] . Estrogens 
can decrease the concentrations of immunoglobulins, while progestogens can 
increase their levels [36] . 
Alterations in vaginal physiology before, during, and after sexual intercourse are 
important factors in the performance of those formulations intended to be used in 
this period. During sexual arousal, genital vasocongestion occurs, leading the clitoris 
and the labia minora to become enlarged with blood and the vagina to increase in 
length and diameter as a result of relaxation of the smooth muscular wall. The 
vaginal canal is lubricated by secretions from the uterus, Bartholin and Skene glands, 
and a fl uid that transudates from the subepithelial vascular tissues, being passively 
transported through the intercellular spaces. Engorgement of the vaginal wall raises 
the pressure inside the capillaries, thus increasing the transudation of plasma through 
the vaginal epithelium [6] . The resulting vaginal fl uid is increased in quantity, less 
acidic, and more diluted, allowing penile penetration and thus preventing the male 
and female genitalia from becoming irritated [3, 4] . 
5.12.2.4 Childhood, Pregnancy, and Menopause 
The vagina undergoes several lifetime changes that may infl uence vaginal formulation 
performance. These changes are particularly important when developing vaginal 
products that are intended to be used in specifi c situations, such as labor inducers 
in pregnant women or hormonal supplements in postmenopausal women. 
The vagina of the newborn exhibits the infl uence of residual maternal estrogens, 
presenting a stratifi ed squamous epithelium rich in glycogen and becoming colonized 
with lactic acid – producing microorganisms shortly after birth. By the fourth 
postnatal week these estrogenic effects disappear, and the vaginal epithelium loses 
its stratifi cation and glycogen content, becoming much thinner and exhibiting alkaline 
or neutral pH because of acid - producing microorganism depletion. These characteristics 
remain throughout childhood, until puberty. By this time, the vagina 
experiences changes due to adrenal and gonadal maturation. This organ increases 
in size, vaginal fornices develop, cervicovaginal secretions start being produced, and 
vaginal milieu becomes acidic. Also, vaginal epithelium thickens and intracellular 
glycogen production increases [37] . 
When the fertile woman becomes pregnant, the connective tissue of the vulva, 
vagina, and perineum relaxes, and the muscle fi bers of the vaginal wall increases in 
size. These alterations prepare the vagina for childbirth. During delivery, perineal 
and vaginal musculature relaxes and the vaginal rugae fl atten, allowing full expansion 
of the vaginal tract, accommodating the passage of the newborn. Normal morphology 
and dimensions of the vagina are recovered after 6 – 12 weeks [37] . The 
vaginal blood supply is substantially increased during pregnancy, which can enhance 
systemic absorption of drugs. High levels of estrogens during pregnancy lead to 
thickening of vaginal epithelium and stimulation of glycogen production. This 
increased glycogen content promotes lactobacilli growth, consequently decreasing 
vaginal pH by the enhancement of lactic acid synthesis. 
THE HUMAN VAGINA 817

818 VAGINAL DRUG DELIVERY 
Menopausal women experience a decline in estrogens, which leads to several 
changes in the genital organs, namely in the vagina. Such changes include atrophy 
of the labia majora and shortening and loss of elasticity of the vaginal barrel. The 
number of epithelial cell layers decreases (from 8 – 10 in premenopausal to 3 – 4 in 
postmenopausal), leading to alterations in the barrier capacity and potential increase 
of mucosal damage and pain and burning sensation during sexual intercourse. 
Vaginal fl uids decrease approximately 50% because of the Bartholin glands atrophy 
and a decrease in the number and maturity of vaginal cells. A decrease in the colonization 
by lactobacilli species is observed in menopausal women as a result of the 
reduction of vaginal glycogen levels, leading to a low production of lactic acid and 
consequently to increased vaginal pH. In postmenopausal women without estrogen 
treatment the vaginal pH is estimated to be 5.5 – 6.8 or even higher [37, 38] . 
5.12.3 GENERAL FEATURES OF VAGINAL DRUG DELIVERY 
Vaginal drug delivery is mostly used in gender - specifi c conditions, although it can 
be a viable alternative for drugs usually administered by other routes. Also, traditionally 
problematic drugs from a delivery point of view (e.g., peptides) may fi nd in 
the vaginal route an interesting and promising way for nonparental administration. 
Limitations and potentialities of vaginal drug administration are intimately connected 
to this route ’ s idiosyncrasy. Hence, acquaintance of particular features of 
vaginal drug delivery is required. 
5.12.3.1 Advantages and Disadvantages of Vaginal Drug Delivery 
The administration of drugs through the vagina, and eventually their absorption, is 
a function for which this organ is not physiologically conceived. Nonetheless, the 
vaginal route of administration presents some advantages. Substances absorbed 
through the vaginal mucosa bypass the liver before entering systemic circulation, 
avoiding hepatic fi rst - pass metabolism. Thus, drugs that undergo extensive hepatic 
fi rst - pass metabolism can benefi t from vaginal administration, usually requiring less 
amount of drug to achieve the same biological effects. Steroids used in hormone 
replacement therapy or contraception are a good example of molecules that are 
largely metabolized in the liver, with approximately 95% of orally administered 
estrogens undergoing hepatic metabolism. Also, these molecules are able to damage 
the liver when administered by the oral route, an event that can be minimized with 
vaginal administration. Gastrointestinal side effects are common for many oral 
administered drugs; the vaginal route may be an alternative to their administration, 
with the benefi t of increased patient compliance. Additionally, vaginal enzymatic 
activity is lower when compared with gastrointestinal activity, lacking even some 
important enzymes enrolled in drug metabolism, such as trypsin and chymotrypsin. 
The vaginal route offers women the possibility of easy self - insertion and removal 
of drug delivery systems as well as avoidance of the pain, tissue damage, and eventual 
infection often associated with parenteral routes. Ocular and buccal administration 
sites frequently become irritated after prolonged contact with drug delivery 
systems; conversely, the vagina presents less sensitivity, allowing the presence of 
drug formulations for long periods of time. Although absorption of substances is 

not a function of the vagina, features such as its relatively large surface area and 
rich blood supply contribute to this organ ’ s high permeability to several drugs, 
allowing higher bioavailability of some active substances when compared to other 
routes [39 – 41] . On the other hand, the vaginal drug delivery route has some limitations. 
Gender specifi city is the most important, as it restricts its use to females only. 
Others, such as misperceptions and cultural issues about genital manipulation and 
insertion of objects in the vagina, personal hygiene, infl uence with sexual intercourse, 
and variability in drug absorption related with menstrual cycle, menopause, 
and pregnancy, can also limit vaginal drug delivery route usage [41, 42] . 
5.12.3.2 Permeability and Drug Absorption 
Although some substances are not desired to be absorbed, such as those targeted 
for local action, permeation of drugs through the vaginal wall into the bloodstream 
must occur if one seeks to obtain a systemic effect. Although vaginal administration 
of drugs has been performed for a long time, the capability of systemic drug absorption 
through this organ was not clarifi ed until the early twentieth century by investigators 
such as Macht or Robinson [43, 44] . Both conducted independent experiments 
with substances such as potassium iodide, morphine, atropine, sodium salicylate, 
quinine, sucrose, and phenol red, which evidenced the permeability of this mucosal 
tissue, thus opening the possibility of systemic drug delivery through the vagina. 
Nonetheless, the currently acknowledged variability of vaginal drug absorption with 
the hormonal status of women, which can limit the potential for systemic drug 
delivery, was documented in the 1940s by authors such as Rock et al. These researchers 
reported that although the vaginal administration of drugs, namely penicillin, 
could provide therapeutic blood levels, the extension of their absorption was highly 
variable [45] . 
Recently, several in vitro experiments substantiated the potential of the human 
vaginal mucosa as a good administration route relating to the degree of permeation 
when compared with other mucosal surfaces. In fact, the vagina can be more permeable 
to some commonly used model substances, such as water, 17 . - estradiol (Figure 
3 ), arecoline, arecaidine, and vasopressin, than colonic or small intestinal mucosa, 
or at least as permeable as when compared to human buccal mucosa [46, 47] . 
In general, systemic drug absorption requires three steps: drug release from the 
delivery system, drug dissolution in the vaginal fl uid, and permeation of the vaginal 
mucosa. Knowledge of the permeability characteristics of the vaginal mucosa is an 
important step when developing a pharmaceutical product, with both the physicochemical 
properties of chemical substances (e.g., chemical nature, degree of ionization, 
molecular weight and size, conformation, and oil/water partition coeffi cient) 
and the biophysicochemical nature of the tissue infl uencing absorption. The epithelial 
layer of the vaginal mucosa presents itself as the main permeability barrier for 
drug absorption. As the epithelium is hormonally dependent, its permeability also 
changes, usually decreasing with higher estrogen levels because of the induced 
membrane thickening. However, contradictory fi ndings of enhanced vaginal absorption 
in postmenopausal women treated with estrogen have been reported [48] . 
These results can be explained by the increment of vaginal mucosa blood fl ow that 
is also induced by estrogen. The transport mechanism of most vaginal absorbed 
substances is simple diffusion. Lipophilic substances are absorbed through the 
GENERAL FEATURES OF VAGINAL DRUG DELIVERY 819

820 VAGINAL DRUG DELIVERY 
intracellular (or transcellular) pathway, whereas hydrophilic substances are absorbed 
through the intercellular (or paracellular) pathway or across aqueous pores present 
in the vaginal mucosa [49] . Also, receptor - mediated transport mechanisms can be 
involved in the absorption of some substances. 
The magnitude of the fl ux rate across the vaginal mucosa is mainly related to the 
molecular size and hydrophobicity of the permeating substances. In general, compounds 
with molecular weight over 300 Da have decreased fl ux rates, while hydrophobic 
properties usually increase permeation [50 – 52] . The infl uence of the penetrant 
hydrophobicity/hidrophilicity in the rate and extent of absorption through the 
vaginal mucosa was demonstrated by Corbo et al. [53, 54] . Experiments performed 
in rabbits showed that hydrophilicity infl uenced mucosal permeability of drugs such 
as progesterone, with increasing hidrophilicity leading to decrease in rate and extent 
of vaginal absorption. Nonetheless, one should keep in mind that a minimum degree 
of aqueous solubility is always required in order to ensure that the drug dissolves 
in the vaginal fl uid. Also, pH, viscosity, and volume of this fl uid change throughout 
the menstrual cycle, potentially infl uencing the extension of drug dissolution: Different 
pH values infl uence the drug degree of ionization and thus its solubility; 
increased viscosity of the vaginal fl uid may enhance drug retention, although it may 
also present a barrier to drug absorption; and higher volumes of fl uid benefi t drug 
dissolution but also increase its clearance from the vagina. 
However, permeability - enhancing strategies may be required, as many substances 
cannot permeate the vaginal mucosa in signifi cant pharmacological levels. An interesting 
and helpful option is the use of several permeation enhancers which can 
increase drug absorption by interacting with epithelial tight junctions, providing a 
new intercellular penetration pathway [55] . Some of these substances, such as citric 
acid, benzalkonium chloride, laureth - 9, lysophosphatidylglycerol, sodium taurodihydrofusidate, 
lysophosphatidylcholine, palmitoylcarnitine chloride, lysophosphatidylglycerol, 
and sodium glycodihydrofusidate, have been successfully tested [56 – 60] . 
FIGURE 3 Overall mean fl ux values of 17 . - estradiol across human vaginal, colonic, and 
small intestinal mucosa, as determined by fl ow - through diffusion cells. Flux values ( J ) were 
calculated as J = Q /( At ), where Q is quantity of 17 . - estradiol crossing mucosa (in dpm), A 
is mucosa area exposed (in cm 2 ), and t is time of exposure (in min). SEM: standard error of 
the mean. (Reprinted with permission from P. van der Bijl and A. D. van Eyk, International 
Journal of Pharmaceutics , 261, 147 – 152, 2003. Copyright 2003 by Elsevier.) 
Vagina Colon Intestine 500 
400 
300 
200 
100 
0 
0 
5 10 15 20 25 
Time (h) Bar represents SEM

Although this strategy may enhance permeability, it presents some disadvantages, 
particularly the possibility of mucosal damage. These unwanted effects are variable 
and not always observed; however, they may often be severe [61] . Also, some drugs 
achieve poor systemic levels after being delivered by the vaginal route, not because 
of poor permeation through the mucosa, but due to their fast inactivation by local 
enzymes, particularly when therapeutic peptides and proteins are considered. 
In these cases, enzymatic inhibitors may be a helpful solution. Vaginal peptidase 
inhibitors, such as ethylene diamine tetraacetic acid (EDTA), thimerosal, amastatin, 
bestatin, leuptin, and pepstatin A, were shown to be useful, promoting peptide 
absorption in rats, as they prevent drug degradation [60, 62] . At this point it is also 
important to notice that some substances can reduce the permeability of the vaginal 
mucosa [63] , this possibility always being important when designing a drug delivery 
system. In addition, other approaches, such as the use of mucoadhesive polymers, 
in situ gelling formulations, or solubility enhancers, have been shown to be useful 
in improving vaginal permeability of several drugs [40] . 
5.12.3.3 First - Uterine - Pass Effect 
The fi rst - uterine - pass effect can be defi ned as a preferential transfer of vaginally 
administered drugs to the uterus. This effect is due to a countercurrent mode of 
exchange, with an upward vagina - to - uterus transport of substances absorbed in 
vaginal and lymphatic vessels and diffusing to nearby arteries [64] . Evidences of 
higher than expected uterine concentration after vaginal administration of drugs, 
namely progesterone, terbutaline, or danazol, led to the postulation and verifi cation 
of this hypothesis [65, 66] . This effect can be of the utmost importance when the 
uterus is the desired locale for a drug to exert its effects, opening new therapeutic 
options for uterus - related conditions. 
Further investigations showed that the placement of a formulation in different 
areas of the vagina dramatically infl uences the observation of the fi rst - uterine pass - 
effect. Experimental fi ndings suggest that this preferential transfer to the uterus is 
only observed when absorption occurs in the outer one - third of the vagina [67] . 
Thus, drugs intended to exert their effects in the uterus should not be inserted 
deeply in the vagina, as it is often recommended, instead they should be placed near 
the vaginal entrance. 
5.12.4 VAGINAL DRUG DELIVERY SYSTEMS 
5.12.4.1 Overview 
A wide range of drug delivery systems have been used, although many of them are 
not specifi cally designed for intravaginal administration. Traditionally used vaginal 
drug delivery systems include solutions, ointments, creams, vaginal suppositories, 
and tablets. Recently, others, such as vaginal rings or vaginal fi lms, have been developed. 
Also, several strategies and improvements have been tested in order to overcome 
natural limitations of drug delivery through this route, particularly low 
retention, limited absorption, and cyclic variations. 
Most of the currently available vaginal formulations, particularly those that have 
been marketed for a longer period of time, have serious limitations such as poor 
VAGINAL DRUG DELIVERY SYSTEMS 821

822 VAGINAL DRUG DELIVERY 
spreadability, messiness, and small capacity of retention in the vagina. Nonetheless, 
recent advances allowed circumventing some of the major diffi culties that hold back 
the use of this route of drug delivery as a serious alternative to the most traditional 
ones, with the consequent increase of commercially available drug delivery systems 
[39] . Drug release of most traditional formulations is rapid, needing frequent administrations 
to sustain therapeutic drug concentrations. Thus, in recent years, sustained 
release has been a new approach to deliver several active substances through the 
vaginal route. Also, vaginal drug delivery systems should ensure either an adequate 
penetration of the drug within the mucosa, in order to enhance the local effects and 
reduce systemic absorption, or an ideal permeation of the active substances into the 
bloodstream in order to assure an effective systemic response. 
Before formulators choose a delivery system for a selected drug, several issues 
should be taken into consideration: physicochemical properties of the active substance, 
intended effect of the active substance, required drug release profi le, excipients 
to be used and their compatibility with the active substance and vaginal mucosa, 
women ’ s preferences, and economical implications. 
5.12.4.2 Excipients 
When vaginal drug delivery is considered, formulators must select a number of suitable 
excipients in order to design a drug delivery system able to ensure the therapeutic 
success of the active substance(s). In fact, it is known that excipients used in 
vaginal formulations can infl uence the pharmacological performance of active substances, 
being able to improve or diminish their activity [68, 69] . The decision of 
which excipients to use depends to a great extent on the fi nal dosage form and 
desired characteristics of the drug delivery system. Some excipients can infl uence 
drug delivery system performance by changing some properties, such as viscosity, 
mucoadhesion, and distribution [70] . Although these variations do not interfere 
directly with the pharmacological effect of the active substances, their availability 
and thus the formulation clinical outcome can be compromised. Thus, excipient 
selection must be performed with utmost caution, taking into consideration the 
quality, safety, and functionality aspects of these materials. Indeed, Garg et al. 
recently compiled a list of excipients that are currently approved or have already 
been investigated for vaginal administration [71] . 
Although by defi nition excipients are deprived of pharmacological effects, some 
have showed that this is not always true. For instance, chitosan, an excipient that 
has attracted a lot of interest in the formulation of vaginal drug delivery systems, 
exhibits antimycotic effects, particularly against the common vaginal pathogen 
Candida albicans [72] . Also, other polymers commonly used in tablets and capsules, 
such as cellulose acetate phthalate, have been investigated in the formulation of 
vaginal microbicides, due to their antiviral effects against HIV [73] . 
Some commonly used excipients can interact with vaginal and cervical fl uids, 
altering their properties. These interactions should be taken into consideration when 
designing a drug delivery system, as they can infl uence in vivo performance. For 
example, small amounts of nonionic (e.g., polyethylene glycol) and cationic (e.g., 
polyvinylpyridine) polymers are able to modify the gel structure of the cervical 
mucus, altering its barrier properties, while like - charged molecules (e.g., polyacrylic 
acid) interact little with this biological fl uid. This approach has been taken into 

account, particularly as a new prevention strategy for pathogens that infect via the 
mucosa, as a new treatment option for diseases that affect the mucous layer itself 
or even as a strategy for systemic drug delivery routes [74, 75] . 
5.12.4.3 Solid Systems 
Solid systems commonly administered by the vaginal route include tablets, capsules, 
and vaginal suppositories. 
Tablets are frequently used as vaginal drug delivery systems, being inexpensive 
and easy to manufacture. They are also easily administered in the vagina, allowing 
a “ clean ” insertion that contrasts with the typical messiness of semisolid drug delivery 
systems. Although very similar to oral tablets, these systems have some particularities, 
such as being round or oval shaped and devoid of sharp edges, in order to 
avoid damage of mucosal tissue. These drug delivery systems are usually designed 
to rapidly release their active substances after being placed in the vagina. In fact, 
disintegration or dissolution problems, mainly due to the scarce amount of vaginal 
fl uid, are important issues to be managed by formulators. This rapid release and 
solubility enhancement of the active substances can be important because of the 
rapid vaginal wash - off and low in situ retention. Increased and faster release of drug 
content has been achieved using effervescent tablets [76] or including specifi c excipients 
that can enhance its disintegration in vaginal fl uids [77] . For instance, Karasulu 
et al. proposed an effervescent tablet made of a mixture of mucoadhesive microcapsules 
loaded with ketoconazole and effervescent granules [76] . This combination 
showed ability to improve retention with rapid onset of action. Additionally, other 
strategies have been used, such as inclusion complexes of poorly soluble drugs with 
cyclodextrins, in order to improve drug solubility, allowing a rapid onset of the 
pharmacological effect. 
Although fast release of the active substances is a frequent goal, controlled - 
release tablets can be used in order to enhance their effi cacy, because of their prolonged 
release, and prevent the irritation of the vaginal mucosa that may be caused 
by some drugs [78] . Nonoxynol - 9, a commonly used microbicide and spermicide, 
known for its irritability when administered in the vagina, is a good example of a 
drug that can benefi t from controlled release. Formulation of double - layer tablets 
(fast - release outer layer and slow - release core) obtained from coprecipitates of 
nonoxynol - 9 with polyvinylpyrrolidone, can provide extended drug release, allowing 
a more prolonged spermicidal effect while reducing its irritating effect [79] . Also, 
controlled release prevents peaks in serum concentration of absorbable drugs, limiting 
possible systemic effects [80] . 
Vaginal tablets containing lactobacilli have been used in order to restore the 
normal vaginal fl ora. Formulation of these delivery systems requires specifi c proceedings 
in order to provide viability of lactobacilli and stability of the fi nal product. 
Freeze drying of bacterial suspensions has been tested to obtain lyophilized powders 
for tablet production [81] . These powders were shown to be processable and tablet 
production was easy and reproducible. Also, the use of double - layer tablets (fast - 
release layer and slow - release layer) seems to be an interesting approach to lactobacilli 
administration. 
It is common to use tablets designed for the oral route in order to deliver drugs 
through the vagina. Nonetheless, issues such as delivery system retention and 
VAGINAL DRUG DELIVERY SYSTEMS 823

824 VAGINAL DRUG DELIVERY 
distribution and drug release can infl uence the fi nal performance of the formulation, 
being preferable to use specifi cally vaginal designed drug delivery systems, or at 
least study the pharmacokinetics of oral tablets after vaginal administration [82] . 
Capsules, particularly soft capsules, have been used as vaginal drug delivery 
systems, but with modest popularity. These systems are relatively stable, particularly 
when compared with semisolid formulations or vaginal suppositories, being an 
adequate way to deliver liquid drugs within a solid dosage form. 
Vaginal suppositories, also referred as ovules or pessaries, are ovoid - shaped, solid 
(but generally malleable) dosage forms specifi cally designed for vaginal administration. 
These systems usually weigh 2 – 3 g, although formulations with up to 16 g have 
been used in the past [83] . Vaginal suppositories have a long history of use as vaginal 
drug delivery systems, mainly in the management of local conditions. Major advantages 
are their reduced price and ease of manufacture. However, they present some 
inconveniences, such as messiness, low retention in the vagina, and poor stability, 
the last feature due to their temperature and moisture sensibility. 
Vaginal suppositories are very close to rectal suppositories in terms of excipient 
nature and manufacturing process. Thus, they are usually prepared by fusion of the 
excipient(s) (referred as “ base ” ) and incorporation of the active substance(s), this 
mixture being subsequently poured into molds and allowed to solidify. Other 
methods, such as by compression, can also be used. Several substances have been 
utilized as bases for the formulation of vaginal suppositories: gelatin and glycerin, 
cocoa butter, semisynthetic glycerides, and polyethylene glycol, among others [83] . 
Composition of vaginal suppositories is importantly related to their melting or dissolution, 
thus infl uencing drug release profi le. Generally, it can be stated that drug 
release rate increases as the melting temperature of a suppository decreases or as 
its dissolution time in vaginal fl uids increases. Also, affi nity of the drug for the base 
infl uences its release from vaginal suppositories: Greater release of drug is expected 
when there is less affi nity between the active substance(s) and the base [84] . The 
melting temperature and melting process of vaginal suppositories can be characterized 
by several techniques, such as differential scanning calorimetry and viscosity 
and dilatometry methods, among others [85] . Specifi c pharmaceutical characterization 
of vaginal suppositories includes the determination of disintegration time and 
breaking hardness. Also, other standard quality control tests include appearance 
description, surface texture evaluation, pH determination, uniformity of content, 
and microbial limit testing [86, 87] . 
Recently, sustained - release vaginal suppositories have been developed in order 
to attain drug delivery systems with improved performance. Sustained release can 
reduce the number of administrations, thus improving patient compliance. A base 
composition consisting of a polymeric gum (carboxymethylcellulose and xanthan 
gum), a dispersing agent (colloidal silicone dioxide), and polyethylene glycol, 
referred as long acting, sustained release of spermicide (LASRS), has recently been 
studied by Zaneveld et al. in order to deliver contraceptives and microbicides [88] . 
Results showed that a LASRS base is able to spread quickly and evenly over the 
mucosa, being retained in place for prolonged periods of time and allowing long - 
lasting effi cacy for several active drugs. Preliminary human trials have confi rmed 
these results [89] . In another study, Mandal developed hydrophilic vaginal suppositories 
comprising mixtures of miconazole cross - linked with poly(vinyl alcohol) by 

freeze thawing and different polyethylene glycols that were able to sustain release 
this antifungal drug for up to 108 h [90] . 
5.12.4.4 Semisolid Systems 
Semisolid systems present several advantages over other drug delivery systems: 
They are easy to use and generally inexpensive and have good acceptability. Among 
their disadvantages, leakage has been one of the most disturbing, mainly because 
many conventional formulations are not mucoadhesive. The simplest way of dealing 
with this problem has been the recommendation for night administration, as the 
supine position diminishes leakage. Also, messiness and discomfort upon application 
and diffi culties in dispensing an accurate dose are important limitations. 
Once widely used, ointments have been largely substituted by creams and gels. 
Nonetheless, some of these drug delivery systems may still be encountered, particularly 
as hydrophilic bases. 
Creams have been used for quite some time as vaginal drug delivery systems, 
particularly for the administration of sexual hormones and antimicrobials. The main 
advantage of creams over other semisolid systems is their ability to easily dissolve 
both hydrophobic and hydrophilic drugs. As most conventional creams do not 
possess bioadhesive properties, incorporation of bioadhesive polymers is an effective 
approach to improve their retention in the vagina. Recently, a new approach 
to vaginal drug delivery was developed using Site Release (SR) technology (KV 
Pharmaceutical, St. Louis, MO). This technology is based on bioadhesive controlled - 
release water - in - oil emulsions, being formulated as a vaginal cream (SR cream). The 
outer oily phase repels moisture (thereby resisting dilution) and retains the dispersed 
water phase containing the drug (allowing controlled release) [91] . The SR 
cream allows minimizing leakage and enhancing clinical outcome, requiring less 
total drug exposure per course of therapy. Site Release technology is currently available 
in the United States in two commercial products: one containing butoconazole 
nitrate 2% (Gynazole - 1, Ther - Rx Co., St. Louis, MO) and the other containing 
clindamycin phosphate 2% (Clindesse, Ther - Rx Co.). Clinical fi ndings demonstrated 
that a single application of Gynazole - 1 makes it possible to achieve more rapid relief 
of vaginal candidiasis symptoms than standard oral therapy with fl uconazole [92] . 
Similarly, Clindesse was show to be able to achieve prolonged local effective concentrations 
while presenting lower systemic bioavailability and thus less systemic 
adverse effects when compared with conventional formulations in the treatment of 
bacterial vaginitis [93] . Also, other drugs have been studied in order to further evaluate 
the potential of this versatile technology [94] . 
Since the pioneer work by Wichterle and Lim in the 1960s [95] , gels have evolved 
greatly from simple formulations to advanced drug delivery systems. These systems 
were soon demonstrated to be good candidates to deliver drugs in the vagina, 
particularly because of their high bioavailability (mainly because of mucoadhesive 
properties), biocompatibility, spreadability, ease of usage, and economical savings 
[96] . Gels are extremely versatile, being used to deliver most of the currently used 
drugs through the vaginal route. 
Recent advances in gel and polymer technology boosted research, opening new 
possibilities for vaginal drug delivery [97] . Indeed, the development of new and 
VAGINAL DRUG DELIVERY SYSTEMS 825

826 VAGINAL DRUG DELIVERY 
improved gelling agents, particularly concerning to their mucoadhesive properties, 
has been of great importance. For example, polycarbophil (Noveon AA - 1, Noveon, 
Cleveland, OH), a mucoadhesive polyacrilic acid polymer, has been widely used as 
a gelling agent in vaginal gel formulations. This polymer is acidic in nature, which 
can be useful in reducing the elevated pH associated with bacterial vaginosis [98] . 
Additionally, acidic polycarbophil gels may be used in the treatment of dry vagina 
and menopause - related stress incontinence [99] . 
Also, gel microemulsions have been recently reported as safe and devoid of 
mucosal toxicity drug delivery systems, presenting intrinsic spermicide activity and 
the possibility of improving vaginal bioavailability of poorly soluble antimicrobial 
agents [100] . 
Although most currently available vaginal gels rapidly release their active 
substance(s), they can also be formulated to achieve modifi ed drug release profi les 
[101] . It is not clear how controlled release is achieved, but the analysis of most 
formulations that claim to possess this feature suggests a combination of dissolution 
and diffusion control [102] . Gels are also known to be promising drug delivery 
systems in protein and peptide administration through the vagina, proving to be 
adequate to accommodate and stabilize sensible molecules such as leuprolide 
[103] . 
5.12.4.5 Liquid Systems 
Vaginal douching with liquids containing antimicrobial drugs such as povidone - 
iodine has been a common practice among women, with the intention of improving 
personal hygiene and treat vaginitis [104] . These liquids are almost immediately 
removed from the vagina after administration, thus being inadequate for controlled 
release. Although vaginal washing is frequently performed by women all over the 
world, its practice is discouraged, as it is associated with increased risk of acquiring 
HIV, particularly when soap or other substances rather than water are used [105, 
106] . Also, bacterial vaginitis and other adverse reproductive health effects are possible 
when vaginal douching is a frequent practice [107, 108] . 
In addition, several solutions are utilized by gynecologists in their offi ce practice. 
For example, glacial acetic acid solutions (3 – 5%) are used to identify cervical dysplasia 
during colposcopy, and Lugol solution is employed to perform Schiller ’ s test 
(diagnosis of cervix cancer). 
5.12.4.6 Vaginal Rings 
Vaginal rings are doughnut - shaped drug delivery systems designed to provide controlled 
release of drugs. Developed systems are made of fl exible, inert, and nonirritating 
polymeric materials, presenting different dimensions, usually 54 – 58 mm in 
diameter and 4 – 9.5 mm in cross - sectional diameter [109, 110] . Vaginal rings present 
several advantages particularly important for hormonal contraceptives delivery: 
(1) They do not require daily attention, allowing higher compliance than with daily 
dosage forms; (2) fl exibility of current rings allow them to be easily inserted and 
removed by the woman herself, not requiring medical assistance as in the case of 
subcutaneous or intrauterine devices; (3) the continuous and prolonged delivery 
(three weeks to one year) of hormones avoids the high peak concentrations and 

fl uctuations seen with daily oral administration; (4) rings are not associated with 
adverse local effects, including cytological and normal fl ora changes; and (5) contraceptive 
rings may be removed from the vagina during sexual intercourse and up 
to 2 h, without compromising their pharmacological effect [110 – 112] . Although 
vaginal rings have been essentially investigated and used for the delivery of sexual 
hormones with contraceptive purposes or as hormone replacement therapy, these 
drug delivery systems can be also useful for the administration of other drugs 
such as bromocriptine mesylate, danazol, oxybutynin, antigens, and microbicides 
[113 – 117] . 
In the 1960s, fi rst reports that implants made of polysiloxane, containing sexual 
steroids, could release their content at constant rates in saline solutions provided 
early information that led to the development of the fi rst vaginal rings [118] . Vaginal 
rings were initially developed in the 1970s as contraceptives. The fi rst system was 
composed of a silicone rubber ring containing medroxyprogesterone acetate as the 
active substance [119] . Nonetheless, the fi rst vaginal rings have just recently reached 
the market, due to several unpredictable obstacles such as formulation diffi culties, 
safety issues, and poor ovulation suppression [120, 121] . Table 3 presents some 
vaginal rings currently available in the market. 
Vaginal rings may present several designs, as seen in Figure 4 . The fi rst 
vaginal rings were made of a homogeneous matrix containing the mixture of 
poly(dimethylsiloxane) (matrix - forming polymer) and the active drug, usually 
referred as a matrix design. Unfortunately, these rings showed an initial burst effect 
due to rapid release of the drug contained in the system ’ s surface followed by persistent 
linear decrease of the drug release rate. This later phenomenon is related to 
the gradually thickening of a drug - depleted boundary between the inner drug - 
loaded region and the release surface, which is created by continuous drug release 
from the outer layers. Thus, their use in clinical practice was compromised, namely 
TABLE 3 Selected Vaginal Rings Currently Available 
Commercial 
Name 
Active 
Substance(s) 
Clinical 
Indications Availability Company 
Nuvaring Etonogestrel 
and ethinyl 
estradiol 
Contraception United States, 
Europe, 
Brazil, Chile 
Organon 
Progering Progesterone Contraception 
in lactating 
women 
Chile Laboratorios 
Silesia 
Femring Estradiol 
acetate 
Relief of vaginal 
and urogenital 
symptoms in 
menopausal 
women 
United States, 
Netherlands 
Warner Chilcott 
Laboratories 
Estring Estradiol Relief of vaginal 
and urogenital 
symptoms in 
menopausal 
women 
United States, 
Canada, 
Europe, 
South Africa 
Pfi zer 
VAGINAL DRUG DELIVERY SYSTEMS 827

828 VAGINAL DRUG DELIVERY 
as contraceptive devices. Later, and in order to improve control of the drug release, 
a layer of poly(dimethylsiloxane) without active substance was applied over the core 
containing the drug, acting as a drug release – limiting sheath (core or reservoir 
design). This strategy allowed achieving a near zero - order drug release profi le. The 
diffusion rate of reservoir design rings is dependent on the drug concentration in 
the core, its partition coeffi cient between the core and membrane, the thickness and 
surface area of the membrane, and the diffusion coeffi cient of the drug in the membrane. 
In order to achieve a constant release rate, the drug should be much more 
permeable through the core than through the membrane [122] . Rings with several 
independent reservoirs containing different drugs have been obtained, thereby 
allowing the administration of two or more active substances from the same device. 
Also, another design has been developed in order to overcome drug release drawbacks, 
comprising a core of poly(dimethylsiloxane), an intermediate layer of 
poly(dimethylsiloxane) containing the active substance, and an outer drug release – 
limiting membrane of poly(dimethylsiloxane) (sandwich or shell design). As the 
drug is closer to the releasing surface, this strategy is particularly suited for substances 
presenting poor polymer diffusion characteristics [123] . As with reservoir 
design rings, a near zero - order drug release profi le is obtained. 
Besides poly(dimethylsiloxane), other elastomeric polymers have been employed 
in the manufacturing of vaginal rings, such as poly(dimethylsiloxane/vinylmethylsiloxane), 
styrene – butadiene – styrene block copolymer, and poly(ethylene - co - vinyl 
acetate) [123 – 125] . In fact, poly(ethylene - co - vinyl acetate) (commonly referred as 
EVA) appeared in the mid 1990s as an alternative to poly(dimethylsiloxane), when 
the manufacturer of this last material stopped supplying it for human use, demonstrating 
it to be very suitable for the production of controlled - release systems. 
At the laboratory scale, silicone vaginal rings are usually obtained by injection 
molding, where poly(dimethylsiloxane) is mixed with a polymerization catalyst and 
the drug, being subsequently injected in ring - shaped molds. The mixture is allowed 
to cure for a period of time at a preestablished temperature, which can range from 
FIGURE 4 Cross sections of three vaginal rings presenting different designs: matrix design 
(left), core or reservoir design (center), and sandwich or shell design (right). Light gray represents 
drug – polymer mixture and dark gray represents polymer only. 

room temperature to 150 ° C and over (higher temperatures allow increasing the 
speed of the ring curing). In fact, curing time and temperature should be optimized 
as they infl uence the fi nal performance of the ring, particularly drug release (Figure 
5 ). This step leads to the formation of a three - dimensional network by means of a 
cross - linking reaction between polymer chains [126] . Afterward, other layers can be 
added, a step that is usually performed by injection molding or a dipping process. 
Although vaginal rings produced at the laboratory scale are useful during preclinical 
and clinical experimentation, the pharmaceutical industry needs other manufacture 
solutions to allow large - scale and fi nancially viable production of these drug delivery 
systems. This process scale - up requires proof of bioequivalence between rings 
obtained by both processes [109] . 
The most common process of obtaining vaginal rings in the pharmaceutical 
industry is hot - melt extrusion (or hot - melt spinning), where the polymer, either 
alone or mixed with drugs or other additives, is melted (usually between 105 and 
120 ° C) and forced by single - or twin - extrusion screws to pass through a die. After 
leaving the die, the obtained coaxial fi ber is cooled and cut, the obtained fragments 
being shaped as rings by gluing both ends with an adequate pharmaceutical adhesive. 
Figure 6 presents a simplifi ed scheme of the manufacturing process of a reservoir 
design ring similar to one used to produce Nuvaring (NV Organon, Oss, The 
Netherlands), an EVA vaginal ring containing etonogestrel and ethiny lestradiol. 
The core [polymer and active substance(s)] and the surrounding membrane polymer 
mixtures are extruded separately through two single - screw extruders (coextrusion) 
FIGURE 5 Effect of curing conditions on in vitro release profi le of estradiol ( . g) from 
core design rings containing estradiol and progesterone: T: polymer - only outer sheath 
[poly(dimethylsiloxane/vinylmethylsiloxane)]; E2 + P: core sheath, comprising mixture of 
polymer [poly(dimethylsiloxane/vinylmethylsiloxane)], estradiol, and progesterone. 
(Reprinted with permission from S. I. Saleh et al., Journal of Pharmaceutical Sciences , 92, 
258 – 265, 2003. Copyright 2003 by Wiley - Liss.) 
T 
E2 + P 
Days 
Cured at 105°C, 30 min 
Cured at 140°C, 15 min 
Cured at 140°C, 30 min 
Amount released, .g/day 
800 
700 
600 
500 
400 
300 
200 
100
0 
0 5 10 15 20 25 30 35 
VAGINAL DRUG DELIVERY SYSTEMS 829

830 VAGINAL DRUG DELIVERY 
that are connected to a spinning pump. In these cylindrical pipelines the polymers 
are melted and extruded through a die at an accurate fl ow rate. Then, the core and 
membrane polymers are combined in a spinneret, forming a coaxial fi ber. The ratio 
between fl ow rates of both spinning pumps determines the thickness of the membrane. 
After leaving the spinneret, this fi ber is cooled, fi rst by air exposure and then 
by immersion in a water bath. At this stage, the fi ber diameter is adjusted to the 
desired value by elongation with take - up rolls. In fact, after leaving the die, the 
obtained fi ber expands its diameter as a result of the viscoelastic behavior of 
the polymers used [122, 127] . 
The drug release profi le is conditioned by the polymeric structure of the systems, 
which is infl uenced by several parameters such as polymer composition, melt spinning 
process variables (namely feeding of polymer mixture and spinning velocity, 
extrusion temperature, spinline stress, cooling rate, and drawing back elongating 
force), and storage conditions [122, 128] . Also, drug release characteristics are largely 
infl uenced by the active substances ’ molecular weight and diffusion coeffi cient and 
solubility in the polymer. For example, extremely hydrophobic drugs and molecules 
with molecular weight above 450 Da are poorly released from silicone. Modulation 
of the drug solubility by adding various excipients (e.g., propylene glycol, polyethylene 
glycol, gelatin, and fl uid silicone) can be used to change the release profi le [129, 
130] . The physical state of the drug is also an important parameter related to drug 
release. Taking the example of steroids, these compounds can be either in a solid 
crystalline state or in a molecularly dissolved state. In the fi rst case, the concentration 
of the drug is fi xed by its saturation solubility, making it possible to control the 
release rate by the thickness and the permeability of an outer membrane. When two 
drugs are present, this concept cannot be used: Both drugs need to be completely 
dissolved, their release rates being controlled by their concentrations [123] . 
FIGURE 6 Schematic of manufacturing process of reservoir design ring by hot - melt 
extrusion. 

5.12.4.7 Vaginal Films 
Vaginal fi lms are polymeric drug delivery systems shaped as thin sheets, usually 
ranging from 220 to 240 . m in thickness. These systems are often square (approximately 
5 cm . 5 cm), colorless, and soft, presenting a homogenous surface. Vaginal 
fi lms have some advantages, such as portability, ease of application, long time of 
retention in the vagina, and good drug stability [131] . Once placed in the vagina, 
the fl uid present in the mucosa hydrates the polymer, covering the mucosa with the 
active substance. This coating may also be helped during sexual intercourse due to 
the spreading motion of the male penis. 
Vaginal fi lms are produced with polymers such as polyacrylates, polyethylene 
glycol, polyvinyl alcohol, and cellulose derivatives. Proper combination of these 
polymer is essential to achieve adequate mucoadhesion and optimal drug release 
profi les. Vaginal fi lms can be produced by casting [132] , in which polymer solutions 
containing the active substance(s) are poured into adequate molds and dried until 
a thin, solid, and fl exible polymeric sheet is formed. Afterward, the sheet is cut in 
small pieces (individual fi lms) and peeled off. 
Vaginal fi lms have been mostly used as spermicides, although they present inferior 
contraceptive success than hormonal methods, condoms, and intrauterine 
devices. A contraceptive fi lm containing 28% nonoxynol - 9 in a polyvinyl alcohol 
base (VCF, Apothecus Pharmaceutical) is currently available in the United States. 
Alternative contraceptive fi lms containing different drugs and fi lm - forming polymers 
have also been investigated, in order to obtain more effective and acceptable 
formulations [133] . 
5.12.4.8 Medicated Vaginal Tampons 
Vaginal tampons have been studied for their feasibility as vaginal drug delivery 
systems. First experiments used commercially available tampons that were impregnated 
with active substances by a simple dipping process [134] . Currently, a medicated 
vaginal tampon, approved as a medical device by the Food and Drug 
Administration (FDA), is available (Ela Tampon, Rostam, Israel). This bifunctional 
tampon contains a polymeric delivery system (strips) that absorbs menstrual fl uid 
while gradually releasing lactic acid and citric acid. These two drugs act by preserving 
the acid vaginal milieu, preventing the proliferation of potential pathogenic 
bacteria [135] . 
5.12.4.9 Vaginal Foams 
Vaginal foams have been tested to deliver drugs in the vagina, mainly microbicides 
or spermicides [136, 137] . The effi cacy of these systems was shows to be limited when 
compared to other available options, leading to a decline of their use. Nonetheless, 
foams are easy to use, providing a good coverage of the vaginal mucosa with 
minimal leaking. Also, most foam bases are nonirritating, unlike some other conventional 
formulations which are reported to cause burning and itching. Thus, 
improving their formulation can be an interesting approach to obtaining new vaginal 
drug delivery systems. In fact, foams containing antimicrobials, local anesthetics, and 
hormones have the potential to gradually substitute several currently available 
dosage forms, namely creams and ovules. 
VAGINAL DRUG DELIVERY SYSTEMS 831

832 VAGINAL DRUG DELIVERY 
5.12.4.10 Vaginal Sponges 
Vaginal sponges were once widely used as vaginal contraceptives, the oldest reference 
to their use in the Talmud (c. 500 b . c. ), where a sponge soaked in vinegar was 
recommended in order to prevent pregnancy [138] . Although these devices have 
been disapproved by some researchers, since their use encouraged colonization and 
proliferation of bacteria in the vagina, predisposing women to vaginitis and other 
genital disorders [139] , sponges containing spermicides are still used as contraceptives. 
These contraceptive devices have the ability to deliver the active drugs while 
absorbing semen and blocking the cervical canal. Also, vaginal sponges are inexpensive 
devices; less messy than creams, gels, and foams; and easier to insert and remove 
than diaphragms [140] . Nonetheless, these systems have demonstrated less effi cacy 
than other contraceptive methods, such as the diaphragm [141] . 
A vaginal sponge (Today, Whitehall Robins) made of soft polyurethane foam 
(diameter of 5.5 cm and 2.5 cm thick) and saturated with 1 g of nonoxynol - 9 was 
authorized as a spermicide in the United States in 1983 [140] . Advantages of this 
formulation include the possibility of being used up to 24 h without requiring additional 
application of spermicide. Although in 1995 the manufacturer discontinued 
its production because of increased costs related to FDA guideline compliance, 
Allendale Pharmaceuticals purchased the rights to the sponge, reintroducing the 
product in U.S. and Canadian markets [142] . Similar sponges are available outside 
the United States containing benzalkonium chloride alone (Pharmatex, Innotech 
International) or a combination of nonoxynol - 9 and sodium cholate (Protectaid, 
Pirri Pharma). 
5.12.4.11 Other Strategies and Vaginal Drug Delivery Systems 
Bioadhesion may be defi ned as the state in which two materials, at least one of which 
is biological in nature, are held together for extended periods of time by interfacial 
forces. Mucoadhesion is a particular case of bioadhesion where one of these materials 
is a mucous membrane or the mucus [143] . Mucoadhesive drug delivery systems 
can circumvent the poor retention that most traditional vaginal formulations present, 
improving residence time, and even their specifi c location in the mucosa. Prolonged 
time of contact and intimate interaction with the vaginal mucosa are able to increase 
drug absorption and bioavailability and thus its therapeutic effect. Currently used 
mucoadhesive materials are polymeric in nature, many of them previously used for 
other specifi c purposes (e.g., as gelling agents in vaginal gels). Several polymers, 
alone or in combination, have been used or investigated for both systemic and local 
vaginal drug delivery in order to obtain pharmaceutical systems with mucoadhesive 
properties (see Table 4 ) [144] . Virtually almost all drug delivery systems may benefi t 
from these mucoadhesives, particularly gels, creams, tablets, vaginal suppositories, 
and fi lms. To date, polyacrylates are the most used and explored mucoadhesive 
polymers, although others such as chitosan, carrageenan, or sodium alginate have 
proved to be advantageous. Also, some widely used mucoadhesives in vaginal formulations, 
such as polycarbophil (Noveon AA - 1, Noveon, Cleveland, OH) present 
the advantage of being useful as controlled - and sustained - release matrices [145] . 
Recently, thiolated polymers, usually referred as thiomers, have been studied in 
vaginal mucoadhesive systems, exhibiting higher mucoadhesion than nonmodifi ed 

polymers. Although thiomers can improve the mucoadhesion of the original polymers, 
they do not interfere with other advantageous characteristics such as the 
ability of controlled release [146, 147] . 
Environmentally sensitive drug delivery systems are characterized by their ability 
to respond to changes in pH, temperature, ionic strength, solvent composition, magnetic 
fi elds, light, and electric current, among others, most commonly used for the 
fi rst two [148] . Thermoreversible systems are fl uids that can be introduced into the 
body in a minimally invasive manner prior to solidifying or gelling within the local 
of administration because of temperature increase from room temperature to body 
temperature [149] . This innovative approach is particularly interesting in vaginal 
drug delivery, as a liquid system is easily introduced in the vaginal cavity, while the 
in situ formed gel facilitates retention. Indeed, some thermosensitive formulations 
have already been suggested to obtain vaginal drug delivery systems, poloxamer 
gels being the most extensively studied. A clotrimazole - containing vaginal thermosensitive 
gel (poloxamer 188 and polycarbophil) was demonstrated to be useful for 
the effective and convenient treatment of vaginal candidiasis in female rats, with 
the advantages of facilitating administration and reducing dosing interval when 
compared with conventional therapy [150] . 
The incorporation of drug - loaded liposomes in adequate formulations can 
improve their stability, allowing their applicability in vaginal drug delivery [151] . 
Additionally, entrapment of drugs in liposomes may improve their solubility and 
TABLE 4 Examples of Mucoadhesive Polymers Used or Investigated in Formulation of 
Vaginal Drug Delivery Systems 
Polymer Classes Examples 
Carrageenan Iota - carrageenan 
Cellulose derivatives Sodium carboxymethylcellulose (NaCMC) 
Methylcellulose (MC) 
Hydroxypropyl methylcellulose (HPMC) 
Hydroxypropyl cellulose (HPC) 
Hydroxyethyl cellulose (HEC) 
Chitosans 
Gelatin 
Gums Pectin 
Tragacanth 
Dextran 
Xanthan 
Hyaluronic acid and derivatives Sodium hyaluronate 
Polyacrylates Carbopol 974P 
Polycarbophil (Noveon ® AA-1) 
Polyethylene glycols (PEGs) PEG 600 
Povidone 
Alginates Sodium alginate 
Starch 
Sulfated polysaccharides Cellulose sulfate 
Thiomers Carbopol ® 974P – cysteine 
Chitosan – thioglycolic acid (TGA) 
Chitosan – 4 - thio - butylamidin - conjugates (TBA) 
Note : As reviewed in ref. 144 . 
VAGINAL DRUG DELIVERY SYSTEMS 833

834 VAGINAL DRUG DELIVERY 
availability at the site of administration, reducing the administered dose and systemic 
effects. In order to administer liposomes, adequate dosage forms must be 
selected. Polyacrylate gels have emerged as good candidates, showing the potential 
to accommodate liposomes for vaginal drug delivery [151, 152] . Incorporation of 
liposomes in these gel bases can be achieved by gently mixing both components, 
which is an advantage over other dosage forms. Also, these novel drug delivery formulations 
enable sustained drug release, combining both the liposome limiting 
release effect and the bioadhesive properties of these gels. 
When liposomes are considered for vaginal administration, their stability should 
be assessed under in situ conditions presented by both pre - and postmenopausal 
women. Drug formulation can help improve their stability. Several gels made of 
Carbopol 974P NF were shown to be suitable in improving the stability of liposomes 
containing clotrimazole, metronidazole, or acyclovir in a vaginal environment simulating 
media when compared to liposomal dispersions [153, 154] . Also, compatibility 
between liposomes and vaginal mucosa (animal or human) should be checked [155] . 
Nonetheless, liposomes still have some stability - related problems that limit their 
shelf life. Formulation of liposomes as proliposomes can prevent these stability 
issues. Proliposomes are dry, free - fl owing products which on addition of water disperse 
to form liposomal suspensions [156] . As proliposomes are dry powders, they 
can be formulated as solid dosage forms, which can be more convenient to administer 
in the vagina. Recent work by Ning et al. demonstrated that clotrimazole - 
containing proliposomes can be suitable for vaginal administration, allowing a 
rapid conversion to sustained - release liposomes upon contact with physiological 
fl uids [157] . 
Niosomes (nonionic surfactant vesicles) have been experimented as controlled - 
and prolonged - release drug delivery systems to administer insulin through the 
vagina. These particles might be good carriers for protein delivery, showing the 
potential to enhance the effects of these drugs when compared to the vaginal administration 
of free insulin. Results obtained in rats showed that niosomes containing 
insulin can achieve comparable bioavailability with subcutaneous administration of 
this protein [158] . Also, experiments performed with vaginal gels containing both 
niosomes and liposomes have been shown to be a promising strategy for the prolonged 
release and safe vaginal administration of clotrimazole [159] . 
Microparticles and nanoparticles present some advantageous features, namely 
mucoadhesive properties. They have demonstrated some potential in vaginal drug 
delivery, particularly in the formulation of delivery systems for vaccines or peptides 
and proteins [160, 161] . Nonetheless, these particles have to be incorporated in 
adequate carrier systems in order to be delivered. This task has been shown to be 
complex, it being hard to achieve controlled - release and steady - release profi les. 
Cyclodextrins are commonly used in pharmaceutics, their applicability being no 
exception in vaginal drug delivery. Drug/cyclodextrin complexes allowed enhancement 
of solubility and achievement of prolonged - release properties of drugs such 
as clotrimazole and itraconazole when included in suitable vaginal drug delivery 
systems such as gels, creams, or tablets [162 – 164] . 
Cervical barrier devices such as diaphragms can be modifi ed to include a reservoir 
that releases active substances such as spermicides or microbicides. Although 
these devices have limited applicability, they can be particularly helpful in prevent

ing pregnancy, as they combine a physical barrier device with a chemical spermicide 
that faces the vagina, after correct placement in the cervix [165] . Lee et al. presented 
a diaphragm made from silicone containing 35% nonoxynol - 9 that was able to 
achieve controlled drug release depending on the device design and size [166] . The 
diaphragm was prepared by using compression molding in a single - cavity aluminum 
mold. 
Patches can provide delivery of very toxic drugs in very limited areas of the 
vaginal mucosa without leaking to the circumventing tissue. McCarron et al. 
described a bioadhesive patch that delivered 5 - aminovulinic acid to intraepithelial 
neoplasia lesions. The proposed delivery system is based on a poly(methyl - 
vinylether/maleic anhydride) matrix that contains the active drug, providing local 
retention of up to 4 h and enhancing effi cacy without damaging healthy epithelium 
[167] . Patchs with bilayer design may also be an option for the treatment of these 
neoplasic lesions, where a bioadhesive drug - loaded matrix bonded to a drug - 
impermeable backing layer is able to prevent drug spillage to healthy tissues, promoting 
unidirectional and deep drug penetration. Woolfson et al. presented a 
5 - fl uorouracil vaginal patch with a bilayer design, comprising a fl exible polyvinyl 
chloride (PVC) emulsion as a backing layer and a drug - loaded bioadhesive fi lm 
made of 2% Carbopol 981 and 1% glycerin as a plasticizer [168] . 
Propess (Ferring Pharmaceuticals) is a commercially available controlled - release 
vaginal insert presented as a thin macrogol 8000 matrix (rectangular in shape with 
radiused corners) containing 10 mg of dinoprostone being used for labor inducement. 
This drug - loaded matrix is included within a knitted polyester retrieval system 
that ends in a long tail to help retrieval at the end of the dosing interval. This insert 
is capable of releasing the active substance at a rate of 0.8 mg/h over 12 h, after being 
exposed to vaginal moisture [169] . Clinical studies indicate that Propess is as equally 
effective and safe in achieving cervical ripening as other commercially available 
vaginal drug delivery systems containing dinoprostone [170] . 
Multiple emulsions have been studied as drug vehicles for vaginal administration 
that are able to include several active substances in the different phases. An antimicrobial 
water – oil – water (W/O/W) multiple emulsion containing lactic acid in the 
internal aqueous phase, octadecylamine in the oily phase, and benzalkonium chloride 
in the external aqueous phase was tested, proving to stabilize the included 
active substances while providing adequate viscosity properties to vaginal administration 
[171] . Nonetheless, the structure of these formulations is destroyed at high 
shear rates (e.g., shear rates observed during coitus), losing or diminishing their 
intended activity and thus limiting their applicability. 
Recently, an interesting approach for protein delivery using genetically engineered 
normal vaginal fl ora as delivery systems has been proposed. This strategy is 
based on the natural affi nity that these microorganisms have to adhere tightly to 
the epithelial surface, providing a direct delivery of the drugs to the mucosa and 
thus minimizing enzymatic and bacterial degradation. Lactic acid bacteria, transformed 
with plasmids that contained a gene encoding for the therapeutic protein 
to be administered, have been used as delivery systems. Obtained results showed 
enhancement of protein delivery when compared with a conventional solution containing 
the same molecule. This strategy looks particularly appealing for the development 
of vaccines that induce mucosal immunization [172, 173] . 
VAGINAL DRUG DELIVERY SYSTEMS 835

836 VAGINAL DRUG DELIVERY 
5.12.4.12 Packaging and Vaginal Applicators 
Vaginal packaging and applicators are an integral part of vaginal products. Packaging 
is designed to accommodate and protect formulations, while applicators should 
allow their convenient administration in the vagina. Several materials have been 
used to manufacture these devices, such as plastics (e.g., polypropylene and polyethylene) 
and nonlatex rubber. Besides compatibility, stability, and suitability issues, 
these materials should be selected regarding the fi nal cost of packaging and 
applicators. 
Applicators are intended to be introduced in the vagina, adequately deliver the 
product, and then be removed. Their design relates to safety (e.g., relationship with 
product purity and stability, avoidance of local trauma associated with insertion or 
use), effi cacy (e.g., consistent delivery of the required amount of product in the 
intended location), and acceptability (comfort, ease of use, convenience, aesthetic 
appeal) [174] . In general, they can be divided as single - use or reusable applicators. 
Single - use applicators are usually prefi lled, while reusable applicators are fi lled by 
women prior to vaginal insertion. Several applicator designs have been used, such 
as barrel - and - plunger and squeeze tube, but they all should be easy to insert, comfortable, 
and deprived of cutting edges. Also, some specifi c formulations, such as 
those intended for vaginal douching, require other types of applicators. Typically, 
squeeze plastic bottles with variable volumes (approximately 100 – 200 mL) are 
used. 
5.12.5 PHARMACEUTICAL EVALUATION OF VAGINAL DRUG 
DELIVERY SYSTEMS 
Evaluation of pharmaceutical systems is consensually recognized as an important 
component of their development and quality control. Although evaluation of general 
features (e.g., drug content) is also required for vaginal formulations, this section 
focuses only upon some of the most important parameters that are intimately 
related to drug delivery systems specifi cally designed to be administered by this 
route. 
5.12.5.1 Legal and Offi cial Compendia Requirements 
There is a lack of well - defi ned guidelines and regulations for vaginal products in 
most countries as well as offi cial compendia information and requirements on 
quality control and other important aspects of vaginal drug delivery systems [39] . 
In fact, the latest editions of the U.S. Pharmacopeia, European Pharmacopoeia, and 
Japanese Pharmacopeia include limited or even no information related to the quality 
control and evaluation of vaginal drug delivery systems. Thus, most of the currently 
used evaluation procedures require standardization, making it diffi cult to compare 
results obtained by different research groups. 
5.12.5.2 Drug Release and Permeability 
When formulating vaginal drug delivery systems, it is important to consider the 
release of the active substances, as different formulations can greatly affect the 

release rate and ultimately their pharmacological effects. The choice of the dissolution 
method should be done on a case - by - case basis, while dissolution profi les can 
then be fi tted to commonly used mathematical models, as reviewed by Costa and 
Sousa Lobo [175] . Conventional procedures and apparatus have been adapted 
taking into consideration vaginal physiological specifi cations such as pH, fl uid 
volume, and temperature, among others. The experimental method may not inevitably 
imitate the vaginal environment, but it should test the main key performance 
indicators of the formulation. Although offi cial methods are not available, some 
have been recommended for specifi c vaginal drug dosage forms [176] . For semisolid 
vaginal formulations, the Franz diffusion cell is considered the most promising 
apparatus for drug release investigation. In the case of hydrophilic vaginal suppositories, 
a basket apparatus, a paddle apparatus, or fl ow - through cells can generally 
be considered as suitable; for hydrophobic vaginal suppositories, modifi ed fl ow - 
through cells would be preferable. Dissolution methods that use a basket instead of 
a paddle can be advantageous for vaginal solid formulations that tend to fl oat, 
particularly those that may include a modifi cation that prevents the formulations to 
form a cake inside the basket, limiting their dissolution [177, 178] . Drug release from 
vaginal rings is usually determined by placing these systems in conical fl asks containing 
an adequate dissolution medium. Flasks are then placed in a water bath controlled 
at 37 ° C and shaken during the time of the assay, with samples being taken 
and release medium being replaced typically every 24 h [113, 129] . Correlation of 
these in vitro release tests with in vivo results proved to be satisfactory for the 
majority of the evaluated vaginal rings [125] . 
Absorption of drugs through the vagina is an important parameter to be evaluated, 
particularly when a systemic effect is required. Also, assessment of the absorption 
potential of drugs intended to locally exert their effects needs to be evaluated, 
as this event can lead to unwanted systemic effects. The evaluation of both formulated 
and unformulated drugs can be performed by in vitro or in vivo methods. 
In vitro permeability studies have been performed in fl ow - through diffusion cells 
using either animal or human vaginal mucosa [179, 180] . After isolation and adequate 
treatment of vaginal mucosa specimens, small tissue disks are mounted in the 
apparatus in order to perform the permeation experiments. At the end of these 
procedures, routine histological examination of the used tissues can be performed 
in order to identify any changes in the normal structure of the vaginal mucosa. 
Several alterations can suggest potential mechanisms of permeation of tested drugs 
[181, 182] . Animal mucosa used in these experiments can be obtained from several 
species, namely rabbits and pigs. Also, porcine vaginal mucosa was demonstrated to 
be a good in vitro permeability model of human vaginal mucosa, particularly when 
hydrophobic substances are tested [50] . This feature can be explained because both 
mucosal tissues are very similar in many aspects, namely their lipid composition and 
histological structure. On the other hand, high molecular weight or charged molecules, 
such as oxytocin, may show different permeability profi les when tested with 
either porcine or human vaginal mucosa. Thus, researchers must be careful when 
interpreting and extrapolating results to human tissues because unexpected differences 
often occur. When human vaginal mucosa is used, samples are usually obtained 
from excess tissue removed from postmenopausal women after vaginal hysterectomy 
[181, 182] . Use of postmenopausal vaginal mucosa is advantageous as it is less 
altered (particularly in thickness) by hormonal stimulation, leading to more uniform 
PHARMACEUTICAL EVALUATION OF VAGINAL DRUG DELIVERY SYSTEMS 837

838 VAGINAL DRUG DELIVERY 
results. Nonetheless, these features do not refl ect the normal histological architecture 
of fertile women, being able to signifi cantly alter the permeability profi les for 
many drugs. Along with vaginal mucosal tissue, other model membranes, such as 
vaginal and cervical cell monolayer membranes, have been suggested in recent years 
in order to predict in vivo absorption [172] . 
It is also noteworthy that many of the in vitro results of vaginal permeability 
studies cited in the literature have limitations related to the experimental conditions 
that were used, particularly pH values at which they have been performed. Differences 
in permeability values are particularly expected when ionization characteristics 
change between experimental pH and vaginal pH [183] . However, in vitro 
results should only be considered as evidence that the vaginal mucosa is able to be 
permeated. 
In vivo studies performed in animals are an important step before considering 
human experimentation. Animal species commonly used in vaginal permeability 
studies include rabbits, rats, and mice [53, 158, 184] . Although potentially more 
accurate in predicting human vaginal absorption of drugs, animal experimentation 
have some limitations. A major problem is the variability of the vaginal epithelium 
properties throughout the estrous cycle, thus infl uencing drug absorption [184, 185] . 
In order to minimize this variability and standardize the thickness of the epithelium, 
animals are usually ovariectomized. Also, vaginal enzymatic activity is an important 
parameter in choosing animal models. It is recommended that the enzymatic profi le 
of such animal should be comparable to that of the woman. Taking this into consideration, 
rats and rabbits seem to be good models for vaginal permeability studies, 
particularly when protein and peptide drugs are considered [27] . 
5.12.5.3 p H and Acid - Buffering Capacity 
As already referred, pH is an important parameter concerning the health and 
normal physiology of the vagina, being important that vaginal formulations do not 
interfere with its normal value. Also, the pH of the vagina can be elevated due to 
changes in its normal physiology (e.g., bacterial vaginitis) or the presence of semen. 
Vaginal formulations presenting good acid - buffering capacity have the potential to 
reestablish normal pH or to prevent it from rising. 
The acid - buffering capacity of a vaginal formulation can be measured by simple 
titration with an inorganic alkali, such as sodium hydroxide. The physiologically 
relevant acid - buffering capacity can be defi ned as the amount of alkali required to 
elevate the pH from its initial value to the maximum desirable value when considering 
the healthy vagina [186] . Also, mixtures of vaginal formulations with semen may 
be useful in determining their buffering capacity, this proceeding being particularly 
helpful when testing products used during sexual intercourse. 
Variations in vaginal pH of the vagina can infl uence drug stability, particularly 
when extreme values are observed. Thus, the adjustment of the formulation pH can 
also be important in order to assure maximum stability or pharmacological activity 
of the active substance(s). As an example, the administration of antibodies in the 
vaginal milieu can compromise their activity because of the acidic pH. Generally 
monoclonal human antibodies are more stable at pH 4 – 7, losing binding and neutralizing 
activity below pH 4 [187] . These fi ndings underline the importance of pH 
buffering when delivering pH - sensible molecules such as antibodies. 

5.12.5.4 Rheological Studies 
Rheological properties of semisolid vaginal formulations are crucial to their suitability 
as drug delivery systems. They experience in vivo a wide range of shear rates 
(from less than 0.1 s . 1 to about 1000 s . 1 ), both steady and transient, while being 
diluted with vaginal fl uids, which infl uences their rheological properties and hence 
their spreading and retention. Events such as passive seeping, sliding, squeezing 
between vaginal walls, and coitus, among others, infl uence the rheological performance 
of vaginal formulations [188] . Thus, knowledge of rheological properties of 
semisolid vaginal drug formulations may assist in improving their design, being 
helpful in the process of predicting which formulations can retain their structural 
stability over time, particularly in the physiological environment [189, 190] . 
Qualitative and quantitative composition of a semisolid vaginal formulation can 
strongly infl uence its rheological properties [191] . This fact is particularly important 
when considering the optimization of a drug - containing formulation and its placebo 
formulation. These two systems should only differ in the absence of the active 
substance(s). However, this small difference can sometimes originate different 
rheological properties that can greatly infl uence the formulation ’ s performance and 
even the results of clinical trials [192] . 
Rheological properties of a formulation can be studied either by simple fl ow 
measurements or by dynamic oscillatory measurements, although the latter are 
preferable as they allow a complete characterization of both elastic and viscous 
components. Also, they are nondestructive and, if the strain is not too high, the 
sample is not disturbed [193] . As already noticed, in vivo conditions, particularly 
temperature and fl uids that may be present in the vagina (vaginal fl uid, cervical 
mucus, and semen), can infl uence the rheology of pharmaceutical formulations. 
Indeed, these factors have to be taken into account when formulating a vaginal drug 
delivery system. Thus, optimization should not only focus upon the rheology of 
undiluted material but also include mixtures of formulations and fl uids that may be 
present in the vagina [194] . However, these biological fl uids are not always available 
and considerable differences between individuals limit their use. In order to abbreviate 
these limitations, some simulants of vaginal fl uid [21] , cervical mucus [26] , and 
semen [195] have been used. 
5.12.5.5 Textural Studies 
Textural profi le analysis is a widely used analytical method based on the measurement 
of the forces involved during the compression/decompression of a probe in a 
sample of the product to be tested. From the obtained results, important parameters 
can be calculated, including hardness (force required to attain a given deformation), 
compressibility/spreadability (the work required to deform the product during the 
fi rst compression cycle of the probe), and adhesiveness of the product. Besides their 
infl uence in the ease of removal from a container (e.g., vaginal applicators), the ease 
of application, or retention, among others, it is consensual that textural properties 
of formulations will infl uence their clinical performance. Therefore, it is important 
to fully characterize these properties during the formulation process [196] . Also, 
these textural parameters can be converted into rheological properties, such as 
shearing stress, shear rate, and viscosity, using dimensional analysis, allowing the 
comparison of results generated by both techniques [197] . 
PHARMACEUTICAL EVALUATION OF VAGINAL DRUG DELIVERY SYSTEMS 839

840 VAGINAL DRUG DELIVERY 
5.12.5.6 Mucoadhesion 
As previously discussed, development of mucoadhesive drug delivery systems is a 
promising strategy in order to enhance vaginal drug administration. Several methods 
for the in vitro evaluation of mucoadhesive properties of vaginal formulations may 
be found in the literature. Although these tests present some limitations, they can 
offer easy and valuable tools in the initial formulation and evaluation of vaginal 
drug delivery systems. Most methods can be categorized as tensile strength or shear 
stress tests, where the force needed to separate a model membrane attached to the 
formulation measures mucoadhesion. Different results can be obtained for the same 
sample because of the different types of forces involved in these two methods [198] . 
For this reason, both types of tests should be performed in order to achieve a more 
complete characterization of the mucoadhesive potential of formulations. 
Alternative methods have also been shown to be useful. For example, mucoadhesive 
properties of semisolid vaginal formulations can be assessed by the rheological 
characterization of the mucoadhesive interface, based on the assumption that 
the interpenetration extension between polymer gels and their mixtures with mucin 
can be detected by measuring differences in rheological parameters. Also, the texture 
analysis of these formulations/mucin interfaces was demonstrated to be a useful 
technique in measuring bioadhesion, with results in the same rank as the ones 
obtained with the rheological technique [199] . Kast et al. tested vaginal tablets for 
their mucoadhesiveness by a simple method, comprising the use of a dissolution 
apparatus [146] . In this procedure, the formulation to be tested is attached to a 
vaginal mucosa that is fi xed on a cylinder; the cylinder is then immersed in the dissolution 
vessel and rotated in an adequate testing solution at approximately 37 ° C. 
The mucoadhesion is evaluated by the detachment time of the formulation. An 
alternative method based on a modifi ed balance, in which the tablet is attached to 
a vaginal mucosa fi xed to one side plate, has also been proposed [200] . In this case, 
mucoadhesion is measured by the weight required to detach the formulation. 
5.12.5.7 Vaginal Distribution and Retention 
Vaginal mucosa coating by a formulation is an important parameter to be determined, 
since its action may depend on the effectiveness of this phenomenon. Nonetheless, 
knowledge about the distribution and retention of commercially available 
products is limited. After being administered, liquid, semisolid, or solid (after liquefaction) 
formulations can spread throughout the vagina, with part of the material 
being able to exit this tube, either to the exterior or to the upper genital tract [201] . 
This distribution is governed by physical forces that include gravity, normal forces 
from contacting tissues, surface tension, and shearing. Formulation fl ow due to these 
forces is affected by many factors, including formulation physical properties, amount 
of formulation applied, surface interactions, surrounding tissue properties, vaginal 
secretions, baseline dimensions of the vagina, ambulation and posture changes of 
the user, and sexual intercourse [202, 203] . Drug delivery systems intended for local 
effect should ideally spread evenly throughout the vaginal mucosa. Also, evaluation 
of the possible erosion of a product coating during its residence in the vagina, particularly 
during sexual intercourse, is extremely important. Thus, vaginal distribution 
and retention studies may clarify some questions, such as the amount of product to 

cover evenly the vaginal surface, the time required to distribute to all areas and to 
be removed from the vagina, and the effect of daily activities such as ambulation 
and sexual activity in these phenomena. 
Simple in vitro tests can be used to quantify the vaginal coating of a vaginal formulation. 
It is important that these tests model the natural history of a product in 
the vagina, from initial application and contact with fl uids that may be present in 
the vagina to the period during and following sexual intercourse [204] . The distribution 
and retention of solid drug delivery systems can be evaluated by a simple 
method proposed by Ceschel et al. [205] , where the formulations are placed in a 
vertical thermostated cellophane tube, the discharged liquid collected and measured 
throughout the time of the experiment. The amount of discharge liquid is related 
to the retention while the distribution of formulations can be assessed by dosing 
the amount of active substance(s) in different sections of the tube at the end of the 
experiment. Also, the contribution of gravity to the vaginal coating fl ows of vaginal 
semisolid drug delivery systems can be evaluated by a simple and objective method 
proposed by Kieweg et al. [203] . The proposed technique is based on the measurement 
of the fl ow behavior of a formulation sample after being placed in an inclined 
plane surface. Obtained results, together with mathematical models, can help formulators 
to select primary candidate formulations before in vivo tests commence. 
In vivo assessment provides more reliable and complete information about the 
vaginal distribution and retention of drug delivery systems, although ethical and 
economical issues limit its applicability to routine evaluation. In recent years, imaging 
techniques, which have been used for other purposes for a long time, emerged as 
valuable tools for the evaluation of vaginal distribution and retention. Magnetic 
resonance imaging proved to be a helpful method, providing cross - sectional images 
of drug delivery systems that are administered in the vagina, both in animals and 
in humans, allowing precise reproducible data regarding the spread of vaginal formulations 
to be achieved [202] . As magnetic resonance imaging refl ects the images 
of a chemical label, such as gadolinium, and not necessarily that of the drug delivery 
systems or carried drugs, it is necessary to perform the association of the chemical 
label with vaginal products and its validation [206] . Another interesting imaging 
technique that has been used in this type of assessment is gamma scintigraphy, 
shown to be a useful tool for evaluating and comparing the distribution, spreading, 
and clearance of vaginal delivery systems [207, 208] . 
Imaging methods, such as gamma scintigraphy and magnetic resonance imaging, 
are useful but have some limitations concerning their resolution, being unable to 
quantify or even identify the presence of vaginal coating layers of just a few hundred 
micrometers. In order to overcome such limitations an optical instrument capable 
of detecting coating layers as thick as 50 . m has been developed by Henderson and 
co - workers [209] . The device is inserted and remains stationary in the vagina, where 
both local video images and fl uorescence intensity measurements of fl uorescein - 
labeled formulations are obtained. Since the tube that is inserted in the vagina is 
shaped and sized like a phallus, the vaginal coating measured is analogous to that 
observed during sexual intercourse. Nonetheless, in vivo fl uorescence - based methods 
present limitations, particularly because of the diffusion of the dye out of the formulation 
and photobleaching, limiting the interval over which measurements can 
be performed accurately. A new technique based on low - coherence interferometry 
that can overcome these diffi culties is being developed, allowing extended time 
PHARMACEUTICAL EVALUATION OF VAGINAL DRUG DELIVERY SYSTEMS 841

842 VAGINAL DRUG DELIVERY 
studies to be performed [210] . This easy, label - free, high - resolution method uses 
broadband light in an interferometry scheme to achieve depth - resolved refl ection 
measurements. Future studies focus on the development of an easy - to - use endoscopic 
device that may be used in clinical studies. 
5.12.5.8 Safety and Toxicology 
It is accepted that a new pharmaceutical product should be assessed for its effects 
on the vaginal mucosa before being approved by drug - licensing agencies. Nonetheless, 
many of the older products that have been used for a long time by women all 
over the world do not have this type of information available. Safety issues are 
particularly important when a vaginal drug formulation is used repeatedly, as in the 
case of microbicides, spermicides, and contraceptive vaginal rings. Also, it is important 
to test vaginal applicators for their safety as they are considered an integral 
part of vaginal products, being able to induce alterations in the mucosa [211] . 
Local effects assessment should include not only short - term but also long - term 
protocols, as some formulations are intended to be used for large periods, in order 
to assess their real impact on vaginal health. Although systemic exposure to drugs 
intended for topical action is expected to be minor, vaginal formulations should also 
be assessed for systemic effects due to possible absorption. Alterations in blood 
parameters and liver and renal function should be investigated [212] . Also, drugs 
and formulations to be administered through the vaginal route must be assessed for 
fertility and teratogenic effects in animal models, before entering clinical trialing 
and human use [213] . 
In vitro testing helps formulation scientists understand and predict the potential 
harmful effects of formulations to the vaginal mucosa. Although animal testing still 
needs to be performed, these in vitro methods can be of great interest in initial 
screening of new products and formulations, reducing the amount of animal testing 
required. Also and unlike animal testing, in vitro testing can often differentiate 
products that are very mild in terms of toxicity potential. Thus, toxicity of vaginal 
products can be assessed using simple tests with epithelial cell monolayers, where 
maintenance of membrane integrity in the presence of testing formulation indicates 
potential safety [214, 215] . Nonetheless, monolayer cell cultures lack histological 
and functional resemblance with native ectocervical and vaginal tissues, which limits 
the interpretation of the obtained results. In order to respond to this and other 
problems, Ayehunie et al. recently proposed a fast and highly reproducible three - 
dimensional organotypic vaginal – ectocervical tissue model that simulates the structure 
of the vaginal epithelium [216] . 
The standard preclinical test of local vaginal irritation and toxicity of pharmaceutical 
products, and the only one recommended by the FDA, is the rabbit vaginal 
irritation test [217, 218] . However, reproducibility problems and differences in 
vaginal physiology when compared to women limit the interpretation of results. 
Other animal models have also been used, namely primates, dogs, guinea pigs, pigs, 
mice, and rats [219 – 223] . Classic animal testing is limited because of the number of 
animals required, which makes testing burdensome, expensive, and ethically questionable, 
and because of differences between species, which may jeopardize the 
extrapolation of results to humans [224] . Thus, simpler toxicity tests performed 
in nonvertebrate organisms, such as gastropods, are interesting alternatives to 

vertebrate animal testing. These tests also proved to be superior than in vitro testing, 
mainly because of the limitations that are intrinsic to simple cell culture models 
[225] . Recently, a simple in vitro test using slugs ( Arion lusitanus ) has been proposed 
as an alternative to vertebrates in order to screen new vaginal semisolid formulations 
for local tolerance early in the development process. The irritation potential 
is evaluated by mucus production, and protein and enzyme release (lactate dehydrogenase 
and alkaline phosphatase). Experimental results showed that the slug 
mucosa irritation test performance is comparable to the classically used rabbit 
vaginal irritation test [226] . 
When initiating human testing, symptoms and signs of genital irritation must be 
assessed [227] . These investigations should be performed comparing results between 
the formulation to be tested (vehicle plus active substance(s) and vehicle only) and 
formulations that are well known for their irritative effects. In the late 1980s and 
early 1990s colposcopy of the vagina and cervix began to be used in the in vivo 
safety assessment of vaginal products, becoming a standard technique [228] . The 
objective of this procedure is to detect epithelial changes, such as breaks in the epithelium, 
infl ammation, or other not well characterized, that may be a consequence 
of vaginal products usage. Although very important, it presents several limitations 
such as costs, specifi c personnel training, and diffi culty in understanding which colposcopic 
fi ndings indicate risk [229] . Also, other techniques, such as Papanicolau 
stained smears or automated cytomorphometric analysis, have been used in order 
to assess the effect of formulations on the vaginal mucosa [230] . 
5.12.5.9 Other Characteristics 
In addition to the discussed evaluation tests and methodologies, other characteristics 
of vaginal formulations may be assessed according to their individual specifi cities. 
For example, the compatibility of vaginal formulations with condoms is an 
important parameter to be determined, particularly when they are used during 
sexual intercourse. These studies are usually performed according to the American 
Society for Testing and Materials (ASTM) norm D3492 - 89 (Standard Specifi cation 
for Rubber Contraceptives), where condoms are tested by accelerated testing for 
their tensile strength and elongation on break point after being exposed to vaginal 
formulations [231, 232] . Also, it is important to consider the effects of vaginal formulations 
that are used during sexual intercourse in the penis and the possibility 
of drug penetration through this organ. Although the human penis is covered 
with keratinized stratifi ed epithelium, and thus the expected absorption should 
be less than that of the vaginal epithelium, it is always a possibility to be taken into 
account [233] . 
5.12.6 CLINICAL USAGE AND POTENTIAL OF 
VAGINAL DRUG DELIVERY 
5.12.6.1 Microbicides 
Microbicides (initially termed “ virucides ” ) are anti - infective drugs formulated for 
topical self - administration in the vagina before sexual intercourse in order to protect 
against HIV and other sexually transmitted pathogens [234] . Once a neglected 
CLINICAL USAGE AND POTENTIAL OF VAGINAL DRUG DELIVERY 843

844 VAGINAL DRUG DELIVERY 
subject in the war against HIV and other sexually transmitted diseases, in the last 
decade microbicide investigations have gained important boosting and interest by 
the scientifi c community, being considered as a new approach for prevention [235] . 
Despite the fact that several microbicides are already in clinical trialing (see Table 
5 ), currently there are no available products on the market. Also, 100% effective 
microbicides are not likely to be achieved, even though only partially effective, 
microbicides can be a big help in reducing the spread of HIV infection. Investigators 
estimate that the use of a 60% effective microbicide in only 20% of all coital acts 
could prevent approximately 2.5 million infections over a period of 3 years [236] . 
While waiting for the fi rst - generation microbicides, preclinical research in new and 
improved microbicides is already in progress. Several candidates, such as PSC - 
RANTES [237] , antimicrobial peptides [238] , monoclonal antibodies [239] , inhibitors 
of virus – cell fusion [240] , and natural products [241] , are currently being 
developed. Also, another strategy that seems to be gaining consensus among the 
TABLE 5 Microbicides Currently Undergoing Clinical Trialing 
Mechanism 
of Action Active Substance(s) 
Candidate 
Product 
Drug 
Delivery 
Systems Developers 
Clinical 
Trial 
Status 
Vaginal 
defense 
enhancer 
— ACIDFORM Gel CONRAD/Instead Phase I 
— BufferGel Gel Reprotect Phases 
II/III 
Membrane 
disruptive 
agent/ 
surfactant 
C31G Savvy Gel CONRAD Phase 
III 
Entry/fusion 
inhibitor 
Carrageenan (PC - 515) Carraguard Gel Population Council Phase 
III 
Cellulose acetate 1,2 - 
benzenedicarboxylate 
Cellacefate/ 
CAP 
Gel Lindsey F. Kimball 
Research 
Institute / Dow 
Pharmaceuticals, 
Inc. 
Phase I 
Cellulose sulfate Cellulose 
Sulfate 
Vaginal Gel 
Gel CONRAD Phase 
III 
Sodium lauryl sulfate Invisible 
Condom 
Gel Laval University Phases 
I/II 
Naphthalene 2 - 
sulfonate polymer 
PRO 2000/5 Gel Indevus 
Pharmaceuticals 
Phase 
III 
SPL7013 VivaGel Gel Starpharma Phase I 
Replication 
inhibitor 
Tenofovir PMPA Gel Gel Gilead Sciences Phases 
II/IIb 
Dapivirine (TMC120) TMC120 
Vaginal Gel 
Gel and 
vaginal 
ring 
International 
Partnership for 
Microbicides 
Phases 
I/II 
UC - 781 UC - 781Gel Gel CONRAD Phase I 
Unknown 
mechanism 
Extracts of Azadirachta 
indica, Sapindus 
mukerossi , and 
Mentha citrata 
Praneem 
polyherbal 
Vaginal 
tablet 
Talwar Research 
Foundation 
Phases 
II/IIb 

scientifi c community is the synergistic association of microbicides with different 
action mechanisms in order to improve protection [242] . 
Early expectations created around the possibility of using nonoxynol - 9 (widely 
used as a vaginal contraceptive) as an effective microbicide, based on its in vitro 
effi cacy against HIV, were frustrated in clinical trials [243] . Several hypotheses for 
this failure were brought up, an inadequate choice of drug delivery system being 
one of them. These results confi rm that preformulation and formulation studies 
play an important role in microbicide rational design and development, being a 
big challenge to overcome. Although clinical development recommendations have 
been extensively reviewed [244, 245] , pharmaceutical development algorithms for 
microbicides are yet to be defi ned. Nonetheless, it is known that preformulation 
parameters such as organoleptic characteristics, stability, permeability, inherent 
bioadhesion and retention features, and compatibility with excipients and condoms 
of candidate drugs, among others, play a crucial role when developing drug delivery 
systems containing microbicides [246] . After collection of this information, 
formulation studies are necessary in order to obtain a fi nal product that fulfi lls 
microbicide objectives and requirements, namely safety, effi cacy, acceptability, 
affordability, and regulatory duties [247] . As well as providing effective protection, 
microbicide formulations must also be safe on multiple exposures over time, chemically 
and physically stable, compatible with latex and other materials used in 
barrier devices, and affordable and acceptable to the end user. Ideally, they should 
be colorless, odorless, tasteless, and nonmessy [234, 248] . Tested microbicides have 
been formulated mostly as gels, although creams, vaginal rings, foams, sponges, 
vaginal suppositories, and fi lms may also be considered. Nonetheless, alternative 
innovative options for the delivery of microbicides have been developed in recent 
years. For instance, the formulation of a safe and inexpensive “ universal ” drug 
carrier for microbicidal substances is an interesting strategy that may potentially 
ensure the effi cacy of most currently researched molecules [249] . Another interesting 
approach for the delivery of microbicides was proposed by Chang et al. These 
researchers studied the possibility of using genetically modifi ed comensal vaginal 
bacteria ( Lactobacillus jensenii ) to produce anti - HIV proteins [250] . In vitro experiments 
showed that this strategy can be a new step toward an effective microbicide 
formulation. 
5.12.6.2 Antimicrobials 
Vaginitis is a common condition in women which can be caused by bacteria, yeasts, 
or protozoa. Treatment of vaginitis has been achieved by oral or vaginal administration 
of antimicrobials, often with similar effi cacy rates [251] . Several drugs are 
currently available for intravaginal treatment of bacterial (e.g., metronidazole, 
clindamycin), fungal (e.g., azoles, boric acid, nystatin), and protozoal (e.g., metronidazole) 
vaginitis [252] . Also, alternative vaginal therapies have been investigated 
in order to treat vaginitis. For instance, herbal formulations, particularly those containing 
essential oils, have been referred as potential antimicrobials for the treatment 
of both fungal and bacterial infections [253, 254] . Several vaginal drug delivery 
systems have been used in order to administer antimicrobial substances, particularly 
gels, creams, tablets, and vaginal suppositories. Current research in antimicrobial 
vaginal drug delivery systems is focused on more convenient single - dose 
CLINICAL USAGE AND POTENTIAL OF VAGINAL DRUG DELIVERY 845

846 VAGINAL DRUG DELIVERY 
formulations that can achieve clinical cure while improving patient compliance 
[255 – 257] . 
Vaginal treatment may be advantageous when compared with oral treatment, as 
systemic adverse effects are less likely to occur [258] . For example, Cunningham 
and co - workers conducted a study where systemic levels of metronidazole after 
vaginal administration (5 g of a 0.75% metronidazole gel) were residual when compared 
to the oral administration of a 500 - mg standard dose despite comparable 
clinical outcome [259] . This low level of vaginal absorption may be attributed to 
metronidazole ’ s poor lipid solubility. Similar results have been obtained for clindamycin 
when comparing intravaginal and intravenous routes [260] . Also, contraindication 
during pregnancy and possible interference with oral contraceptives are 
situations that probably recommend vaginal administration of these therapeutic 
agents over oral administration in women requiring treatment [261] . 
As previously discussed, vaginal pH plays an important role in the normal physiology 
of the vagina, being elevated in bacterial vaginitis. Vaginal acidifi ers, such 
as vitamin C in the form of tablets, can be effective in the treatment of vaginitis, 
with the advantage of maintaining or even improving the normal vaginal fl ora 
equilibrium, particularly in diabetic and pregnant women or those with recurrent 
bacterial vaginitis episodes [78, 262] . Also, other formulations such as acid - buffering 
gels were demonstrated to be potential helpers in the maintenance of a healthy 
vaginal milieu and as adjuvant of antimicrobials in the treatment of bacterial vaginosis 
[263, 264] . In fact, some gels may even be used in the treatment of bacterial 
vaginosis. For example, a mucoadhesive gel containing two polymers, polycarbophil 
and Carbopol 974P, was demonstrated to be effective in the treatment of bacterial 
vaginosis, even when compared with the clinical cure rate of vaginal metronidazole 
or tinidazole [265] . 
5.12.6.3 Hormonal Contraceptives and Hormonal Replacement Therapy 
The vaginal route is one of the many available routes for estrogen and progestogen 
delivery. When compared to the oral and transdermal route, vaginal administration 
of these hormones presents the advantage of needing a smaller quantity of drug 
to achieve similar systemic effects with less relative variability [266, 267] . Also, 
the inconveniences associated with subcutaneous implants are circumvented. Thus, 
vaginal hormonal contraception and replacement therapy were soon considered 
as a possibility. 
Hormonal contraception requires drug delivery systems that are able to achieve 
sustained blood levels of these substances, either by multiple administrations, as in 
the case of oral contraceptives, or by sustained drug release. Since daily vaginal 
administration would not be feasible and acceptable by most women, sustained - 
release systems were developed. This need led to the development of several 
devices, including vaginal rings [109] . Progestogen - only rings were initially developed, 
but menstrual bleeding problems and ineffi cacious control of ovulation led 
to discontinuation in several studies. Thus, combined estrogen – progestogen rings 
were the next natural step toward effective and acceptable formulations. Various 
combined estrogen – progestogen rings have proven to be highly effective as contraceptives, 
providing excellent inhibition of ovulation [41] . Also, serious vaginal 
lesions were shown to be unlikely to occur with short - and medium - term exposure 

to currently available rings, mainly due to their improved fl exibility and small 
dimensions [268] . Although other alternatives have also been studied, the most 
popular schedule for contraceptive vaginal rings is three weeks in, one week out. 
With this regimen the ring is inserted in the vagina on day 5 of the menstrual cycle 
and left in place for three weeks. On week 4 the ring is removed, allowing menstrual 
bleeding. A new ring (one - month ring) or the same one (over - one - month ring) is 
inserted after one week ring free [112] . Vaginal rings are easy to use, being self - 
administered by women. Once inserted in the vagina, the ring fi ts in the upper 
vagina and delivers the active substance(s) by contact with the vaginal mucosa (see 
Figure 7 ). 
Despite all the investigational work already performed, only one contraceptive 
vaginal ring, Nuvaring (Organon), is available in the United States and Europe. This 
three weeks in – one week out vaginal ring was the fi rst approved by the FDA, being 
available in the market since 2002. It is a fl exible and transparent ring containing 
etonogestrel and ethinyl estradiol in a poly(ethylene - co - vinylacetate) matrix, with 
an outer diameter of 54 mm and a cross section of 4 mm [269] . When placed in the 
vagina it releases 120 . g/day of etonogestrel and 15 . g/day of ethinyl estradiol. 
Clinical studies showed that Nuvaring is as effective and reliable as commonly used 
oral contraceptives, being well tolerated, convenient, and highly acceptable to most 
women and their partners [270 – 272] . Moreover, this contraceptive ring is able to 
provide lower and more stable systemic exposure to estrogens than other contraceptive 
options, namely combined oral contraceptives or transdermal patches, thereby 
reducing drug - related side effects (Figure 8 ) [267] . Other contraceptive vaginal rings 
that are still in premarket research include a one - year contraceptive ring being 
developed by the Population Council, New York. This nestorone and ethinyl estradiol 
containing ring has been shown to be as effi cacious as oral contraceptives when 
used up to 12 months on a 3 weeks in – 1 week out regimen [273, 274] . 
FIGURE 7 X - ray image of vaginal ring after placement in human vagina. ( Reprinted with 
permission from K. Malcolm et al., Journal of Controlled Release , 90, 217 – 225, 2003. Copyright 
2003 by Elsevier .) 
CLINICAL USAGE AND POTENTIAL OF VAGINAL DRUG DELIVERY 847

848 VAGINAL DRUG DELIVERY 
Although conventional emergency contraception usually comprises the use of 
orally administered levonorgestrel (either alone or in combination with ethinyl 
estradiol), the vaginal route has been shown to be an effi cacious alternative. The 
obtained hormonal plasma levels for the same oral dose are lower, but presumably 
high enough to prevent pregnancy [275] . Nonetheless, increases in the administered 
dose by the vaginal route have been suggested [276, 277] . 
Hormone replacement therapy has been a common practice for a long time in 
order to improve the quality of life of women suffering from acute symptoms related 
to menopause. Several formulations for oral, buccal, subcutaneous, parenteral, intrauterine, 
nasal, transdermal, or vaginal administration have been used for this purpose 
[278] . Although in recent years hormone replacement therapy has been associated 
with an increased risk of fatal breast cancer [279] , some postmenopausal women 
may still benefi t from this treatment. Estrogens have been used through the vaginal 
route for the treatment of vaginal symptoms associated with hormone decline in 
menopausal woman, such as dryness, dyspareunia, pruritus, irritation, discomfort, 
and atrophy and has been shown to be as effective as systemic therapy [280, 281] . 
Creams and vaginal suppositories were the fi rst vaginal drug delivery systems to 
be used to deliver estrogens. Recently, estradiol rings for urogenital symptoms 
therapy (low - dose rings) or vasomotor plus vaginal symptoms relief (usually higher 
dose rings) were approved in some countries, providing a new option for the 
FIGURE 8 Mean serum ethinyl estradiol concentration versus time for subjects treated 
during 21 days with Nuvaring ( n = 8), transdermal contraceptive patch (Evra, OrthoMcNeil 
Pharmaceutical; releases 20 . g ethinyl estradiol and 150 . g norelgestromin daily; n = 6), and 
combined oral contraceptive (COC) (Microgynon, Schering AG; contains 30 . g ethinyl estradiol 
and 150 . g levonorgestrel; n = 8). Subject exposure to ethinyl estradiol in Nuvaring group 
was on average 3.4 and 2 times lower than for subjects in patch and combined oral contraceptive 
groups, respectively. ( Reprinted with permission from M. W. van den Heuvel et al., Contraception 
, 72, 168 – 174, 2005. Copyright 2005 by Elsevier .) 

administration of these drugs [282] . For instance, Estring (Pharmacia & Upjohn), a 
currently marketed vaginal ring containing low - dose estradiol (releases 7.5 . g/day 
when placed in the vagina), proved to be effi cacious, well tolerated, and safe when 
used for up to a year in the treatment of urogenital symptoms in postmenopausal 
women [283, 284] . This silicone polymer ring has a diameter of 55 mm and a cross - 
sectional diameter of 9 mm and is used up to three months. Another similar ring 
containing estradiol acetate (Femring, Warner Chilcott), with a diameter of 56 mm 
and a cross - sectional diameter of 7.6 mm, is also currently available. 
Estrogens and progestogens have also been administered through the vagina in 
order to manage other conditions. In fact, vaginally administered progesterone is 
commonly used for luteal - phase support in women undergoing assisted reproduction 
treatment, allowing optimal uterine concentrations without the high serum 
levels observed by other routes (oral and intramuscular), possibly due to the fi rst - 
uterine - pass effect [285] . Several drug delivery systems such as capsules, tablets, 
vaginal suppositories, or gels have been used and have been shown to be equally 
effi cacious in increasing the chance of becoming pregnant. Nonetheless, sustained - 
release formulations allow fewer administrations per day with lower doses [286] . 
Also, progesterone vaginal rings have been successfully used for luteal - phase 
support [287] . 
5.12.6.4 Spermicides 
The vaginal use of spermicidal substances during sexual intercourse is perhaps the 
oldest method of contraception. However, the introduction of oral contraceptives 
and the intrauterine device in the 1960s led to the decay in their use. Since many of 
these substances also offer protection against sexually transmitted diseases, interest 
and investigation in this fi eld have recently increased [288] . Also, the development 
of new potential spermicides, namely antibodies [289] , contributed to further awareness. 
Currently used spermicides include nonoxynol - 9, octoxynol, benzalkonium 
chloride, and chlorhexidine. 
Nonoxynol - 9 has been used for more than 30 years as a spermicide in over - the - 
counter vaginal products, such as semisolid formulations, sponges, foams, fi lms, and 
others, in order to prevent pregnancy. Although relatively safe and effective, nonoxynol 
- 9 formulations are still not able to achieve the same decrease in pregnancy 
risk obtained with hormonal methods [290] . Thus, some strategies have been used 
to enhance nonoxynol - 9 effects while reducing its toxic effects. For instance, some 
synergistic associations with chelating agents that have themselves little spermicide 
activity, such as EDTA and ethylene glycol tetraacetic acid (EGTA), have been 
shown to be promising in this matter [291] . Also, coprecipitation of nonoxynol - 9 
with polyvinylpyrrolidone by a freeze - drying method can be useful, particularly 
when the formulation of a solid system is desirable, as this process is necessary to 
alter the chemical state (liquid to solid) of nonoxynol - 9 [292, 293] . Using these 
coprecipitates, a tablet with an inner core that provides slow release of nonoxynol - 9 
after its fast release of the outer core was shown to be an effi cient and safer way of 
delivering this spermicide in rabbits [294] . 
Semisolid formulations are often used as contraceptives, particularly gels. These 
drug delivery systems were demonstrated to be useful, namely in reducing the toxicity 
of nonoxynol - 9 [295] . Adequate formulation of these products has also been 
CLINICAL USAGE AND POTENTIAL OF VAGINAL DRUG DELIVERY 849

850 VAGINAL DRUG DELIVERY 
shown to be essential for their effi cacy. For example, the contraceptive effect of 
spermicides can be enhanced by adequate consistency of the formulation. When the 
viscosity of a formulation increases, contraceptive effi cacy may increase as a result 
of becoming more tenacious and more resistant to sperm migration, consequently 
decreasing its capacity of reaching the site of fertilization [296] . Other gel properties, 
such as pH and osmolarity, may also infl uence its spermicidal effects [297] . Moreover, 
vaginal rings and inserts have been proposed as adequate vehicles for the 
delivery of nonhormonal contraceptives [298, 299] . These systems may be advantageous 
due to their controlled and prolonged release properties. 
5.12.6.5 Labor Inducers and Abortifacients 
Cervical ripening for the induction of labor has been a common practice in modern 
obstetrics. Several drugs have been tested with this purpose, misoprostol, dinoprostone, 
and oxytocin the three most frequently used. These labor inducers are normally 
administered by the vaginal route as gels, tablets, suppositories, or inserts, all 
demonstrated to be effective and safe. Nonetheless, drug formulation and choice of 
drug delivery are important factors to be considered. In fact, different times to 
achieve cervical ripening have been observed in several studies due to different drug 
release profi les [300, 301] . 
Vaginal administration of prostaglandins can be useful in the termination of 
pregnancy. Misoprostol has been used to terminate unwanted pregnancies, demonstrating 
a relatively high effi cacy, even when different regimens have been tested 
[302 – 304] . Indeed, improved results with vaginal administered misoprostol may be 
expected, because of higher and prolonged serum drug concentrations obtained, 
when compared with the oral route [305] . It is also noteworthy that since vaginal 
formulations are usually not available, oral tablets containing misoprostol (Cytotec, 
Pfi zer) have been routinely used to administer this drug by the vaginal route. As 
these tablets are not specifi cally designed to be administered in the vagina, suboptimal 
clinical results may occur. 
5.12.6.6 Proteins and Peptides 
A few proteins and peptides, such as insulin, leuprolide, and salmon calcitonin, have 
been tested for their possible administration by the intravaginal route. In spite of 
the rich blood supply, relatively large surface area, and good permeability, vaginal 
absorption of peptides and proteins is infl uenced by hormonal - induced changes in 
the mucosa histology and enzymatic activity, thus limiting their administration 
through this route [306] . Nonetheless, rational design of adequate and innovative 
drug delivery carriers allowed considerable progress in protein and peptide vaginal 
delivery. For example, microspheres have shown good potential to deliver peptides 
and proteins, such as calcitonin, being able to increase drug stability and absorption. 
This enhancement of absorption is thought to be related with the intimate contact 
between the microspheres and the mucosa, resulting in high local concentrations at 
the site of absorption [307, 308] . Another tested approach to vaginal administration 
of proteins and peptides has been polymeric matrices, identical to the ones used in 
the design of vaginal rings. For instance, antibodies have been successfully administered 
in mice using poly(ethylene - co - vinyl acetate) disks [309] . Also, these and other 

polymeric matrices can provide long - term (up to several years), controlled, and 
high - dose topical delivery of antibodies [310, 311] . 
Although considered a route that offers little or no real opportunities for insulin 
administration, mainly because of low and variable levels of absorption, efforts have 
been made to systemically deliver insulin through the vaginal mucosa. In the early 
1980s, studies by Morimoto et al. showed that polyacrylic acid gels containing insulin 
were able to induce and maintain (up to 30 min) hypoglycemia when administered 
to rats and rabbits. However, sustained release was necessary to achieve longer time 
of hypoglycemia [312] . Later, Richardson et al. observed that insulin is almost not 
absorbed by the vaginal mucosa of ovariectomized rats in the absence of permeation 
enhancers, but the coadministration of substances such as sodium taurodihydrofusidate, 
polyoxyethylene - 9 - laurylether, lysophosphatidyl choline, palmitoylcarnitine 
chloride, and lysophosphatidyl glycerol signifi cantly increased its absorption and 
consequently hypoglycemia [59] . Also, the use of mucoadhesive microspheres as 
delivery systems for insulin improved the absorption rate of this drug in sheep, 
particularly when associated with permeation enhancers [55] . Recently, Degim et 
al. developed vaginal chitosan gels as carriers for insulin. Studies performed in 
rabbits showed that chitosan gels containing 5% dimethyl - . - cyclodextrin as a penetration 
enhancer may provide longer insulin release, offering a potential alternative 
to the parenteral route [313] . Also, Ning et al. investigated the suitability of niosomes 
as insulin carriers for vaginal administration in rats. Results demonstrated that the 
bioavailability of insulin when administered through the vaginal route was comparable 
to that of the subcutaneous route [158] . 
5.12.6.7 Vaccines 
Although once considered not to be a very promising approach, intravaginal vaccines 
have emerged in recent years as a potential noninvasive immunization strategy, 
particularly for the prevention of HIV transmission [314] . In fact, some animal 
and human experiments suggest that the obtained female genital tract immunization 
can be superior with vaginal administration of vaccines when compared to other 
routes, such as oral, nasal, or rectal [315 – 317] . Also, it is known that effective immunization 
against sexually transmitted diseases will require strong local genital tract 
as well as strong systemic antibody responses [318] . Mucosal vaccination presents 
several advantages over systemic immunization: improved safety profi le, minimization 
of adverse effects, ease of administration, and potentially lower costs. However, 
limitations such as epithelium changes with menstrual cycle, which leads to reduced 
mucosa permeability and poor antigen presentation during certain stages, and inactivation 
of antigens by exposure to the vaginal environment can modulate the 
magnitude of immune response inducement [319] . 
Phosphate - buffered saline has been conveniently used as a vehicle for the delivery 
of antigens. Although these liquid formulations facilitate vaginal administration, 
they allow poor retention, controlled release, and protection of the delivered antigens. 
Thus, development of adequate vaginal drug delivery systems plays an important 
role in the success of vaginal immunization. Thermosensitive delivery systems 
with adequate mucoadhesiveness may be useful in enhancing the mucosal and systemic 
immune responses, as they can increase the exposure and contact of antigens 
to the vaginal mucosa [320] . The combination of thermosensitive polymers such as 
CLINICAL USAGE AND POTENTIAL OF VAGINAL DRUG DELIVERY 851

852 VAGINAL DRUG DELIVERY 
poloxamers and mucoadhesive polymers such as polycarbophil were shown to be a 
good strategy in the development of mucoadhesive, thermosensitive, and controlled - 
release formulations for vaginal delivery of vaccines [321] . Also, antigen susceptibility 
to the vaginal environment can be circumvented by administering these molecules 
in protective systems, such as the one described by Shen et al., which comprises 
poly(ethylene - co - vinyl acetate) matrices containing plasmid DNA [322] . These disk - 
shaped devices, produced by a solvent evaporation technique described by Luo 
et al. [323] , were shown to be effective in inducing local immunity in mice, protecting 
and providing controlled and sustained delivery of plasmid DNA. In addition, other 
strategies for vaginal antigen administration have also been proposed, such as the 
use of nonpathogenic bacterial vectors [324 – 326] and microspheres [327] . 
5.12.6.8 Other Uses 
Many other drugs have been administered in the vagina for the management of 
either local or systemic conditions. Table 6 summarizes some of these reports. 
5.12.7 ACCEPTABILITY AND PREFERENCES OF WOMEN RELATED 
TO VAGINAL DRUG DELIVERY 
Usage of vaginal products is intimately related to taboos and presumptions associated 
with the knowledge and handling of genitalia. Results from a recent international 
survey showed that, despite improvements in recent decades, society ’ s attitude 
toward the vagina and its use as a drug delivery route is not very open and not as 
open as women would like it to be. Also, this study revealed that the vagina is not 
commonly recognized as a possible drug delivery route, with only approximately 
35% of women acknowledging this fact [342] . Although frequently overlooked, 
acceptability studies of vaginal products are important in predicting women ’ s compliance 
and thus their effectiveness. Dedicated studies to women ’ s acceptability and 
preferences toward vaginal drug delivery are not common, most information available 
resulting from parallel research to clinical trials. Also, most of these studies 
have focused on microbicides and spermicides, since their acceptance is decisive for 
their consistent use [343 – 345] . In fact, there can be a sense of guilt or negative 
feeling with this kind of vaginal product, contrasting with others that are prescribed 
by a physician for a specifi c gynecological problem, which seem to be deprived of 
moral issues due to the “ legitimacy ” of their use [346] . Acceptability studies should 
also be extended to sexual partners, particularly when evaluating products meant 
to be used during sexual intercourse [347] . Data collected from previous acceptability 
studies must be considered during the development of formulations in order 
to improve their suitability. For example, lubrication provided by a vaginal product 
seems to be important in determining its acceptability, as this feature may be 
regarded advantageous or not by different users [348] . Also, negative perceptions 
regarding product characteristics should be identifi ed during clinical trials in order 
to improve formulations. 
Although results found in the literature related to women ’ s preferences toward 
the vaginal route of drug administration may vary signifi cantly, particularly when 
different locations and cultures are considered, some general statements can be 

TABLE 6 Selected Drugs Administered in the Vagina 
Drugs Intended Use 
Drug Delivery 
Systems Comments References 
5 - Fluorouracil Treatment of intravaginal warts 
Gel Demonstrated to be effective, 
safe, 
and tolerable 328 
Maintenance therapy of cervical 
dysplasias after standard excisional 
or ablative therapy 
Cream Reduction of recurrence was achieved 
329 
Bromocriptine Therapy of hyperprolactinemia 
Oral tablet and 
vaginal 
suppository 
Proved to be effective and safe, without the adverse 
effects of oral administration; vaginal suppository 
obtained higher reduction in serum prolactin 
330 
Cabergoline Therapy of hyperprolactinemia Oral tablet Proved to be effective and safe, without adverse 
effects of oral administration 
331 
Chlorhexidine Prevention of peripartum infections of 
newborn 
Aqueous solution Proved to be useful 332 
Danazol Treatment of pelvic endometriosis 
Ring and vaginal 
suppository 
Effective without increased serum concentrations 
observed during oral therapy 
114 
Etoposide Management of cervical dysplasias 
associated with human papilloma 
virus (HPV) 
Vaginal 
suppository 
Demonstrated to be safe and tolerable 333 
Imiquimod Treatment of high - grade vaginal 
intraepithelial neoplasia 
Cream Results suggest it can be an alternative conservative 
therapy 
334 
Indomethacin Tocolysis in preterm labor 
Rectal 
suppository 
Vaginal administration proved to be more effective 
than conventional rectal plus oral administration 
335 
Lignocaine Cervical anesthesia during insertion of 
tenaculum 
Spray and gel No difference in pain management between spray 
and gel 
336 
Morphine Pain control as alternative to 
parenteral administration 
Tablet and 
vaginal 
suppository 
Requires close monitoring due to unpredictable 
bioavailability 
337 
Oxybutinin Treatment of urge urinary incontinence Insert Proved to be effective and safe in rabbits 338 
Propranolol Control of tachycardia Tablet Obtained serum levels were within . - blocking 
range and comparable to those achieved by oral 
route 
339 
Sildenafi 
l Endometrial development for embryo 
implantation 
Vaginal 
suppository 
Enhanced endometrial development was achieved 340 
Trichloroacetic 
acid 
Treatment of low - grade vaginal 
intraepithelial neoplasia 
Solution Proved to be effective as well as inexpensive and 
easy to perform 
341 
853

854 VAGINAL DRUG DELIVERY 
made. Ideally, formulations should be easy and comfortable to use, colorless, odorless, 
and messiness free. Also, products that are better retained in the vagina seem 
to be favored by women, since leakage is one of the most undesired feature of 
vaginal formulations [349] . Concerning preferred vaginal dosage forms, gels and 
creams seem to be the most popular among women. On the other hand, vaginal 
suppositories and tablets are among the most disliked dosage forms. Others, such 
as fi lms, present ambiguous results, mostly related with diffi culties during insertion 
[350, 351] . Vaginal rings have been shown to be highly acceptable by both women 
and their sexual partners, even during sexual intercourse. A trial conducted in North 
America and Europe showed that couples rarely felt the device during penile penetration, 
and when the ring was noticed, almost none of the partners seemed to 
mind [352] . Furthermore, insertion and removal of vaginal rings are judged to be 
easy by users [353] . 
Packaging and applicators may also infl uence women ’ s choice. Products that are 
placed in the vagina by means of an applicator seem to be preferred because it 
facilitates administration and avoids direct touching of genitalia during insertion. 
Applicator characteristics, namely length, width, color, fi lling features (single - use or 
reusable applicators), and ease of usage, are also known to infl uence women ’ s 
acceptability [351, 354] . 
5.12.8 VETERINARY VAGINAL DRUG DELIVERY 
As with humans, veterinary vaginal drug administration has been performed for a 
long time, particularly for the treatment of local infections, traditionally involving 
the use of vaginal suppositories, liquid formulations, or gels. Advantages of this route 
include the avoidance of damage to the skin or to tissue that is associated with 
injections, minor stress infl icted to the animals, and possibility of ceasing drug delivery 
at will [355] . Nonetheless, the fi rst major studies on veterinary drug delivery 
have been performed by Robinson in the 1960s with progestogen - impregnated 
polyurethane sponges [356] . Since then, the major use of the vaginal route has been 
the control of the estrus cycle in livestock by delivering progestogens and estrogens 
in a controlled fashion. Also, the administration of these hormones showed good 
results in treating reproductive disorders, such as ovarian quiescence, cystic ovary 
or cystic corpus luteum [357] . Estrus synchronicity is advantageous as it allows 
insemination of all or selected females in a herd or fl ock to occur during a single 
period of several hours or days [358] . With this purpose, several drug delivery 
systems have been developed, being generally based on polymeric matrices that 
are able to control release of drug content. These devices also present two common 
features: Retention is guaranteed by means of a gentle pressure applied to the 
mucosa and the existence of a mechanism (e.g., an attached string) that allows their 
easy removal at the end of the treatment. Table 7 presents a synopsis of these 
systems, as reviewed by Rathbone et al. [358, 359] . Also, some of these devices are 
shown in Figure 9 . In addition to the vaginal administration of progestogens and 
estrogens, delivery of other therapeutic agents through this route for both local and 
systemic effects has been investigated, namely 1,25 - dihydroxy vitamin D3 [360] , 
lactic acid – producing lactobacilli [361] , antimicrobials [362] , local anesthetics [363] , 
and vaccines [364] . 

TABLE 7 Selected Veterinary Vaginal Drug Delivery Systems for Control of Estrus Cycle in Livestock 
Drug Delivery 
Systems 
Active 
Substance(s) 
Matrix - Forming 
Polymers Brief Description Comments 
Sponges Progesterone, 
estradiol, and 
several 
progestins 
Polyurethane Polymeric devices impregnated with active 
substance(s), usually cylindrical shaped; 
alternative designs allow achieving zero - 
order release profi le 
Variable vaginal 
retention; inexpensive 
and simple to prepare 
PRID 
(Progesterone 
Releasing 
Intravaginal 
Device, InterAg, 
Hamilton, NZ) 
Progesterone Poly(dimethylsiloxane) Spiral - shaped device obtained by molding and 
curing polymer (containing active substance) 
onto stainless steel spiral by high - 
temperature ( . 190 ° C) injection molding; 
also 
may enclose hard gelatin capsule (containing 
estradiol) glued to device 
Excellent vaginal 
retention 
CIDR (Controlled 
Internal Drug 
Release, CEVA, 
Libourne, 
France) 
Progesterone Poly(dimethylsiloxane) T - shaped nylon spine over which the polymer 
(containing active substance) is molded and 
cured by high - temperature injection molding; 
several types developed: CIDR - S (rabbit 
eared in shape), CIDR - G (slimmer, straight 
T - shaped), 
CIDR - B (similar to CIDR - G but 
with increased dimensions) 
Excellent vaginal 
retention; only types G 
and B currently 
available 
INVAS 
(Intravaginal 
Application 
System) 
Progesterone Poly(dimethylsiloxane) Similar to CIDR but obtained by lower 
temperature ( < 120 ° C) method 
Not commercially 
available 
Rings — Poly(dimethylsiloxane) Similar to human vaginal rings Not commercially 
available; abandoned 
because of poor 
retention properties 
Rajamehendran 
intravaginal 
device 
17 . - estradiol/ 
progesterone 
Poly(dimethylsiloxane) Two C - shaped polymer tubes (impregnated 
with active substances) tied together, forming 
“ umbrella - shaped ” 
device 
Excellent vaginal 
retention; not 
commercially available 
IBD (Intelligent 
Breeding Device, 
Advanced 
Animal 
Technology, 
Hamilton, NZ) 
Progesterone/ 
estradiol/ 
prostaglandin 
Poly(dimethylsiloxane) Composed by outer plastic sheath [designed to 
protect circuit board (which controls drug 
release) and two batteries, four polymeric 
drug reservoirs (a large one at base and 
three small ones at head of device)], 
retention mechanism, and tail 
Allows release of active 
substances at different 
rates and at specifi c 
times 
Source : From refs. 358 and 359 . 
855

856 VAGINAL DRUG DELIVERY 
In order to improve the performance of drug delivery systems, several strategies 
have been tested. For example, improvement of currently available progestogen - 
and estrogen - delivering devices and development of new ones have been a common 
gold of vaginal drug delivery investigators [365] . In other therapeutic fi eld, Gavini 
et al. used bioadhesive chitosan microspheres compressed into tablets with several 
excipients to deliver acrifl avine [362] . In vitro results demonstrated that these 
systems have good mucoadhesive properties, allowing increased residence time in 
the vagina. Recently, Cross et al. proposed an interesting electronic device that 
allows controlling drug release in response to in loco stimuli (e.g., vaginal temperature) 
or external commanding via radio wireless link [366] . This device is placed 
inside and behind the piston of a modifi ed syringe, being able to control the production 
of hydrogen from a gas cell. The increased gas pressure behind the piston 
propels a viscous pharmaceutical vehicle that fi lls the syringe. This drug release and 
monitoring unit (DMU) have been shown to be a promising strategy in controlling 
the delivery of active substances to cows while simultaneously collecting physiological 
data in the vaginal environment. 
5.12.9 CONCLUSIONS AND FUTURE TRENDS 
As other routes of drug administration presented serious diffi culties to deliver some 
active substances, the vagina emerged as a feasible alternative. Undoubtedly, the 
vaginal route of drug delivery has attracted the interest of the scientifi c community, 
particularly in the last few decades. The latest developments in this fi eld, namely 
mucoadhesive formulations, vaginal rings, and other controlled - release drug delivery 
systems, have boosted research and clinical use of this once - neglected route of 
drug administration. Also, women ’ s emancipation in the last century has slowly led 
to higher acceptability of vaginal formulations, as old fears and preconceived ideas 
are being demystifi ed. 
FIGURE 9 Some vaginal veterinary drug delivery systems used or investigated to control 
estrus cycle in livestock: sponge (1), PRID (2), CIDR - S (3), CIDR - G (4), CIDR - B (5), 
Rajamehendran intravaginal device (6), and IBD (7). 

REFERENCES 857 
Much work remains to be done, particularly in specifi c fi elds, such as the delivery 
of macromolecular drugs (e.g., proteins and peptides) and other substances that 
are poorly absorbed through the vaginal mucosa. Another promising area that 
needs further investigation is vaginal administration of vaccines and microbicides. 
Indeed, in an era where HIV and other sexually transmitted diseases are an increasing 
concern, vaginal preventive strategies are required. Formulating scientists can 
contribute decisively to these objectives, as optimization of drug delivery systems 
seems to be essential. Issues such as poor vaginal distribution and retention, inadequate 
drug release, limited drug protection from vaginal “ aggressors, ” and adverse 
effects of currently available drug delivery systems still need to be solved. 
ACKNOWLEDGMENTS 
The authors would like to express their gratitude to Bruno Sarmento and Cl a udia 
Carneiro for their kind review and useful comments on the manuscript. 
REFERENCES 
1. O ’ Dowd , M. J. ( 2001 ), The History of Medications for Women: Materia Medica Woman , 
1st ed., Taylor & Francis , London, pp 53 – 58. 
2. O ’ Dowd , M. J. , and Philipp , E. E. ( 2000 ), The History of Obstetrics and Gynecology , 1st 
ed., Taylor & Francis , London, pp 1 – 40. 
3. Stenchever , M. A. , Droegemueller , W. , and Herbst , A. L. ( 2002 ), Comprehensive Gynecology 
, 4th ed., Mosby , St. Louis, MO , pp 40 – 44 . 
4. Van De Graff , K. ( 2001 ), Human Anatomy , 6th ed., McGraw - Hill , London , pp 725 – 753 . 
5. Barnhart , K. T. , Izquierdo , A. , Pretorius , E. S. , Shera , D. M. , Shabbout , M. , and Shaunik , 
A. ( 2006 ), Baseline dimensions of the human vagina , Hum. Reprod. , 21 , 1618 – 1622 . 
6. Berman , J. R. , and Bassuk , J. ( 2002 ), Physiology and pathophysiology of female sexual 
function and dysfunction , World J. Urol. , 20 , 111 – 118 . 
7. Pendergrass , P. B. , Belovicz , M. W. , and Reeves , C. A. ( 2003 ), Surface area of the human 
vagina as measured from vinyl polysiloxane casts , Gynecol. Obstet. Invest. , 55 , 110 – 113 . 
8. Barnhart , K. T. , Pretorius , E. S. , and Malamud , D. ( 2004 ), Lesson learned and dispelled 
myths: Three - dimensional imaging of the human vagina , Fertil. Steril. , 81 , 1383 – 1384 . 
9. Fawcett , D. W. , and Raviola , E. ( 1994 ), Bloom and Fawcett, A Textbook of Histology , 12th 
ed., Chapman & Hall , New York , pp 857 – 858 . 
10. Patton , D. L. , Thwin , S. S. , Meier , A. , Hooton , T. M. , Stapleton , A. E. , and Eschenbach , 
D. A. ( 2000 ), Epithelial cell layer thickness and immune cell populations in the normal 
human vagina at different stages of the menstrual cycle , Am. J. Obstet. Gynecol. , 183 , 
967 – 973 . 
11. Ildgruben , A. K. , Sjoberg , I. M. , and Hammarstrom , M. L. ( 2003 ), Infl uence of hormonal 
contraceptives on the immune cells and thickness of human vaginal epithelium , Obstet. 
Gynecol. , 102 , 571 – 582 . 
12. Mauck , C. K. , Callahan , M. M. , Baker , J. , Arbogast , K. , Veazey , R. , Stock , R. , Pan , Z. , 
Morrison , C. S. , Chen - Mok , M. , Archer , D. F. , and Gabelnick , H. L. ( 1999 ), The effect of 
one injection of Depo - Provera on the human vaginal epithelium and cervical ectopy , 
Contraception , 60 , 15 – 24 . 

858 VAGINAL DRUG DELIVERY 
13. Wagner , G. , and Ottesen , B. ( 1982 ), Vaginal physiology during menstruation , Ann. Intern. 
Med. , 96 , 921 – 923 . 
14. Caillouette , J. C. , Sharp , C. F. , Jr. , Zimmerman , G. J. , and Roy , S. ( 1997 ), Vaginal pH as a 
marker for bacterial pathogens and menopausal status , Am. J. Obstet. Gynecol. , 176 , 
1270 – 1275 . 
15. Ramsey , P. S. , Ogburn , P. L. , Jr. , Harris , D. Y. , Heise , R. H. , and Ramin , K. D. ( 2002 ), 
Effect of vaginal pH on effi cacy of the dinoprostone gel for cervical ripening/labor 
induction , Am. J. Obstet. Gynecol. , 187 , 843 – 846 . 
16. Boskey , E. R. , Cone , R. A. , Whaley , K. J. , and Moench , T. R. ( 2001 ), Origins of vaginal 
acidity: High D/L lactate ratio is consistent with bacteria being the primary source , Hum. 
Reprod. , 16 , 1809 – 1813 . 
17. Boris , S. , and Barb e s , C. ( 2000 ), Role played by lactobacilli in controlling the population 
of vaginal pathogens , Microbes Infect. , 2 , 543 – 546 . 
18. Zhou , X. , Bent , S. J. , Schneider , M. G. , Davis , C. C. , Islam , M. R. , and Forney , L. J. ( 2004 ), 
Characterization of vaginal microbial communities in adult healthy women using 
cultivation - independent methods , Microbiology , 150 , 2565 – 2573 . 
19. Larsen , B. , and Galask, R. P. (1982), Vaginal microbial fl ora: Composition and infl uences 
of host physiology , Ann. Intern. Med. , 96 , 926 – 930 . 
20. Woolfson , A. D. , Malcolm , R. K. , and Gallagher , R. ( 2000 ), Drug delivery by the intravaginal 
route , Crit. Rev. Ther. Drug Carrier Syst. , 17 , 509 – 555 . 
21. Owen , D. H. , and Katz , D. F. ( 1999 ), A vaginal fl uid simulant , Contraception , 59 , 
91 – 95 . 
22. Widmaier , E. P. , Raff , H. , and Strang , K. T. ( 2004 ), Vander, Sherman, Luciano ’ s Human 
Physiology: The Mechanisms of Body Function , 9th ed., McGraw - Hill , New York , 
pp 658 – 688 . 
23. Terranova , P. F. ( 2003 ), The female reproductive system , in Rhoades , R. A. , and Tanner , 
G. A. , Eds. Medical Physiology , 2nd ed., Lippincott, Williams & Wilkins , Philadelphia , 
pp 667 – 683 . 
24. Valore , E. V. , Park , C. H. , Igreti , S. L. , and Ganz , T. ( 2002 ), Antimicrobial components of 
vaginal fl uid , Am. J. Obstet. Gynecol. , 187 , 561 – 568 . 
25. Eggert - Kruse , W. , Botz , I. , Pohl , S. , Rohr , G. , and Strowitzki , T. ( 2000 ), Antimicrobial 
activity of human cervical mucus , Hum. Reprod. , 15 , 778 – 784 . 
26. Burruano , B. T. , Schnaare , R. L. , and Malamud , D. ( 2002 ), Synthetic cervical mucus formulation 
, Contraception , 66 , 137 – 140 . 
27. Acarturk , F. , Parlatan , Z. I. , and Saracoglu , O. F. ( 2001 ), Comparison of vaginal aminopeptidase 
enzymatic activities in various animals and in humans , J. Pharm. Pharmacol. , 
53 , 1499 – 1504 . 
28. Gibbs , D. F. , Labrum , A. H. , and Stagg , B. H. ( 1968 ), Vaginal fl uid enzymology. A new 
assay method with enzyme - potassium ratios , Am. J. Obstet. Gynecol. , 102 , 982 – 988 . 
29. Belec , L. ( 2002 ), D e fenses non immunes, pr e - immunes et immunes du tractus 
g e nital f e minin contre les infections , J. Gynecol. Obstet. Biol. Reprod. (Paris) , 31 , 
034302.1 – 034302.7 . 
30. Mestecky , J. , and Fultz , P. N. ( 1999 ), Mucosal immune system of the human genital tract , 
J. Infect. Dis. , 179 ( Suppl. 3 ), S470 – 474 . 
31. Russell, M. W. (2002), Immunization for protection of the reproductive tract: A review , 
Am. J. Reprod. Immunol. , 47 , 265 – 268 . 
32. Mestecky , J. , Moldoveanu , Z. , and Russell , M. W. ( 2005 ), Immunologic uniqueness of 
the genital tract: Challenge for vaccine development , Am. J. Reprod. Immunol. , 53 , 
208 – 214 . 

REFERENCES 859 
33. Hocini , H. , Barra , A. , Belec , L. , Iscaki , S. , Preud ’ homme , J. L. , Pillot , J. , and Bouvet , J. P. 
( 1995 ), Systemic and secretory humoral immunity in the normal human vaginal tract , 
Scand. J. Immunol. , 42 , 269 – 274 . 
34. Beagley , K. W. , and Gockel , C. M. ( 2003 ), Regulation of innate and adaptive immunity 
by the female sex hormones oestradiol and progesterone , FEMS Immunol. Med. Microbiol. 
, 38 , 13 – 22 . 
35. Wira , C. R. , Rossoll , R. M. , and Kaushic , C. ( 2000 ), Antigen - presenting cells in the female 
reproductive tract: Infl uence of estradiol on antigen presentation by vaginal cells , Endocrinology 
, 141 , 2877 – 2885 . 
36. Franklin , R. D. , and Kutteh , W. H. ( 1999 ), Characterization of immunoglobulins and 
cytokines in human cervical mucus: Infl uence of exogenous and endogenous hormones , 
J. Reprod. Immunol. , 42 , 93 – 106 . 
37. Farage , M. , and Maibach , H. ( 2006 ), Lifetime changes in the vulva and vagina , Arch. 
Gynecol. Obstet. , 273 , 195 – 202 . 
38. Nilsson , K. , Risberg , B. , and Heimer , G. ( 1995 ), The vaginal epithelium in the postmenopause 
— Cytology, histology and pH as methods of assessment , Maturitas , 21 , 51 – 56 . 
39. Vermani , K. , and Garg , S. ( 2000 ), The scope and potential of vaginal drug delivery , 
Pharm. Sci. Technol. Today , 3 , 359 – 364 . 
40. Bernkop - Schnurch , A. , and Hornof , M. ( 2003 ), Intravaginal drug delivery systems , Am. 
J. Drug Deliv. , 1 , 241 – 254 . 
41. Alexander , N. J. , Baker , E. , Kaptein , M. , Karck , U. , Miller , L. , and Zampaglione , E. 
( 2004 ), Why consider vaginal drug administration ? Fertil. Steril. , 82 , 1 – 12 . 
42. Hussain , A. , and Ahsan , F. ( 2005 ), The vagina as a route for systemic drug delivery , 
J. Controlled Release , 103 , 301 – 313 . 
43. Macht , D. I. ( 1918 ), On the absorption of drugs and poisons through the vagina , J. Pharmacol. 
Exp. Ther. , 10 , 509 – 522 . 
44. Robinson, G. D. (1925), Absorption from the human vagina, BJOG , 32 , 496 – 504 . 
45. Rock , J. , Barker , R. H. , and Bacon , W. B. ( 1947 ), Vaginal absorption of penicillin , Science , 
105 , 13 . 
46. van der Bijl , P. , and van Eyk , A. D. ( 2003 ), Comparative in vitro permeability of human 
vaginal, small intestinal and colonic mucosa , Int. J. Pharm. , 261 , 147 – 152 . 
47. van Eyk , A. D. , and van der Bijl , P. ( 2004 ), Comparative permeability of various chemical 
markers through human vaginal and buccal mucosa as well as porcine buccal and mouth 
fl oor mucosa , Arch. Oral Biol. , 49 , 387 – 392 . 
48. Villanueva , B. , Casper , R. F. , and Yen , S. S. ( 1981 ), Intravaginal administration of progesterone: 
Enhanced absorption after estrogen treatment , Fertil. Steril. , 35 , 433 – 437 . 
49. Sassi , A. B. , McCullough , K. D. , Cost , M. R. , Hillier , S. L. , and Rohan , L. C. ( 2004 ), Permeability 
of tritiated water through human cervical and vaginal tissue , J. Pharm. Sci. , 93 , 
2009 – 2016 . 
50. van Eyk , A. D. , and van der Bijl , P. ( 2005 ), Porcine vaginal mucosa as an in vitro permeability 
model for human vaginal mucosa , Int. J. Pharm. , 305 , 105 – 111 . 
51. van der Bijl , P. , and van Eyk , A. D. ( 2004 ), Human vaginal mucosa as a model of buccal 
mucosa for in vitro permeability studies: An overview , Curr. Drug Deliv. , 1 , 129 – 135 . 
52. van der Bijl , P. , Penkler , L. , and van Eyk , A. D. ( 2000 ), Permeation of sumatriptan 
through human vaginal and buccal mucosa , Headache , 40 , 137 – 141 . 
53. Corbo , D. C. , Liu , J. C. , and Chien , Y. W. ( 1989 ), Drug absorption through mucosal 
membranes: Effect of mucosal route and penetrant hydrophilicity , Pharm. Res. , 6 , 
848 – 852 . 

860 VAGINAL DRUG DELIVERY 
54. Corbo , D. C. , Liu , J. C. , and Chien , Y. W. ( 1990 ), Characterization of the barrier properties 
of mucosal membranes , J. Pharm. Sci. , 79 , 202 – 206 . 
55. Richardson , J. L. , Farraj , N. F. , and Illum , L. ( 1992 ), Enhanced vaginal absorption of 
insulin in sheep using lysophosphatidylcholine and a bioadhesive microsphere delivery 
system , Int. J. Pharm. , 88 , 319 – 325 . 
56. Okada , H. , Yamazaki , I. , Yashiki , T. , and Mima , H. ( 1983 ), Vaginal absorption of a potent 
luteinizing hormone - releasing hormone analogue (leuprolide) in rats II: Mechanism of 
absorption enhancement with organic acids , J. Pharm. Sci. , 72 , 75 – 78 . 
57. van der Bijl , P. , van Eyk , A. D. , Gareis , A. A. , and Thompson , I. O. ( 2002 ), Enhancement 
of transmucosal permeation of cyclosporine by benzalkonium chloride , Oral Dis. , 8 , 
168 – 172 . 
58. Richardson , J. L. , Thomas , N. W. , and Illum , L. ( 1991 ), Recovery of the rat vaginal epithelium 
from the histological effects of absorptions enhancers , Int. J. Pharm. , 77 , 
75 – 78 . 
59. Richardson , J. L. , Illum , L. , and Thomas , N. W. ( 1992 ), Vaginal absorption of insulin in 
the rat: Effect of penetration enhancers on insulin uptake and mucosal histology , Pharm. 
Res. , 9 , 878 – 883 . 
60. Sayani , A. P. , Chun , I. K. , and Chien , Y. W. ( 1993 ), Transmucosal delivery of leucine 
enkephalin: Stabilization in rabbit enzyme extracts and enhancement of permeation 
through mucosae , J. Pharm. Sci. , 82 , 1179 – 1185 . 
61. Richardson , J. L. , Minhas , P. S. , Thomas , N. W. , and Illum , L. ( 1989 ), Vaginal administration 
of gentamicin to rats. Pharmaceutical and morphological studies using absorption 
enhancers , Int. J. Pharm. , 56 , 29 – 35 . 
62. Nakada , Y. , Miyake , M. , and Awata , N. ( 1993 ), Some factors affecting the vaginal absorption 
of human calcitonin in rats , Int. J. Pharm. , 89 , 169 – 175 . 
63. van der Bijl , P. , and van Eyk , A. D. ( 2001 ), Areca nut extract lowers the permeability of 
vaginal mucosa to reduced arecoline and arecaidine , J. Oral Pathol. Med. , 30 , 537 – 541 . 
64. Cicinelli , E. , Einer - Jensen , N. , Galantino , P. , Alfonso , R. , and Nicoletti , R. ( 2004 ), The 
vascular cast of the human uterus: From anatomy to physiology , Ann. N. Y. Acad. Sci. , 
1034 , 19 – 26 . 
65. Cicinelli , E. , Cignarelli , M. , Sabatelli , S. , Romano , F. , Schonauer , L. M. , Padovano , R. , 
and Einer - Jensen , N. ( 1998 ), Plasma concentrations of progesterone are higher in the 
uterine artery than in the radial artery after vaginal administration of micronized progesterone 
in an oil - based solution to postmenopausal women , Fertil. Steril. , 69 , 
471 – 473 . 
66. Bulletti , C. , de Ziegler , D. , Flamigni , C. , Giacomucci , E. , Polli , V. , Bolelli , G. , and 
Franceschetti , F. ( 1997 ), Targeted drug delivery in gynaecology: The fi rst uterine pass 
effect , Hum. Reprod. , 12 , 1073 – 1079 . 
67. Cicinelli , E. , Di Naro , E. , De Ziegler , D. , Matteo , M. , Morgese , S. , Galantino , P. , Brioschi , 
P. A. , and Schonauer , A. ( 2003 ), Placement of the vaginal 17beta - estradiol tablets in the 
inner or outer one third of the vagina affects the preferential delivery of 17beta - estradiol 
toward the uterus or periurethral areas, thereby modifying effi cacy and endometrial 
safety , Am. J. Obstet. Gynecol. , 189 , 55 – 58 . 
68. Thorgeirsdottir , T. O. , Hilmarsson , H. , Thormar , H. , and Kristmundsdottir , T. ( 2005 ), 
Development of a virucidal cream containing the monoglyceride monocaprin , Pharmazie , 
60 , 897 – 899 . 
69. Thorgeirsdottir , T. O. , Thormar , H. , and Kristmundsdottir , T. ( 2005 ), The infl uence of 
formulation variables on stability and microbicidal activity of monoglyceride monocaprin 
, J. Drug Deliv. Sci. Tech. , 15 , 233 – 236 . 

REFERENCES 861 
70. Kristmundsdottir , T. , Sigurdsson , P. , and Thormar , H. ( 2003 ), Effect of buffers on the 
properties of microbicidal hydrogels containing monoglyceride as the active ingredient , 
Drug Dev. Ind. Pharm. , 29 , 121 – 129 . 
71. Garg , S. , Tambwekar , K. R. , Vermani , K. , Garg , A. , Kaul , C. L. , and Zaneveld , J. D. ( 2001 ), 
Compendium of pharmaceutical excipients for vaginal formulations , Pharm. Technol. , 
25 , 14 – 24 . 
72. Knapczyk , J. , Macura , A. B. , and Pawlik , B. ( 1992 ), Simple test demonstrating the antimycotic 
effect of chitosan , Int. J. Pharm. , 80 , 33 – 38 . 
73. Manson , K. H. , Wyand , M. S. , Miller , C. , and Neurath , A. R. ( 2000 ), Effect of a cellulose 
acetate phthalate topical cream on vaginal transmission of simian immunodefi ciency 
virus in rhesus monkeys , Antimicrob. Agents Chemother. , 44 , 3199 – 3202 . 
74. Katz , D. F. , and Dunmire , E. N. ( 1993 ), Cervical mucus: Problems and opportunities for 
drug delivery via the vagina and cervix , Adv. Drug Deliv. Rev. , 11 , 385 – 401 . 
75. Willits , R. K. , and Saltzman , W. M. ( 2001 ), Synthetic polymers alter the structure of cervical 
mucus , Biomaterials , 22 , 445 – 452 . 
76. Karasulu , H. Y. , Taneri , F. , Sanal , E. , Guneri , T. , and Ertan , G. ( 2002 ), Sustained release 
bioadhesive effervescent ketoconazole microcapsules tabletted for vaginal delivery , 
J. Microencapsul. , 19 , 357 – 362 . 
77. Knapczyk, J. (1992), Antimycotic buccal and vaginal tablets with chitosan, Int. J. Pharm. , 
88 , 9 – 14 . 
78. Petersen , E. E. , and Magnani , P. ( 2004 ), Effi cacy and safety of vitamin C vaginal tablets 
in the treatment of non - specifi c vaginitis. A randomised, double blind, placebo - 
controlled study , Eur. J. Obstet. Gynecol. Reprod. Biol. , 117 , 70 – 75 . 
79. Digenis , G. A. , Nosek , D. , Mohammadi , F. , Darwazeh , N. B. , Anwar , H. S. , and Zavos , 
P. M. ( 1999 ), Novel vaginal controlled - delivery systems incorporating coprecipitates 
of nonoxynol - 9 , Pharm. Dev. Technol. , 4 , 421 – 430 . 
80. Notelovitz , M. , Funk , S. , Nanavati , N. , and Mazzeo , M. ( 2002 ), Estradiol absorption from 
vaginal tablets in postmenopausal women , Obstet. Gynecol. , 99 , 556 – 562 . 
81. Maggi , L. , Mastromarino , P. , Macchia , S. , Brigidi , P. , Pirovano , F. , Matteuzzi , D. , 
and Conte , U. ( 2000 ), Technological and biological evaluation of tablets containing different 
strains of lactobacilli for vaginal administration , Eur. J. Pharm. Biopharm. , 50 , 
389 – 395 . 
82. Bates , C. D. , Nicoll , A. E. , Mullen , A. B. , Mackenzie , F. , Thomson , A. J. , and Norman , 
J. E. ( 2003 ), Serum profi le of isosorbide mononitrate after vaginal administration in the 
third trimester , BJOG , 110 , 64 – 67 . 
83. Prista , L. N. , Alves , A. C. , and Morgado , R. ( 1996 ), Tecnologia Farmac e utica , 4th ed., 
Funda c a o Calouste Gulbenkian , Lisboa , pp 1585 – 1592 . 
84. Ozyazici , M. , Turgut , E. H. , Taner , M. S. , Koseoglu , K. , and Ertan , G. ( 2003 ), In - vitro 
evaluation and vaginal absorption of metronidazole suppositories in rabbits , J. Drug 
Target. , 11 , 177 – 185 . 
85. Bergren , M. S. , Battle , M. M. , Halstead , G. W. , and Theis , D. L. ( 1989 ), Investigation of 
the relationship between melting - related parameters and in vitro drug release from 
vaginal suppositories , J. Pharm. Biomed. Anal. , 7 , 549 – 561 . 
86. Kale , V. V. , Trivedi , R. V. , Wate , S. P. , and Bhusari , K. P. ( 2005 ), Development and evaluation 
of a suppository formulation containing lactobacillus and its application in vaginal 
diseases , Ann. N. Y. Acad. Sci. , 1056 , 359 – 365 . 
87. Mahaguna , V. , McDermott , J. M. , Zhang , F. , and Ochoa , F. ( 2004 ), Investigation of 
product quality between extemporaneously compounded progesterone vaginal 

862 VAGINAL DRUG DELIVERY 
suppositories and an approved progesterone vaginal gel , Drug. Dev. Ind. Pharm. , 30 , 
1069 – 1078 . 
88. Zaneveld , L. J. , Waller , D. P. , Ahmad , N. , Quigg , J. , Kaminski , J. , Nikurs , A. , and De Jonge , 
C. ( 2001 ), Properties of a new, long - lasting vaginal delivery system (LASRS) for contraceptive 
and antimicrobial agents , J. Androl. , 22 , 481 – 490 . 
89. Ladipo , O. A. , De Castro , M. P. , Filho , L. C. , Coutinho , E. , Waller , D. P. , Cone , F. , and 
Zaneveld , L. J. ( 2000 ), A new vaginal antimicrobial contraceptive formulation: Phase I 
clinical pilot studies , Contraception , 62 , 91 – 97 . 
90. Mandal , T. K. ( 2000 ), Swelling - controlled release system for the vaginal delivery of 
miconazole , Eur. J Pharm. Biopharm. , 50 , 337 – 343 . 
91. Merabet , J. , Thompson , D. , and Saul Levinson , R. ( 2005 ), Advancing vaginal drug delivery 
, Expert Opin. Drug. Deliv. , 2 , 769 – 777 . 
92. Seidman , L. S. , and Skokos , C. K. ( 2005 ), An evaluation of butoconazole nitrate 2% site 
release vaginal cream (Gynazole - 1) compared to fl uconazole 150 mg tablets (Difl ucan) 
in the time to relief of symptoms in patients with vulvovaginal candidiasis , Infect. Dis. 
Obstet. Gynecol. , 13 , 197 – 206 . 
93. Levinson , R. S. , Mitan , S. J. , Steinmetz , J. I. , Gattermeir , D. J. , Schumacher , R. J. , and 
Joffrion , J. L. ( 2005 ), An open - label, two - period, crossover study of the systemic bioavailability 
in healthy women of clindamycin phosphate from two vaginal cream formulations 
, Clin. Ther. , 27 , 1894 – 1900 . 
94. del Palacio , A. , Sanz , F. , Sanchez - Alor , G. , Garau , M. , Calvo , M. T. , Boncompte , E. , 
Alguero , M. , Pontes , C. , and Gomez de la Camara , A. ( 2000 ), Double - blind randomized 
dose - fi nding study in acute vulvovaginal candidosis. Comparison of fl utrimazole site - 
release cream (1, 2 and 4%) with placebo site - release vaginal cream , Mycoses , 43 , 
355 – 365 . 
95. Wichterle , O. , and Lim , D. ( 1960 ), Hydrophilic gels in biologic use , Nature , 185 , 117 . 
96. Justin - Temu , M. , Damian , F. , Kinget , R. , and Van Den Mooter , G. ( 2004 ), Intravaginal 
gels as drug delivery systems , J. Womens Health (Larchmt.) , 13 , 834 – 844 . 
97. das Neves , J. , and Bahia , M. F. ( 2006 ), Gels as vaginal drug delivery systems , Int. J. Pharm. , 
318 , 1 – 14 . 
98. Milani , M. , Molteni , B. , and Silvani , I. ( 2000 ), Effect on vaginal pH of a polycarbophil 
vaginal gel compared with an acidic douche in women with suspected bacterial vaginosis: 
A randomized, controlled study , Curr. Ther. Res. Clin. Exp. , 61 , 781 – 788 . 
99. Robinson , J. R. , and Bologna , W. J. ( 1994 ), Vaginal and reproductive system treatments 
using a bioadhesive polymer , J. Controlled Release , 28 , 87 – 94 . 
100. D ’ Cruz , O. J. , and Uckun , F. M. ( 2001 ), Gel - microemulsions as vaginal spermicides and 
intravaginal drug delivery vehicles , Contraception , 64 , 113 – 123 . 
101. Knuth , K. , Amiji , M. , and Robinson , J. R. ( 1993 ), Hydrogel delivery systems for vaginal 
and oral applications: Formulation and biological considerations , Adv. Drug Deliv. Rev. , 
11 , 137 – 167 . 
102. Venkatraman , S. , Davar , N. , Chester , A. , and Kleiner , L. ( 2005 ), An overview of controlled 
release systems , in Wise , D. L. , Ed., Handbook of Pharmaceutical Controlled 
Release Technology , Marcel Dekker , New York , pp 431 – 464 . 
103. Okada , H. , Yamazaki , I. , Ogawa , Y. , Hirai , S. , Yashiki , T. , and Mima , H. ( 1982 ), Vaginal 
absorption of a potent luteinizing hormone - releasing hormone analog (leuprolide) in 
rats I: Absorption by various routes and absorption enhancement , J. Pharm. Sci. , 71 , 
1367 – 1371 . 
104. Rosenberg , M. J. , Phillips , R. S. , and Holmes , M. D. ( 1991 ), Vaginal douching. Who and 
why ? J. Reprod. Med. , 36 , 753 – 758 . 

REFERENCES 863 
105. McClelland , R. S. , Lavreys , L. , Hassan , W. M. , MAndaliya , K. , Ndinya - Achola , J. O. , and 
Baeten , J. M. ( 2006 ), Vaginal washing and increased risk of HIV - 1 acquisition among 
African women: A 10 - year prospective study , AIDS , 20 , 269 – 273 . 
106. Myer , L. , Kuhn , L. , Stein , Z. A. , Wright , T. C. , Jr., and Denny , L. ( 2005 ), Intravaginal 
practices, bacterial vaginosis, and women ’ s susceptibility to HIV infection: Epidemiological 
evidence and biological mechanisms , Lancet Infect. Dis. , 5 , 786 – 794 . 
107. Rajamanoharan , S. , Low , N. , Jones , S. B. , and Pozniak , A. L. ( 1999 ), Bacterial vaginosis, 
ethnicity, and the use of genital cleaning agents: A case control study , Sex. Transm. Dis. , 
26 , 404 – 409 . 
108. Zhang , J. , Thomas , A. G. , and Leybovich , E. ( 1997 ), Vaginal douching and adverse health 
effects: A meta-analysis , Am. J. Public Health , 87 , 1207 – 1211 . 
109. Sam , A. P. ( 1992 ), Controlled release contraceptive devices: A status report , J. Controlled 
Release , 22 , 35 – 46 . 
110. Johansson , E. D. , and Sitruk - Ware , R. ( 2004 ), New delivery systems in contraception: 
Vaginal rings , Am. J. Obstet. Gynecol. , 190 , S54 – 59 . 
111. Timmer , C. J. , and Mulders , T. M. ( 2000 ), Pharmacokinetics of etonogestrel and ethinylestradiol 
released from a combined contraceptive vaginal ring , Clin. Pharmacokinet. , 39 , 
233 – 242 . 
112. Sitruk - Ware , R. ( 2005 ), Vaginal delivery of contraceptives , Expert Opin. Drug Deliv. , 2 , 
729 – 736 . 
113. Acarturk , F. , and Altug , N. ( 2001 ), In - vitro and in - vivo evaluation of a matrix - controlled 
bromocriptine mesilate - releasing vaginal ring , J. Pharm. Pharmacol. , 53 , 1721 – 1726 . 
114. Igarashi , M. , Iizuka , M. , Abe , Y. , and Ibuki , Y. ( 1998 ), Novel vaginal danazol ring therapy 
for pelvic endometriosis, in particular deeply infi ltrating endometriosis , Hum. Reprod. , 
13 , 1952 – 1956 . 
115. Woolfson , A. D. , Malcolm , R. K. , and Gallagher , R. J. ( 2003 ), Design of a silicone reservoir 
intravaginal ring for the delivery of oxybutynin , J. Controlled Release , 91 , 465 – 
476 . 
116. Wyatt , T. L. , Whaley , K. J. , Cone , R. A. , and Saltzman , W. M. ( 1998 ), Antigen - releasing 
polymer rings and microspheres stimulate mucosal immunity in the vagina , J. Controlled 
Release , 50 , 93 – 102 . 
117. Malcolm, R. K. , Woolfson, A. D. , Toner , C. F. , Morrow , R. J. , and McCullagh, S. D. ( 2005 ), 
Long - term, controlled release of the HIV microbicide TMC120 from silicone elastomer 
vaginal rings , J. Antimicrob. Chemother. , 56 , 954 – 956 . 
118. Dziuk , P. J. , and Cook , B. ( 1966 ), Passage of steroids through silicone rubber , Endocrinology 
, 78 , 208 – 211 . 
119. Mishell , D. R. , Jr. , Talas , M. , Parlow , A. F. , and Moyer , D. L. ( 1970 ), Contraception by 
means of a silastic vaginal ring impregnated with medroxyprogesterone acetate , Am. J. 
Obstet. Gynecol. , 107 , 100 – 107 . 
120. Landgren , B. M. , Johannisson , E. , Masironi , B. , and Diczfalusy , E. ( 1979 ), Pharmacokinetic 
and pharmacodynamic effects of small doses of norethisterone released from 
vaginal rings continuously during 90 days , Contraception , 19 , 253 – 271 . 
121. Mishell , D. R. , Jr. , Lumkin , M. , and Jackanicz , T. ( 1975 ), Initial clinical studies of 
intravaginal rings containing norethindrone and norgestrel , Contraception , 12 , 253 – 
260 . 
122. van Laarhoven , H. , Veurink , J. , Kruft , M. A. , and Vromans , H. ( 2004 ), Infl uence of spinline 
stress on release properties of a coaxial controlled release device based on EVA 
polymers , Pharm. Res. , 21 , 1811 – 1817 . 

864 VAGINAL DRUG DELIVERY 
123. van Laarhoven , J. A. , Kruft , M. A. , and Vromans , H. ( 2002 ), In vitro release properties 
of etonogestrel and ethinyl estradiol from a contraceptive vaginal ring , Int. J. Pharm. , 
232 , 163 – 173 . 
124. Vartiainen , J. , Wahlstrom , T. , and Nilsson , C. G. ( 1993 ), Effects and acceptability of a new 
17 beta - oestradiol - releasing vaginal ring in the treatment of postmenopausal complaints , 
Maturitas , 17 , 129 – 137 . 
125. Nash , H. A. , Brache , V. , Alvarez - Sanchez , F. , Jackanicz , T. M. , and Harmon , T. M. ( 1997 ), 
Estradiol delivery by vaginal rings: Potential for hormone replacement therapy , Maturitas 
, 26 , 27 – 33 . 
126. Saleh , S. I. , Khidr , S. H. , Ahmed , S. M. , Jackanicz , T. M. , and Nash , H. A. ( 2003 ), 
Estradiol - progesterone interaction during the preparation of vaginal rings , J. Pharm. 
Sci. , 92 , 258 – 265 . 
127. Metzger , A. P. , and Matlack , J. D. ( 1968 ), Comparative swelling behavior of various 
thermoplastic polymers , Polym. Eng. Sci. , 8 , 110 – 115 . 
128. Chokshi , R. , and Zia , H. ( 2004 ), Hot - melt extrusion technique: A review , Iran J. Pharm. 
Res. , 3 , 3 – 16 . 
129. Malcolm , K. , Woolfson , D. , Russell , J. , Tallon , P. , McAuley , L. , and Craig , D. ( 2003 ), Infl uence 
of silicone elastomer solubility and diffusivity on the in vitro release of drugs from 
intravaginal rings , J. Controlled Release , 90 , 217 – 225 . 
130. Lee , C. H. , Bhatt , P. P. , and Chien, Y. W. (1997), Effect of excipient on drug release and 
permeation from silicone - based barrier devices , Int. J. Pharm. , 43 , 283 – 290 . 
131. Garg , S. , Vermani , K. , Garg , A. , Anderson , R. A. , Rencher , W. B. , and Zaneveld , L. J. 
( 2005 ), Development and characterization of bioadhesive vaginal fi lms of sodium polystyrene 
sulfonate (PSS), a novel contraceptive antimicrobial agent , Pharm. Res. , 22 , 
584 – 595 . 
132. Yoo , J. W. , Dharmala , K. , and Lee , C. H. ( 2006 ), The physicodynamic properties of mucoadhesive 
polymeric fi lms developed as female controlled drug delivery system , Int. J. 
Pharm. , 309 , 139 – 145 . 
133. Mauck , C. K. , Baker , J. M. , Barr , S. P. , Abercrombie , T. J. , and Archer , D. F. ( 1997 ), A 
phase I comparative study of contraceptive vaginal fi lms containing benzalkonium chloride 
and nonoxynol - 9. Postcoital testing and colposcopy , Contraception , 56 , 89 – 96 . 
134. Chien , Y. W. , Oppermann , J. , Nicolova , B. , and Lambert , H. J. ( 1982 ), Medicated tampons: 
Intravaginal sustained administration of metronidazole and in vitro – in vivo relationships 
, J. Pharm. Sci. , 71 , 767 – 771 . 
135. Brzezinski , A. , Stern , T. , Arbel , R. , Rahav , G. , and Benita , S. ( 2004 ), Effi cacy of a novel 
pH - buffering tampon in preserving the acidic vaginal pH during menstruation , Int. J. 
Gynaecol. Obstet. , 85 , 298 – 300 . 
136. Bernstein , G. S. ( 1974 ), Conventional methods of contraception: Condom, diaphragm, 
and vaginal foam , Clin. Obstet. Gynecol. , 17 , 21 – 33 . 
137. Youssef , H. , Crofton , V. A. , Smith , S. C. , and Siemens , A. J. ( 1987 ), A clinical trial of Neo 
Sampoon vaginal tablets and Emko foam in Alexandria, Egypt , Contraception , 35 , 
101 – 110 . 
138. Bullough , V. L. , and Bullough , B. ( 1990 ), Contraception: A Guide to Birth Control 
Methods , Prometheus Books , Buffalo, NY . 
139. Smith , C. B. , Noble , V. , Bensch , R. , Ahlin , P. A. , Jacobson , J. A. , and Latham , R. H. ( 1982 ), 
Bacterial fl ora of the vagina during the menstrual cycle: Findings in users of tampons, 
napkins, and sea sponges , Ann. Intern. Med. , 96 , 948 – 951 . 
140. Kafka , D. , and Gold , R. B. ( 1983 ), Food and Drug Administration approves vaginal 
sponge , Fam. Plann. Perspect. , 15 , 146 – 148 . 

REFERENCES 865 
141. Kuyoh , M. A. , Toroitich - Ruto , C. , Grimes , D. A. , Schulz , K. F. , and Gallo , M. F. ( 2003 ), 
Sponge versus diaphragm for contraception: A Cochrane review , Contraception , 67 , 
15 – 18 . 
142. Bowers , R. , Ed. ( 2001 ), U. S. women are waiting for contraceptive sponge , Contracept. 
Technol. Update , 22 , 6 – 7 . 
143. Smart , J. D. ( 2005 ), The basics and underlying mechanisms of mucoadhesion , Adv. Drug 
Deliv. Rev. , 57 , 1556 – 1568 . 
144. Valenta , C. ( 2005 ), The use of mucoadhesive polymers in vaginal delivery , Adv. Drug 
Deliv. Rev. , 57 , 1692 – 1712 . 
145. Hosny , E. A. ( 1993 ), Polycarbophil as a controlled release matrix , Int. J. Pharm. , 98 , 
235 – 238 . 
146. Kast , C. E. , Valenta , C. , Leopold , M. , and Bernkop - Schnurch , A. ( 2002 ), Design and in 
vitro evaluation of a novel bioadhesive vaginal drug delivery system for clotrimazole , 
J. Controlled Release , 81 , 347 – 354 . 
147. Valenta , C. , Kast , C. E. , Harich , I. , and Bernkop - Schnurch , A. ( 2001 ), Development and 
in vitro evaluation of a mucoadhesive vaginal delivery system for progesterone , J. Controlled 
Release , 77 , 323 – 332 . 
148. Peppas , N. A. , Bures , P. , Leobandung , W. , and Ichikawa , H. ( 2000 ), Hydrogels in pharmaceutical 
formulations , Eur. J. Pharm. Biopharm. , 50 , 27 – 46 . 
149. Ruel - Gariepy , E. , and Leroux , J. C. ( 2004 ), In situ - forming hydrogels — Review of temperature 
- sensitive systems , Eur. J. Pharm. Biopharm. , 58 , 409 – 426 . 
150. Chang , J. Y. , Oh , Y. K. , Kong , H. S. , Kim , E. J. , Jang , D. D. , Nam , K. T. , and Kim , C. K. 
( 2002 ), Prolonged antifungal effects of clotrimazole - containing mucoadhesive thermosensitive 
gels on vaginitis , J. Controlled Release , 82 , 39 – 50 . 
151. Pavelic , Z. , Skalko - Basnet , N. , and Schubert , R. ( 2001 ), Liposomal gels for vaginal drug 
delivery , Int. J. Pharm. , 219 , 139 – 149 . 
152. Pavelic , Z. , Skalko - Basnet , N. , and Jalsenjak , I. ( 2004 ), Liposomal gel with chloramphenicol: 
Characterisation and in vitro release , Acta Pharm. , 54 , 319 – 330 . 
153. Pavelic , Z. , Skalko - Basnet , N. , and Jalsenjak , I. ( 2005 ), Characterisation and in vitro 
evaluation of bioadhesive liposome gels for local therapy of vaginitis , Int. J. Pharm. , 301 , 
140 – 148 . 
154. Pavelic , Z. , Skalko - Basnet , N. , Filipovic - Grcic , J. , Martinac , A. , and Jalsenjak , I. ( 2005 ), 
Development and in vitro evaluation of a liposomal vaginal delivery system for acyclovir 
, J. Controlled Release , 106 , 34 – 43 . 
155. Pavelic , Z. , Skalko - Basnet , N. , and Jalsenjak , I. ( 1999 ), Liposomes containing drugs for 
treatment of vaginal infections , Eur. J. Pharm. Sci. , 8 , 345 – 351 . 
156. Payne , N. I. , Timmins , P. , Ambrose , C. V. , Ward , M. D. , and Ridgway , F. ( 1986 ), Proliposomes: 
A novel solution to an old problem , J. Pharm. Sci. , 75 , 325 – 329 . 
157. Ning , M. Y. , Guo , Y. Z. , Pan , H. Z. , Yu , H. M. , and Gu , Z. W. ( 2005 ), Preparation and 
evaluation of proliposomes containing clotrimazole , Chem. Pharm. Bull. (Tokyo) , 53 , 
620 – 624 . 
158. Ning , M. , Guo , Y. , Pan , H. , Yu , H. , and Gu , Z. ( 2005 ), Niosomes with sorbitan 
monoester as a carrier for vaginal delivery of insulin: Studies in rats , Drug Deliv. , 12 , 
399 – 407 . 
159. Ning , M. , Guo , Y. , Pan , H. , Chen , X. , and Gu , Z. ( 2005 ), Preparation, in vitro and in vivo 
evaluation of liposomal/niosomal gel delivery systems for clotrimazole , Drug Dev. Ind. 
Pharm. , 31 , 375 – 383 . 
160. Akagi , T. , Kawamura , M. , Ueno , M. , Hiraishi , K. , Adachi , M. , Serizawa , T. , Akashi , M. , 
and Baba , M. ( 2003 ), Mucosal immunization with inactivated HIV - 1 - capturing 

866 VAGINAL DRUG DELIVERY 
nanospheres induces a signifi cant HIV1 - specifi c vaginal antibody response in mice , 
J. Med. Virol. , 69 , 163 – 172 . 
161. Bonucci , E. , Ballanti , P. , Ramires , P. A. , Richardson , J. L. , and Benedetti , L. M. ( 1995 ), 
Prevention of ovariectomy osteopenia in rats after vaginal administration of Hyaff 11 
microspheres containing salmon calcitonin , Calcif. Tissue Int. , 56 , 274 – 279 . 
162. Bilensoy , E. , Rouf , M. A. , Vural , I. , Sen , M. , and Hincal , A. A. ( 2006 ), Mucoadhesive, 
thermosensitive, prolonged - release vaginal gel for clotrimazole:beta - cyclodextrin 
complex , AAPS PharmSciTech , 7 , E38 . 
163. Francois , M. , Snoeckx , E. , Putteman , P. , Wouters , F. , De Proost , E. , Delaet , U. , Peeters , 
J. , and Brewster , M. E. ( 2003 ), A mucoadhesive, cyclodextrin - based vaginal cream formulation 
of itraconazole , AAPS PharmSci , 5 , E5 . 
164. Ahmed , M. O. , El - Gibaly , I. , and Ahmed , S. M. ( 1998 ), Effect of cyclodextrins on the 
physicochemical properties and antimycotic activity of clotrimazole , Int. J. Pharm. , 171 , 
111 – 121 . 
165. Shihata , A. ( 2004 ), New FDA - approved woman - controlled, latex - free barrier contraceptive 
device “ Fem Cap , ” Int. Congr. Ser. , 1271 , 303 – 306 . 
166. Lee , C. H. , Bagdon , R. E. , Bhatt , P. P. , and Chien , Y. W. ( 1997 ), Development of silicone - 
based barrier devices for controlled delivery of spermicidal agents , J. Controlled Release , 
44 , 43 – 53 . 
167. McCarron , P. A. , Donnelly , R. F. , Gilmore , B. F. , Woolfson , A. D. , McClelland , R. , 
Zawislak , A. , and Price , J. H. ( 2004 ), Phototoxicity of 5 - aminolevulinic acid in the HeLa 
cell line as an indicative measure of photodynamic effect after topical administration to 
gynecological lesions of intraepithelial form , Pharm. Res. , 21 , 1871 – 1879 . 
168. Woolfson , A. D. , McCafferty , D. F. , McCarron , P. A. , and Price , J. H. ( 1995 ), A bioadhesive 
patch cervical drug delivery system for the administration of 5 - fl uorouracil to cervical 
tissue , J. Controlled Release , 35 , 49 – 58 . 
169. Mazouni , C. , Provensal , M. , Menard , J. P. , Heckenroth , H. , Guidicelli , B. , Gamerre , M. , 
and Bretelle , F. ( 2006 ), Utilisation du dispositif vaginal Propess dans le d e clenchement 
du travail: Effi cacit e et innocuit e , Gynecol. Obstet. Fertil. , 34 , 489 – 492 . 
170. Vollebregt , A. , van ’ t Hof , D. B. , and Exalto , N. ( 2002 ), Prepidil compared to Propess for 
cervical ripening , Eur. J. Obstet. Gynecol. Reprod. Biol. , 104 , 116 – 119 . 
171. Tedajo , G. M. , Bouttier , S. , Grossiord , J. L. , Marty , J. P. , Seiller , M. , and Fourniat , J. ( 2002 ), 
In vitro microbicidal activity of W/O/W multiple emulsion for vaginal administration , 
Int. J. Antimicrob. Agents , 20 , 50 – 56 . 
172. Kaushal , G. , Trombetta , L. , Ochs , R. S. , and Shao , J. ( 2006 ), Delivery of TEM beta - 
lactamase by gene - transformed Lactococcus lactis subsp. lactis through cervical cell 
monolayer , Int. J. Pharm. , 313 , 29 – 35 . 
173. Yao , X. Y. , Yuan , M. M. , and Li , D. J. ( 2006 ), Mucosal inoculation of Lactobacillus 
expressing hCGbeta induces an anti - hCGbeta antibody response in mice of different 
strains , Methods , 38 , 124 – 132 . 
174. Vail , J. G. , Cohen , J. A. , and Kelly , K. L. ( 2004 ), Improving topical microbicide applicators 
for use in resource - poor settings , Am. J. Public Health , 94 , 1089 – 1092 . 
175. Costa , P. , and Sousa Lobo , J. M. ( 2001 ), Modeling and comparison of dissolution profi les , 
Eur. J. Pharm. Sci. , 13 , 123 – 133 . 
176. Siewert , M. , Dressman , J. , Brown , C. K. , and Shah , V. P. ( 2003 ), FIP/AAPS guidelines to 
dissolution/in vitro release testing of novel/special dosage forms , AAPS PharmSciTech , 
4 , E7 . 
177. Karasulu , H. Y. , Hilmioglu , S. , Metin , D. Y. , and Guneri , T. ( 2004 ), Effi cacy of a new 
ketoconazole bioadhesive vaginal tablet on Candida albicans , Farmaco , 59 , 163 – 167 . 

REFERENCES 867 
178. Ondracek , J. , Stoll , B. , and Krifter , R. ( 1988 ), New basket dissolution method for vaginal 
suppositories , Acta Pharm. Technol. , 34 , 169 – 171 . 
179. Chun , I. K. , and Chien , Y. W. ( 1993 ), Transmucosal delivery of methionine enkephalin. 
I: Solution stability and kinetics of degradation in various rabbit mucosa extracts , 
J. Pharm. Sci. , 82 , 373 – 378 . 
180. Bechgaard , E. , Riis , K. J. , and Jorgensen , L. ( 1994 ), The development of an Ussing 
chamber for isolated human vaginal mucosa, and the viability of the in vitro system , Int. 
J. Pharm. , 106 , 237 – 242 . 
181. van der Bijl , P. , van Eyk , A. D. , and Thompson , I. O. ( 1998 ), Penetration of human vaginal 
and buccal mucosa by 4.4 - kd and 12 - kd fl uorescein - isothiocyanate - labeled dextrans , 
Oral Surg. Oral. Med. Oral Pathol. Oral Radiol. Endod. , 85 , 686 – 691 . 
182. van der Bijl , P. , van Eyk , A. D. , and Thompson , I. O. ( 1998 ), Permeation of 17beta - 
estradiol through human vaginal and buccal mucosa , Oral Surg. Oral Med. Oral Pathol. 
Oral Radiol. Endod. , 85 , 393 – 398 . 
183. van der Bijl , P. , van Eyk , A. D. , van Wyk , C. W. , and Stander , I. A. ( 2001 ), Diffusion of 
reduced arecoline and arecaidine through human vaginal and buccal mucosa , J. Oral 
Pathol. Med. , 30 , 200 – 205 . 
184. Hsu , C. C. , Park , J. Y. , Ho , N. F. , Higuchi , W. I. , and Fox , J. L. ( 1983 ), Topical vaginal drug. 
delivery I: Effect of the estrous cycle on vaginal membrane permeability and diffusivity 
of vidarabine in mice , J. Pharm. Sci. , 72 , 674 – 680 . 
185. Okada , H. , Yashiki , T. , and Mima , H. ( 1983 ), Vaginal absorption of a potent luteinizing 
hormone - releasing hormone analogue (leuprolide) in rats III: Effect of estrous cycle on 
vaginal absorption of hydrophilic model compounds , J. Pharm. Sci. , 72 , 173 – 176 . 
186. Garg , S. , Anderson , R. A. , Chany , C. J. , 2nd, Waller , D. P. , Diao , X. H. , Vermani , K. , and 
Zaneveld , L. J. ( 2001 ), Properties of a new acid - buffering bioadhesive vaginal formulation 
(ACIDFORM) , Contraception , 64 , 67 – 75 . 
187. Castle , P. E. , Karp , D. A. , Zeitlin , L. , Garcia - Moreno , E. B. , Moench , T. R. , Whaley , K. J. , 
and Cone , R. A. ( 2002 ), Human monoclonal antibody stability and activity at vaginal 
pH , J. Reprod. Immunol. , 56 , 61 – 76 . 
188. Owen , D. H. , Peters , J. J. , and Katz , D. F. ( 2000 ), Rheological properties of contraceptive 
gels , Contraception , 62 , 321 – 326 . 
189. Chang , J. Y. , Oh , Y. K. , Choi , H. G. , Kim , Y. B. , and Kim , C. K. ( 2002 ), Rheological evaluation 
of thermosensitive and mucoadhesive vaginal gels in physiological conditions , Int. 
J. Pharm. , 241 , 155 – 163 . 
190. Thorgeirsdottir , T. O. , Kjoniksen , A. L. , Knudsen , K. D. , Kristmundsdottir , T. , and 
Nystrom , B. ( 2005 ), Viscoelastic and structural properties of pharmaceutical hydrogels 
containing monocaprin , Eur. J. Pharm. Biopharm. , 59 , 333 – 342 . 
191. Thorgeirsdottir , T. O. , Thormar , H. , and Kristmundsdottir , T. ( 2006 ), Viscoelastic properties 
of a virucidal cream containing the monoglyceride monocaprin: Effects of formulation 
variables: A technical note , AAPS PharmSciTech , 7 , E44 . 
192. Owen , D. H. , Peters , J. J. , Katz , and D. F. ( 2001 ), Comparison of the rheological 
properties of Advantage - S and Replens , Contraception , 64 , 393 – 396 . 
193. Madsen , F. , Eberth , K. , and Smart , J. D. ( 1998 ), A rheological examination of the mucoadhesive/
mucus interaction: The effect of mucoadhesive type and concentration , 
J. Controlled Release , 50 , 167 – 178 . 
194. Owen , D. H. , Peters , J. J. , Lavine , M. L. , and Katz , D. F. ( 2003 ), Effect of temperature 
and pH on contraceptive gel viscosity , Contraception , 67 , 57 – 64 . 
195. Owen , D. H. , and Katz , D. F. ( 2005 ), A review of the physical and chemical properties 
of human semen and the formulation of a semen simulant , J. Androl. , 26 , 459 – 469 . 

868 VAGINAL DRUG DELIVERY 
196. Jones , D. S. , Irwin , C. R. , Woolfson , A. D. , Djokic , J. , and Adams , V. ( 1999 ), Physicochemical 
characterization and preliminary in vivo effi cacy of bioadhesive, semisolid 
formulations containing fl urbiprofen for the treatment of gingivitis , J. Pharm. Sci. , 88 , 
592 – 598 . 
197. Jones , D. S. , Lawlor , M. S. , and Woolfson , A. D. ( 2002 ), Examination of the fl ow rheological 
and textural properties of polymer gels composed of poly(methylvinylether - 
co - maleic anhydride) and poly(vinylpyrrolidone): Rheological and mathematical 
interpretation of textural parameters , J. Pharm. Sci. , 91 , 2090 – 2101 . 
198. Vermani , K. , Garg , S. , and Zaneveld , L. J. ( 2002 ), Assemblies for in vitro measurement 
of bioadhesive strength and retention characteristics in simulated vaginal environment , 
Drug Dev. Ind. Pharm. , 28 , 1133 – 1146 . 
199. Tamburic , S. , and Craig , D. Q. M. ( 1997 ), A comparision of different in vitro methods 
for measuring mucoadhesive performance , Eur. J. Pharm. Biopharm. , 44 , 159 – 167 . 
200. El - Kamel , A. , Sokar , M. , Naggar , V. , and Al Gamal , S. ( 2002 ), Chitosan and sodium 
alginate - based bioadhesive vaginal tablets , AAPS PharmSci , 4 , E44 . 
201. Barnhart , K. T. , Stolpen , A. , Pretorius , E. S. , and Malamud , D. ( 2001 ), Distribution of a 
spermicide containing Nonoxynol - 9 in the vaginal canal and the upper female reproductive 
tract , Hum. Reprod. , 16 , 1151 – 1154 . 
202. Barnhart , K. T. , Pretorius , E. S. , Shera , D. M. , Shabbout , M. , and Shaunik , A. ( 2006 ), The 
optimal analysis of MRI data to quantify the distribution of a microbicide , Contraception 
, 73 , 82 – 87 . 
203. Kieweg , S. L. , Geonnotti , A. R. , and Katz , D. F. ( 2004 ), Gravity - induced coating 
fl ows of vaginal gel formulations: In vitro experimental analysis , J. Pharm. Sci. , 93 , 
2941 – 2952 . 
204. Geonnotti , A. R. , Peters , J. J. , and Katz , D. F. ( 2005 ), Erosion of microbicide formulation 
coating layers: Effects of contact and shearing with vaginal fl uid or semen , J. Pharm. 
Sci. , 94 , 1705 – 1712 . 
205. Ceschel , G. C. , Maffei , P. , Lombardi Borgia , S. , Ronchi , C. , and Rossi , S. ( 2001 ), Development 
of a mucoadhesive dosage form for vaginal administration , Drug Dev. Ind. Pharm. , 
27 , 541 – 547 . 
206. Barnhart , K. , Pretorius , E. S. , Stolpen , A. , and Malamud , D. ( 2001 ), Distribution of 
topical medication in the human vagina as imaged by magnetic resonance imaging , Fertil. 
Steril. , 76 , 189 – 195 . 
207. Brown , J. , Hooper , G. , Kenyon , C. J. , Haines , S. , Burt , J. , Humphries , J. M. , Newman , 
S. P. , Davis , S. S. , Sparrow , R. A. , and Wilding , I. R. ( 1997 ), Spreading and retention of 
vaginal formulations in postmenopausal women as assessed by gamma scintigraphy , 
Pharm. Res. , 14 , 1073 – 1078 . 
208. Chatterton , B. E. , Penglis , S. , Kovacs , J. C. , Presnell , B. , and Hunt , B. ( 2004 ), Retention 
and distribution of two 99mTc - DTPA labelled vaginal dosage forms , Int. J. Pharm. , 271 , 
137 – 143 . 
209. Henderson , M. H. , Peters , J. J. , Walmer , D. K. , Couchman , G. M. , and Katz , D. F. ( 2005 ), 
Optical instrument for measurement of vaginal coating thickness by drug delivery formulations 
, Rev. Sci. Instrum. , 76 , 034302 . 
210. Braun , K. E. , Boyer , J. D. , Henderson , M. H. , Katz , D. F. , and Wax , A. ( 2006 ), Label - free 
measurement of microbicidal gel thickness using low - coherence interferometry , J. 
Biomed. Opt. , 11 , 020504.1 – 020504.3 . 
211. Brache , V. , Cohen , J. A. , Cochon , L. , and Alvarez , F. ( 2006 ), Evaluating the clinical safety 
of three vaginal applicators: A pilot study conducted in the Dominican Republic , Contraception 
, 73 , 72 – 77 . 

REFERENCES 869 
212. Bagga , R. , Raghuvanshi , P. , Gopalan , S. , Das , S. K. , Baweja , R. , Suri , S. , Malhotra , D. , Khare , 
S. , and Talwar , G. P. ( 2006 ), A polyherbal vaginal pessary with spermicidal and antimicrobial 
action: Evaluation of its safety , Trans. R. Soc. Trop. Med. Hyg. , 100 , 1164 – 1167 . 
213. D ’ Cruz , O. J. , and Uckun , F. M. ( 2001 ), Lack of adverse effects on fertility of female 
CD - 1 mice exposed to repetitive intravaginal gel - microemulsion formulation of a dual - 
function anti - HIV agent: Aryl phosphate derivative of bromo - methoxy - zidovudine 
(compound WHI - 07) , J. Appl. Toxicol. , 21 , 317 – 322 . 
214. Krebs , F. C. , Miller , S. R. , Catalone , B. J. , Fichorova , R. , Anderson , D. , Malamud , D. , 
Howett , M. K. , and Wigdahl , B. ( 2002 ), Comparative in vitro sensitivities of human 
immune cell lines, vaginal and cervical epithelial cell lines, and primary cells to candidate 
microbicides nonoxynol 9, C31G, and sodium dodecyl sulfate , Antimicrob. Agents Chemother. 
, 46 , 2292 – 2298 . 
215. Dezzutti , C. S. , James , V. N. , Ramos , A. , Sullivan , S. T. , Siddig , A. , Bush , T. J. , Grohskopf , 
L. A. , Paxton , L. , Subbarao , S. , and Hart , C. E. ( 2004 ), In vitro comparison of topical 
microbicides for prevention of human immunodefi ciency virus type 1 transmission , 
Antimicrob. Agents Chemother. , 48 , 3834 – 3844 . 
216. Ayehunie , S. , Cannon , C. , Lamore , S. , Kubilus , J. , Anderson , D. J. , Pudney , J. , and 
Klausner , M. ( 2006 ), Organotypic human vaginal - ectocervical tissue model for irritation 
studies of spermicides, microbicides, and feminine - care products , Toxicol. In Vitro , 20 , 
689 – 698 . 
217. Eckstein , P. , Jackson , M. C. , Millman , N. , and Sobrero , A. J. ( 1969 ), Comparison of vaginal 
tolerance tests of spermicidal preparations in rabbits and monkeys , J. Reprod. Fertil. , 20 , 
85 – 93 . 
218. D ’ Cruz , O. J. , and Uckun , F. M. ( 2001 ), Intravaginal toxicity studies of a gel - 
microemulsion formulation of spermicidal vanadocenes in rabbits , Toxicol. Appl. Pharmacol. 
, 170 , 104 – 112 . 
219. Patton , D. L. , Sweeney , Y. C. , Tsai , C. C. , and Hillier , S. L. ( 2004 ), Macaca fascicularis vs. 
Macaca nemestrina as a model for topical microbicide safety studies , J. Med. Primatol. , 
33 , 105 – 108 . 
220. Gray , J. E. , Weaver , R. N. , Lohrberg , S. M. , and Larsen , E. R. ( 1984 ), Comparative 
responses of vaginal mucosa to chronic pyrimidinone - induced irritation , Toxicol. Pathol. , 
12 , 228 – 234 . 
221. D ’ Cruz , O. J. , Erbeck , D. , and Uckun , F. M. ( 2005 ), A study of the potential of the pig 
as a model for the vaginal irritancy of benzalkonium chloride in comparison to the 
nonirritant microbicide PHI - 443 and the spermicide vanadocene dithiocarbamate , 
Toxicol. Pathol. , 33 , 465 – 476 . 
222. D ’ Cruz , O. J. , Waurzyniak , B. , Yiv , S. H. , and Uckun , F. M. ( 2000 ), Evaluation of subchronic 
(13 - week) and reproductive toxicity potential of intravaginal gel - microemulsion 
formulation of a dual - function phenyl phosphate derivative of bromo - methoxy zidovudine 
(compound whi - 07) in B(6)C(3)F(1) mice , J. Appl. Toxicol. , 20 , 319 – 325 . 
223. Chvapil , M. , Droegemueller , W. , Owen , J. A. , Eskelson , C. D. , and Betts , K. ( 1980 ), 
Studies of nonoxynol - 9. I. The effect on the vaginas of rabbits and rats , Fertil. Steril. , 33 , 
445 – 450 . 
224. CPMP/EMEA ( 1997 ), Replacement of Animal Studies by In Vitro Models , CPMP/EMEA , 
London . 
225. Adriaens , E. , and Remon , J. P. ( 1999 ), Gastropods as an evaluation tool for screening 
the irritating potency of absorption enhancers and drugs , Pharm. Res. , 16 , 1240 – 1244 . 
226. Dhondt , M. M. , Adriaens , E. , Roey , J. V. , and Remon , J. P. ( 2005 ), The evaluation of the 
local tolerance of vaginal formulations containing dapivirine using the Slug Mucosal 

870 VAGINAL DRUG DELIVERY 
Irritation test and the rabbit vaginal irritation test , Eur. J. Pharm. Biopharm. , 60 , 
419 – 425 . 
227. Mauck , C. K. , Weiner , D. H. , Ballagh , S. A. , Creinin , M. D. , Archer , D. F. , Schwartz , J. L. , 
Pymar , H. C. , Lai , J. J. , Rencher , W. F. , and Callahan , M. M. ( 2004 ), Single and multiple 
exposure tolerance study of polystyrene sulfonate gel: A phase I safety and colposcopy 
study , Contraception , 70 , 77 – 83 . 
228. Mauck, C. K. , Baker , J. M. , Birnkrant, D. B. , Rowe , P. J. , and Gabelnick, H. L. (2000), The 
use of colposcopy in assessing vaginal irritation in research , AIDS , 14 , 2221 – 2227 . 
229. WHO/CONRAD ( 2004 ), Manual for the Standardization of Colposcopy for the Evaluation 
of Vaginal products, Update 2004 , CONRAD/WHO, Geneva. 
230. van der Laak , J. A. , de Bie , L. M. , de Leeuw , H. , de Wilde , P. C. , and Hanselaar , A. G. 
( 2002 ), The effect of Replens on vaginal cytology in the treatment of postmenopausal 
atrophy: Cytomorphology versus computerised cytometry , J. Clin. Pathol. , 55 , 446 – 451 . 
231. Rosen , A. D. , and Rosen , T. ( 1999 ), Study of condom integrity after brief exposure to 
over - the - counter vaginal preparations , South Med. J. , 92 , 305 – 307 . 
232. Haineault , C. , Gourde , P. , Perron , S. , Desormeaux , A. , Piret , J. , Omar , R. F. , Tremblay , 
R. R. , and Bergeron , M. G. ( 2003 ), Thermoreversible gel formulation containing sodium 
lauryl sulfate as a potential contraceptive device , Biol. Reprod. , 69 , 687 – 694 . 
233. Ansbacher , R. ( 2004 ), Intravaginal medications , Fertil Steril , 82 , 1474 (Letter). 
234. Stone , A. ( 2002 ), Microbicides: A new approach to preventing HIV and other sexually 
transmitted infections , Nat. Rev. Drug Discov. , 1 , 977 – 985 . 
235. Rowe , P. M. ( 1995 ), Research into topical microbicides against STDs , Lancet , 345 , 
1231 . 
236. Watts , C. , and Vickerman , P. ( 2001 ), The impact of microbicides on HIV and STD transmission: 
Model projections , AIDS , 15 ( Suppl. 1 ), S43 – 44 . 
237. Lederman , M. M. , Veazey , R. S. , Offord , R. , Mosier , D. E. , Dufour , J. , Mefford , M. , Piatak , 
M. , Jr. , Lifson , J. D. , Salkowitz , J. R. , Rodriguez , B. , Blauvelt , A. , and Hartley , O. ( 2004 ), 
Prevention of vaginal SHIV transmission in rhesus macaques through inhibition of 
CCR5 , Science , 306 , 485 – 487 . 
238. Yedery , R. D. , and Reddy , K. V. ( 2005 ), Antimicrobial peptides as microbicidal contraceptives: 
Prophecies for prophylactics — A mini review , Eur. J. Contracept. Reprod. Health 
Care , 10 , 32 – 42 . 
239. Veazey , R. S. , Shattock , R. J. , Pope , M. , Kirijan , J. C. , Jones , J. , Hu , Q. , Ketas , T. , Marx , 
P. A. , Klasse , P. J. , Burton , D. R. , and Moore , J. P. ( 2003 ), Prevention of virus transmission 
to macaque monkeys by a vaginally applied monoclonal antibody to HIV - 1 gp120 , Nat. 
Med. , 9 , 343 – 346 . 
240. Veazey , R. S. , Klasse , P. J. , Schader , S. M. , Hu , Q. , Ketas , T. J. , Lu , M. , Marx , P. A. , Dufour , 
J. , Colonno , R. J. , Shattock , R. J. , Springer , M. S. , and Moore , J. P. ( 2005 ), Protection of 
macaques from vaginal SHIV challenge by vaginally delivered inhibitors of virus - cell 
fusion , Nature , 438 , 99 – 102 . 
241. Neurath , A. R. , Strick , N. , Li , Y. Y. , and Debnath , A. K. ( 2005 ), Punica granatum (pomegranate) 
juice provides an HIV - 1 entry inhibitor and candidate topical microbicide , Ann. 
N. Y. Acad. Sci. , 1056 , 311 – 327 . 
242. Liu , S. , Lu , H. , Neurath , A. R. , and Jiang , S. ( 2005 ), Combination of candidate microbicides 
cellulose acetate 1,2 - benzenedicarboxylate and UC781 has synergistic and complementary 
effects against human immunodefi ciency virus type 1 infection , Antimicrob. 
Agents Chemother. , 49 , 1830 – 1836 . 
243. Roddy , R. E. , Zekeng , L. , Ryan , K. A. , Tamoufe , U. , Weir , S. S. , and Wong , E. L. ( 1998 ), 
A controlled trial of nonoxynol 9 fi lm to reduce male - to - female transmission of sexually 
transmitted diseases , N. Engl. J. Med. , 339 , 504 – 510 . 

REFERENCES 871 
244. International Working Group On Vaginal Microbicides ( 1996 ), Recommendations for 
the development of vaginal microbicides , AIDS , 10 , 1 – 6 . 
245. Mauck , C. , Rosenberg , Z. , and Van Damme , L. ( 2001 ), Recommendations for the clinical 
development of topical microbicides: An update , AIDS , 15 , 857 – 868 . 
246. Garg , S. , Kandarapu , R. , Vermani , K. , Tambwekar , K. R. , Garg , A. , Waller , D. P. , and 
Zaneveld , L. J. ( 2003 ), Development pharmaceutics of microbicide formulations. Part I: 
Preformulation considerations and challenges , AIDS Patient Care STDS , 17 , 17 – 32 . 
247. Garg , S. , Tambwekar , K. R. , Vermani , K. , Kandarapu , R. , Garg , A. , Waller , D. P. , and 
Zaneveld , L. J. ( 2003 ), Development pharmaceutics of microbicide formulations. Part II: 
Formulation, evaluation, and challenges , AIDS Patient Care STDS , 17 , 377 – 399 . 
248. Bax , R. , Douville , K. , McCormick , D. , Rosenberg , M. , Higgins , J. , and Bowden , M. ( 2002 ), 
Microbicides — Evaluating multiple formulations of C31G , Contraception , 66 , 365 – 368 . 
249. D ’ Cruz , O. J. , Samuel , P. , and Uckun , F. M. ( 2005 ), Conceival, a novel noncontraceptive 
vaginal vehicle for lipophilic microbicides , AAPS PharmSciTech , 6 , E56 – 64 . 
250. Chang , T. L. , Chang , C. H. , Simpson , D. A. , Xu , Q. , Martin , P. K. , Lagenaur , L. A. , 
Schoolnik , G. K. , Ho , D. D. , Hillier , S. L. , Holodniy , M. , Lewicki , J. A. , and Lee , P. P. ( 2003 ), 
Inhibition of HIV infectivity by a natural human isolate of Lactobacillus jensenii engineered 
to express functional two - domain CD4 , Proc. Natl. Acad. Sci. U.S.A. , 100 , 
11672 – 11677 . 
251. Watson, M. C. , Grimshaw , J. M. , Bond, C. M. , Mollison, J. , and Ludbrook, A. (2002), Oral 
versus intravaginal imidazole and triazole anti - fungal agents for the treatment of uncomplicated 
vulvovaginal candidiasis (thrush): A systematic review , BJOG , 109 , 85 – 95 . 
252. Sobel, J. D. (1997), Vaginitis , N. Engl. J. Med. , 337 , 1896 – 1903 . 
253. SaiRam , M. , Ilavazhagan , G. , Sharma , S. K. , Dhanraj , S. A. , Suresh , B. , Parida , M. M. , 
Jana , A. M. , Devendra , K. , and Selvamurthy , W. ( 2000 ), Anti - microbial activity of a new 
vaginal contraceptive NIM - 76 from neem oil (Azadirachta indica) , J. Ethnopharmacol. , 
71 , 377 – 382 . 
254. Pina - Vaz , C. , Goncalves Rodrigues , A. , Pinto , E. , Costa - de - Oliveira , S. , Tavares , C. , 
Salgueiro , L. , Cavaleiro , C. , Goncalves , M. J. , and Martinez - de - Oliveira , J. ( 2004 ), 
Antifungal activity of Thymus oils and their major compounds , J. Eur. Acad. Dermatol. 
Venereol. , 18 , 73 – 78 . 
255. Brown , D. , Henzl , M. R. , and Kaufman , R. H. ( 1999 ), Butoconazole nitrate 2% for 
vulvovaginal candidiasis. New, single - dose vaginal cream formulation vs. seven - day 
treatment with miconazole nitrate. Gynazole 1 Study Group , J. Reprod. Med. , 44 , 
933 – 938 . 
256. Faro , S. , and Skokos , C. K. ( 2005 ), The effi cacy and safety of a single dose of Clindesse 
vaginal cream versus a seven - dose regimen of Cleocin vaginal cream in patients with 
bacterial vaginosis , Infect. Dis. Obstet. Gynecol. , 13 , 155 – 160 . 
257. Upmalis , D. H. , Cone , F. L. , Lamia , C. A. , Reisman , H. , Rodriguez - Gomez , G. , Gilderman , 
L. , and Bradley , L. ( 2000 ), Single - dose miconazole nitrate vaginal ovule in the treatment 
of vulvovaginal candidiasis: Two single - blind, controlled studies versus miconazole 
nitrate 100 mg cream for 7 days , J. Womens Health Gend. Based Med. , 9 , 421 – 429 . 
258. Mikamo , H. , Kawazoe , K. , Izumi , K. , Watanabe , K. , Ueno , K. , and Tamaya , T. ( 1997 ), 
Comparative study on vaginal or oral treatment of bacterial vaginosis , Chemotherapy , 
43 , 60 – 68 . 
259. Cunningham , F. E. , Kraus , D. M. , Brubaker , L. , and Fischer , J. H. ( 1994 ), Pharmacokinetics 
of intravaginal metronidazole gel , J. Clin. Pharmacol. , 34 , 1060 – 1065 . 
260. Borin , M. T. ( 1990 ), Systemic absorption of clindamycin following intravaginal application 
of clindamycin phosphate 1% cream , J. Clin. Pharmacol. , 30 , 33 – 38 . 

872 VAGINAL DRUG DELIVERY 
261. Czeizel , A. E. , Kazy , Z. , and Vargha , P. ( 2003 ), A population - based case - control teratological 
study of vaginal econazole treatment during pregnancy , Eur. J. Obstet. Gynecol. 
Reprod. Biol. , 111 , 135 – 140 . 
262. Polatti , F. , Rampino , M. , Magnani , P. , and Mascarucci , P. ( 2006 ), Vaginal pH - lowering 
effect of locally applied vitamin C in subjects with high vaginal pH , Gynecol. Endocrinol. 
, 22 , 230 – 234 . 
263. Amaral , E. , Perdigao , A. , Souza , M. H. , Mauck , C. , Waller , D. , Zaneveld , L. , and Faundes , 
A. ( 2006 ), Vaginal safety after use of a bioadhesive, acid - buffering, microbicidal contraceptive 
gel (ACIDFORM) and a 2% nonoxynol - 9 product , Contraception , 73 , 542 – 
547 . 
264. Decena , D. C. , Co , J. T. , Manalastas , R. M. , Jr. , Palaypayon , E. P. , Padolina , C. S. , Sison , 
J. M. , Dancel , L. A. , and Lelis , M. A. ( 2006 ), Metronidazole with Lactacyd vaginal gel in 
bacterial vaginosis , J. Obstet. Gynaecol. Res. , 32 , 243 – 251 . 
265. Fiorilli , A. , Molteni , B. , and Milani , M. ( 2005 ), Successful treatment of bacterial vaginosis 
with a policarbophil - carbopol acidic vaginal gel: Results from a randomised double - 
blind, placebo - controlled trial , Eur. J. Obstet. Gynecol. Reprod. Biol. , 120 , 202 – 205 . 
266. Levine , H. , and Watson , N. ( 2000 ), Comparison of the pharmacokinetics of crinone 8% 
administered vaginally versus Prometrium administered orally in postmenopausal 
women , Fertil. Steril. , 73 , 516 – 521 . 
267. van den Heuvel , M. W. , van Bragt , A. J. , Alnabawy , A. K. , and Kaptein , M. C. ( 2005 ), 
Comparison of ethinylestradiol pharmacokinetics in three hormonal contraceptive formulations: 
The vaginal ring, the transdermal patch and an oral contraceptive , Contraception 
, 72 , 168 – 174 . 
268. Fraser , I. S. , Lacarra , M. , Mishell , D. R. , Alvarez , F. , Brache , V. , Lahteenmaki , P. , Elomaa , 
K. , Weisberg , E. , and Nash , H. A. ( 2000 ), Vaginal epithelial surface appearances in 
women using vaginal rings for contraception , Contraception , 61 , 131 – 138 . 
269. Bjarnad o ttir , R. I. ( 2003 ), Update on contraceptive vaginal rings , Rev. Gyn. Pract. , 3 , 
156 – 159 . 
270. Roumen , F. J. , Apter , D. , Mulders , T. M. , and Dieben , T. O. ( 2001 ), Effi cacy, tolerability 
and acceptability of a novel contraceptive vaginal ring releasing etonogestrel and ethinyl 
oestradiol , Hum. Reprod. , 16 , 469 – 475 . 
271. Oddsson , K. , Leifels - Fischer , B. , de Melo , N. R. , Wiel - Masson , D. , Benedetto , C. , 
Verhoeven , C. H. , and Dieben , T. O. ( 2005 ), Effi cacy and safety of a contraceptive vaginal 
ring (NuvaRing) compared with a combined oral contraceptive: A 1 - year randomized 
trial , Contraception , 71 , 176 – 182 . 
272. Dieben , T. O. , Roumen , F. J. , and Apter , D. ( 2002 ), Effi cacy, cycle control, and user acceptability 
of a novel combined contraceptive vaginal ring , Obstet. Gynecol. , 100 , 585 – 593 . 
273. Sivin , I. , Mishell , D. R. , Jr. , Alvarez , F. , Brache , V. , Elomaa , K. , Lahteenmaki , P. , Massai , 
R. , Miranda , P. , Croxatto , H. , Dean , C. , Small , M. , Nash , H. , and Jackanicz , T. M. ( 2005 ), 
Contraceptive vaginal rings releasing Nestorone and ethinylestradiol: A 1 - year dose - 
fi nding trial , Contraception , 71 , 122 – 129 . 
274. Rad , M. , Kluft , C. , Menard , J. , Burggraaf , J. , de Kam , M. L. , Meijer , P. , Sivin , I. , and 
Sitruk - Ware , R. L. ( 2006 ), Comparative effects of a contraceptive vaginal ring delivering 
a nonandrogenic progestin and continuous ethinyl estradiol and a combined oral contraceptive 
containing levonorgestrel on hemostasis variables , Am. J. Obstet. Gynecol. , 
195 , 72 – 77 . 
275. Devoto , L. , Fuentes , A. , Palomino , A. , Espinoza , A. , Kohen , P. , Ranta , S. , and von Hertzen , 
H. ( 2005 ), Pharmacokinetics and endometrial tissue levels of levonorgestrel after administration 
of a single 1.5 - mg dose by the oral and vaginal route , Fertil. Steril. , 84 , 46 – 51 . 

REFERENCES 873 
276. Mor , E. , Saadat , P. , Kives , S. , White , E. , Reid , R. L. , Paulson , R. J. , and Stanczyk , F. Z. 
( 2005 ), Comparison of vaginal and oral administration of emergency contraception , 
Fertil. Steril. , 84 , 40 – 45 . 
277. Kives , S. , Hahn , P. M. , White , E. , Stanczyk , F. Z. , and Reid , R. L. ( 2005 ), Bioavailability 
of the Yuzpe and levonorgestrel regimens of emergency contraception: Vaginal vs. oral 
administration , Contraception , 71 , 197 – 201 . 
278. Yoo , J. W. , and Lee , C. H. ( 2006 ), Drug delivery systems for hormone therapy , J. Controlled 
Release , 112 , 1 – 14 . 
279. Beral , V. ( 2003 ), Breast cancer and hormone - replacement therapy in the Million Women 
Study , Lancet , 362 , 419 – 427 . 
280. Cardozo , L. , Bachmann , G. , McClish , D. , Fonda , D. , and Birgerson , L. ( 1998 ), Meta - 
analysis of estrogen therapy in the management of urogenital atrophy in postmenopausal 
women: Second report of the Hormones and Urogenital Therapy Committee , 
Obstet. Gynecol. , 92 , 722 – 727 . 
281. Crandall , C. ( 2002 ), Vaginal estrogen preparations: A review of safety and effi cacy for 
vaginal atrophy . J. Womens Health (Larchmt.) , 11 , 857 – 877 . 
282. Ballagh , S. A. ( 2004 ), Vaginal rings for menopausal symptom relief , Drugs Aging , 21 , 
757 – 766 . 
283. Henriksson , L. , Stjernquist , M. , Boquist , L. , Cedergren , I. , and Selinus , I. ( 1996 ), A one - 
year multicenter study of effi cacy and safety of a continuous, low - dose, estradiol - 
releasing vaginal ring (Estring) in postmenopausal women with symptoms and signs of 
urogenital aging , Am. J. Obstet. Gynecol. , 174 , 85 – 92 . 
284. Eriksen , B. ( 1999 ), A randomized, open, parallel - group study on the preventive effect 
of an estradiol - releasing vaginal ring (Estring) on recurrent urinary tract infections in 
postmenopausal women , Am. J. Obstet. Gynecol. , 180 , 1072 – 1079 . 
285. Tavaniotou , A. , Smitz , J. , Bourgain , C. , and Devroey , P. ( 2000 ), Comparison between 
different routes of progesterone administration as luteal phase support in infertility 
treatments , Hum. Reprod. Update , 6 , 139 – 148 . 
286. Kleinstein , J. ( 2005 ), Effi cacy and tolerability of vaginal progesterone capsules (Utrogest 
200) compared with progesterone gel (Crinone 8%) for luteal phase support during 
assisted reproduction , Fertil. Steril. , 83 , 1641 – 1649 . 
287. Zegers - Hochschild , F. , Balmaceda , J. P. , Fabres , C. , Alam , V. , Mackenna , A. , Fernandez , 
E. , Pacheco , I. M. , Sepulveda , M. S. , Chen , S. , Borrero , C. , and Borges , E. ( 2000 ), Prospective 
randomized trial to evaluate the effi cacy of a vaginal ring releasing progesterone 
for IVF and oocyte donation , Hum. Reprod. , 15 , 2093 – 2097 . 
288. Gupta , G. ( 2005 ), Microbicidal spermicide or spermicidal microbicide ? Eur. J. Contracept. 
Reprod. Health Care , 10 , 212 – 218 . 
289. Castle , P. E. , Whaley , K. J. , Hoen , T. E. , Moench , T. R. , and Cone , R. A. ( 1997 ), Contraceptive 
effect of sperm - agglutinating monoclonal antibodies in rabbits , Biol. Reprod. , 
56 , 153 – 159 . 
290. Raymond , E. G. , Chen , P. L. , and Luoto , J. ( 2004 ), Contraceptive effectiveness and 
safety of fi ve nonoxynol - 9 spermicides: A randomized trial , Obstet. Gynecol. , 103 , 
430 – 439 . 
291. Lee , C. H. , Bagdon , R. , and Chien , Y. W. ( 1996 ), Comparative in vitro spermicidal activity 
of chelating agents and synergistic effect with nonoxynol - 9 on human sperm functionality 
, J. Pharm. Sci. , 85 , 91 – 95 . 
292. Zavos , P. M. , Correa , J. R. , and Zarmakoupis - Zavos , P. N. ( 1998 ), Assessment of a tablet 
drug delivery system incorporating nonoxynol - 9 coprecipitated with polyvinylpyrrolidone 
in preventing the onset of pregnancy in rabbits , Fertil. Steril. , 69 , 768 – 773 . 

874 VAGINAL DRUG DELIVERY 
293. Fowler , P. T. , Doncel , G. F. , Bummer , P. M. , and Digenis , G. A. ( 2003 ), Coprecipitation of 
nonoxynol - 9 with polyvinylpyrrolidone to decrease vaginal irritation potential while 
maintaining spermicidal potency , AAPS PharmSciTech , 4 , E30 . 
294. Zavos , P. M. , and Correa , J. R. ( 1997 ), Vaginal delivery of new formulations of 
nonoxynol - 9 coprecipitated with polyvinylpyrrolidone in rabbits. Comparisons between 
two formulation delivery systems , Contraception , 56 , 123 – 127 . 
295. Gagne , N. , Cormier , H. , Omar , R. F. , Desormeaux , A. , Gourde , P. , Tremblay , M. J. , Juhasz , 
J. , Beauchamp , D. , Rioux , J. E. , and Bergeron , M. G. ( 1999 ), Protective effect of a thermoreversible 
gel against the toxicity of nonoxynol - 9 , Sex Transm. Dis. , 26 , 177 – 183 . 
296. El - Gizawy , S. A. , and Aglan , N. I. ( 2003 ), Formulation and evaluation of metronidazole 
acid gel for vaginal contraception , J. Pharm. Pharmacol. , 55 , 903 – 909 . 
297. Owen , D. H. , Dunmire , E. N. , Plenys , A. M. , and Katz , D. F. ( 1999 ), Factors infl uencing 
nonoxynol - 9 permeation and bioactivity in cervical mucus , J. Controlled Release , 60 , 
23 – 34 . 
298. Saxena , B. B. , Singh , M. , Gospin , R. M. , Chu , C. C. , and Ledger , W. J. ( 2004 ), Effi cacy of 
nonhormonal vaginal contraceptives from a hydrogel delivery system , Contraception , 
70 , 213 – 219 . 
299. Saltzman , W. M. , and Tena , L. B. ( 1991 ), Spermicide permeation through biocompatible 
polymers , Contraception , 43 , 497 – 505 . 
300. Gauger , L. J. , and Curet , L. B. ( 1991 ), Comparative effi cacy of intravaginal prostaglandin 
E2 in the gel and suppository forms for cervical ripening, DICP Ann. Pharmacother. , 25 , 
456 – 460 . 
301. Chyu , J. K. , and Strassner , H. T. ( 1997 ), Prostaglandin E2 for cervical ripening: 
A randomized comparison of Cervidil versus Prepidil , Am. J. Obstet. Gynecol. , 177 , 
606 – 611 . 
302. Carbonell , J. L. , Rodriguez , J. , Delgado , E. , Sanchez , C. , Vargas , F. , Valera , L. , Mari , J. , 
Valero , F. , Salvador , I. , and Llorente , M. ( 2004 ), Vaginal misoprostol 800 microg every 
12 h for second - trimester abortion , Contraception , 70 , 55 – 60 . 
303. Carbonell , J. L. , Rodriguez , J. , Velazco , A. , Tanda , R. , Sanchez , C. , Barambio , S. , Chami , 
S. , Valero , F. , Mari , J. , de Vargas , F. , and Salvador , I. ( 2003 ), Oral and vaginal misoprostol 
800 microg every 8 h for early abortion , Contraception , 67 , 457 – 462 . 
304. Carbonell , J. L. , Rodriguez , J. , Aragon , S. , Velazco , A. , Tanda , R. , Sanchez , C. , Barambio , 
S. , Chami , S. , and Valero , F. ( 2001 ), Vaginal misoprostol 1000 microg for early abortion , 
Contraception , 63 , 131 – 136 . 
305. Zieman , M. , Fong , S. K. , Benowitz , N. L. , Banskter , D. , and Darney , P. D. ( 1997 ), Absorption 
kinetics of misoprostol with oral or vaginal administration , Obstet. Gynecol. , 90 , 
88 – 92 . 
306. Richardson , J. L. , and Illum , L. ( 1992 ), (D) Routes of delivery: Case studies: (8) 
the vaginal route of peptide and protein drug delivery , Adv. Drug Deliv. Rev. , 8 , 
341 – 366 . 
307. Richardson , J. L. , Ramires , P. A. , Miglietta , M. R. , Rochira , M. , Bacelle , L. , Callegaro , L. , 
and Benedetti , L. ( 1995 ), Novel vaginal delivery systems for calcitonin: I. Evaluation of 
HYAFF/calcitonin microspheres in rats , Int. J. Pharm. , 115 , 9 – 15 . 
308. Rochira , M. , Miglietta , M. R. , Richardson , J. L. , Ferrari , L. , Beccaro , M. , and Benedetti , 
L. ( 1996 ), Novel vaginal delivery systems for calcitonin: II. Preparation and characterization 
of HYAFF ® microspheres containing calcitonin , Int. J. Pharm. , 144 , 19 – 26 . 
309. Sherwood , J. K. , Zeitlin , L. , Whaley , K. J. , Cone , R. A. , and Saltzman , M. ( 1996 ), Controlled 
release of antibodies for long - term topical passive immunoprotection of female 
mice against genital herpes , Nat. Biotechnol. , 14 , 468 – 471 . 

REFERENCES 875 
310. Saltzman , W. M. , Sherwood , J. K. , Adams , D. R. , Castle , P. , and Haller , P. ( 2000 ), Long - 
term vaginal antibody delivery: Delivery systems and biodistribution , Biotechnol. 
Bioeng. , 67 , 253 – 264 . 
311. Saltzman , W. M. , and Langer , R. ( 1989 ), Transport rates of proteins in porous materials 
with known microgeometry , Biophys. J. , 55 , 163 – 171 . 
312. Morimoto , K. , Takeeda , T. , Nakamoto , Y. , and Morisaka , K. ( 1982 ), Effective vaginal 
absorption of insulin in diabetic rats and rabbits using polyacrylic acid aqueous gel bases , 
Int. J. Pharm. , 12 , 107 – 111 . 
313. Degim , Z. , Degim , T. , Acarturk , F. , Erdogan , D. , Ozogul , C. , and Koksal , M. ( 2005 ), Rectal 
and vaginal administration of insulin - chitosan formulations: An experimental study in 
rabbits , J. Drug Target. , 13 , 563 – 572 . 
314. Bogers , W. M. , Bergmeier , L. A. , Ma , J. , Oostermeijer , H. , Wang , Y. , Kelly , C. G. , Ten 
Haaft , P. , Singh , M. , Heeney , J. L. , and Lehner , T. ( 2004 ), A novel HIV - CCR5 receptor 
vaccine strategy in the control of mucosal SIV/HIV infection , AIDS , 18 , 25 – 36 . 
315. Livingston , J. B. , Lu , S. , Robinson , H. , and Anderson , D. J. ( 1998 ), Immunization of the 
female genital tract with a DNA - based vaccine , Infect. Immun. , 66 , 322 – 329 . 
316. Kozlowski , P. A. , Cu - Uvin , S. , Neutra , M. R. , and Flanigan , T. P. ( 1997 ), Comparison of 
the oral, rectal, and vaginal immunization routes for induction of antibodies in rectal 
and genital tract secretions of women , Infect. Immun. , 65 , 1387 – 1394 . 
317. Kozlowski , P. A. , Cu - Uvin , S. , Neutra , M. R. , and Flanigan , T. P. ( 1999 ), Mucosal vaccination 
strategies for women , J. Infect. Dis. , 179 ( Suppl. 3 ), S493 – 498 . 
318. Wassen , L. , Schon , K. , Holmgren , J. , Jertborn , M. , and Lycke , N. ( 1996 ), Local intravaginal 
vaccination of the female genital tract , Scand. J. Immunol. , 44 , 408 – 414 . 
319. Kozlowski , P. A. , Williams , S. B. , Lynch , R. M. , Flanigan , T. P. , Patterson , R. R. , Cu - Uvin , 
S. , and Neutra , M. R. ( 2002 ), Differential induction of mucosal and systemic antibody 
responses in women after nasal, rectal, or vaginal immunization: Infl uence of the menstrual 
cycle , J. Immunol. , 169 , 566 – 574 . 
320. Han , I. K. , Kim , Y. B. , Kang , H. S. , Sul , D. , Jung , W. W. , Cho , H. J. , and Oh , Y. K. ( 2006 ), 
Thermosensitive and mucoadhesive delivery systems of mucosal vaccines , Methods , 38 , 
106 – 111 . 
321. Oh , Y. K. , Park , J. S. , Yoon , H. , and Kim , C. K. ( 2003 ), Enhanced mucosal and systemic 
immune responses to a vaginal vaccine coadministered with RANTES - expressing 
plasmid DNA using in situ - gelling mucoadhesive delivery system , Vaccine , 21 , 
1980 – 1988 . 
322. Shen , H. , Goldberg , E. , and Saltzman , W. M. ( 2003 ), Gene expression and mucosal 
immune responses after vaginal DNA immunization in mice using a controlled delivery 
matrix , J. Controlled Release , 86 , 339 – 348 . 
323. Luo , D. , Woodrow - Mumford , K. , Belcheva , N. , and Saltzman , W. M. ( 1999 ), Controlled 
DNA delivery systems , Pharm. Res. , 16 , 1300 – 1308 . 
324. Oggioni , M. R. , Medaglini , D. , Maggi , T. , and Pozzi , G. ( 1999 ), Engineering the gram - 
positive cell surface for construction of bacterial vaccine vectors , Methods , 19 , 
163 – 173 . 
325. Medaglini , D. , Oggioni , M. R. , and Pozzi , G. ( 1998 ), Vaginal immunization with recombinant 
gram - positive bacteria , Am. J. Reprod. Immunol. , 39 , 199 – 208 . 
326. Singh , S. R. , Hulett , K. , Pillai , S. R. , Dennis , V. A. , Oh , M. K. , and Scissum - Gunn , K. 
( 2006 ), Mucosal immunization with recombinant MOMP genetically linked with modi- 
fi ed cholera toxin confers protection against Chlamydia trachomatis infection , Vaccine , 
24 , 1213 – 1224 . 

876 VAGINAL DRUG DELIVERY 
327. O ’ Hagan , D. T. , Rafferty , D. , Wharton , S. , and Illum , L. ( 1993 ), Intravaginal immunization 
in sheep using a bioadhesive microsphere antigen delivery system , Vaccine , 11 , 660 – 
664 . 
328. Syed , T. A. , Qureshi , Z. A. , Ahmad , S. A. , and Ali , S. M. ( 2000 ), Management of intravaginal 
warts in women with 5 - fl uorouracil (1%) in vaginal hydrophilic gel: A placebo - 
controlled double - blind study , Int. J. STD AIDS , 11 , 371 – 374 . 
329. Maiman , M. , Watts , D. H. , Andersen , J. , Clax , P. , Merino , M. , and Kendall , M. A. ( 1999 ), 
Vaginal 5 - fl uorouracil for high - grade cervical dysplasia in human immunodefi ciency 
virus infection: A randomized trial , Obstet. Gynecol. , 94 , 954 – 961 . 
330. Darwish , A. M. , Hafez , E. , El - Gebali , I. , and Hassan , S. B. ( 2005 ), Evaluation of a novel 
vaginal bromocriptine mesylate formulation: A pilot study . Fertil. Steril. , 83 , 1053 – 1055 . 
331. Motta , T. , de Vincentiis , S. , Marchini , M. , Colombo , N. , and D ’ Alberton , A. ( 1996 ), 
Vaginal cabergoline in the treatment of hyperprolactinemic patients intolerant to oral 
dopaminergics , Fertil. Steril. , 65 , 440 – 442 . 
332. Stray - Pedersen , B. , Bergan , T. , Hafstad , A. , Normann , E. , Grogaard , J. , and Vangdal , M. 
( 1999 ), Vaginal disinfection with chlorhexidine during childbirth , Int. J. Antimicrob. 
Agents , 12 , 245 – 251 . 
333. Garcia - Lopez , P. , Coll , M. , Cervera , E. , Reyes-Vermot, L. , Torres , M. A. , Abrego-Perez, 
G. , Hernandez - Pajaro , A. I. , Castaneda - HernAndez , G. , Mohar - Betancourt , A. , and 
Meneses , A. ( 2006 ), The systemic absorption of etoposide after intravaginal administration 
in patients with cervical intraepithelial lesions associated with human papillomavirus 
infection , Pharm. Res. , 23 , 378 – 383 . 
334. Diakomanolis , E. , Haidopoulos , D. , and Stefanidis , K. ( 2002 ), Treatment of high - grade 
vaginal intraepithelial neoplasia with imiquimod cream , N. Engl. J. Med. , 347 , 374 
(Letter). 
335. Abramov , Y. , Nadjari , M. , Weinstein , D. , Ben - Shachar , I. , Plotkin , V. , and Ezra , Y. ( 2000 ), 
Indomethacin for preterm labor: A randomized comparison of vaginal and rectal - oral 
routes , Obstet. Gynecol. , 95 , 482 – 486 . 
336. Costello , M. F. , Steigrad , S. , and Collet , A. ( 2005 ), A prospective, randomised, single - 
blinded, controlled trial comparing two topical anaesthetic modalities for the application 
of a tenaculum to the cervix , J. Obstet. Gynaecol. , 25 , 781 – 785 . 
337. Ostrop , N. J. , Lamb , J. , and Reid , G. ( 1998 ), Intravaginal morphine: An alternative route 
of administration , Pharmacotherapy , 18 , 863 – 865 . 
338. Schroder , A. , Levin , R. M. , Kogan , B. A. , Das , A. K. , Kay , F. , and Mahashabde , A. ( 2000 ), 
Absorption of oxybutynin from vaginal inserts: Drug blood levels and the response of 
the rabbit bladder , Urology , 56 , 1063 – 1067 . 
339. Smith , R. C. , and Berge , M. ( 1985 ), Systemic absorption after intravaginal propranolol , 
Lancet , 1 , 112 (Letter). 
340. Sher , G. , and Fisch , J. D. ( 2002 ), Effect of vaginal sildenafi l on the outcome of in vitro 
fertilization (IVF) after multiple IVF failures attributed to poor endometrial development 
, Fertil. Steril. , 78 , 1073 – 1076 . 
341. Lin , H. , Huang , E. Y. , Chang , H. Y. , and ChangChien , C. C. ( 2005 ), Therapeutic effect of 
topical applications of trichloroacetic acid for vaginal intraepithelial neoplasia after 
hysterectomy , Jpn. J. Clin. Oncol. , 35 , 651 – 654 . 
342. Nappi , R. E. , Liekens , G. , and Brandenburg , U. ( 2006 ), Attitudes, perceptions and knowledge 
about the vagina: The International Vagina Dialogue Survey , Contraception , 73 , 
493 – 500 . 
343. Hammett , T. M. , Mason , T. H. , Joanis , C. L. , Foster , S. E. , Harmon , P. , Robles , R. R. , Finlinson 
, H. A. , Feudo , R. , Vining - Bethea , S. , Jeter , G. , Mayer , K. H. , Doherty - Iddings , P. , 
and Seage , G. R. , 3rd ( 2000 ), Acceptability of formulations and application methods for 

REFERENCES 877 
vaginal microbicides among drug - involved women: Results of product trials in three 
cities , Sex Transm. Dis. , 27 , 119 – 126 . 
344. Mantell , J. E. , Myer , L. , Carballo - Dieguez , A. , Stein , Z. , Ramjee , G. , Morar , N. S. , and 
Harrison , P. F. ( 2005 ), Microbicide acceptability research: Current approaches and future 
directions , Soc. Sci. Med. , 60 , 319 – 330 . 
345. Raymond , E. , Alvarado , G. , Ledesma , L. , Diaz , S. , Bassol , S. , Morales , E. , Fernandez , V. , 
and Carlos , G. ( 1999 ), Acceptability of two spermicides in fi ve countries , Contraception , 
60 , 45 – 50 . 
346. Hardy , E. , de Padua , K. S. , Hebling , E. M. , Osis , M. J. , and Zaneveld , L. J. ( 2003 ), Women ’ s 
preferences for vaginal antimicrobial contraceptives. V: Attitudes of Brazilian women 
to the insertion of vaginal products , Contraception , 67 , 391 – 395 . 
347. Bentley , M. E. , Fullem , A. M. , Tolley , E. E. , Kelly , C. W. , Jogelkar , N. , Srirak , N. , 
Mwafulirwa , L. , Khumalo - Sakutukwa , G. , and Celentano , D. D. ( 2004 ), Acceptability of 
a microbicide among women and their partners in a 4 - country phase I trial , Am. J. Public 
Health , 94 , 1159 – 1164 . 
348. Braunstein , S. , and van de Wijgert , J. ( 2005 ), Preferences and practices related to vaginal 
lubrication: Implications for microbicide acceptability and clinical testing , J. Womens 
Health (Larchmt.) , 14 , 424 – 433 . 
349. Aubeny , E. , Colau , J. C. , and Nandeuil , A. ( 2000 ), Local spermicidal contraception: A 
comparative study of the acceptability and safety of a new pharmaceutical formulation 
of benzalkonium chloride, the vaginal capsule, with a reference formulation, the pessary , 
Eur. J. Contracept. Reprod. Health Care , 5 , 61 – 67 . 
350. Coggins , C. , Elias , C. J. , Atisook , R. , Bassett , M. T. , Ettiegnene - Traore , V. , Ghys , P. D. , 
Jenkins - Woelk , L. , Thongkrajai , E. , and VanDevanter , N. L. ( 1998 ), Women ’ s preferences 
regarding the formulation of over - the - counter vaginal spermicides , AIDS , 12 , 1389 – 
1391 . 
351. Hardy , E. , Jimenez , A. L. , de Padua , K. S. , and Zaneveld , L. J. ( 1998 ), Women ’ s preferences 
for vaginal antimicrobial contraceptives. III. Choice of a formulation, applicator, 
and packaging , Contraception , 58 , 245 – 249 . 
352. Novak , A. , de la Loge , C. , Abetz , L. , and van der Meulen , E. A. ( 2003 ), The combined 
contraceptive vaginal ring, NuvaRing: An international study of user acceptability , Contraception 
, 67 , 187 – 194 . 
353. Roumen , F. J. , and Dieben , T. O. ( 1999 ), Clinical acceptability of an ethylene - vinyl - 
acetate nonmedicated vaginal ring , Contraception , 59 , 59 – 62 . 
354. Hardy , E. , de Padua , K. S. , Jimenez , A. L. , and Zaneveld , L. J. ( 1998 ), Women ’ s preferences 
for vaginal antimicrobial contraceptives. II. Preferred characteristics according to 
women ’ s age and socioeconomic status , Contraception , 58 , 239 – 244 . 
355. Rothen - Weinhold , A. , Dahn , M. , and Gurny , R. ( 2000 ), Formulation and technology 
aspects of controlled drug delivery in animals , Pharm. Sci. Technol. Today , 3 , 222 – 231 . 
356. Robinson , T. J. ( 1965 ), Use of progestagen - impregnated sponges inserted intravaginally 
or subcutaneously for the control of the oestrous cycle in the sheep , Nature , 206 , 
39 – 41 . 
357. Kim , S. , Kengaku , K. , Tanaka , T. , and Kamomae , H. ( 2004 ), The therapeutic effects of a 
progesterone - releasing intravaginal device (PRID) with attached estradiol capsule on 
ovarian quiescence and cystic ovarian disease in postpartum dairy cows , J. Reprod. Dev. , 
50 , 341 – 348 . 
358. Rathbone , M. J. , Macmillan , K. L. , Bunt , C. R. , and Burggraaf , S. ( 1997 ), Conceptual and 
commercially available intravaginal veterinary drug delivery systems , Adv. Drug Deliv. 
Rev. , 28 , 363 – 392 . 

878 VAGINAL DRUG DELIVERY 
359. Rathbone , M. J. , Macmillan , K. L. , Inskeep , K. , Burggraaf , S. , and Bunt , C. R. ( 1998 ), 
Fertility regulation in cattle , J. Controlled Release , 54 , 117 – 148 . 
360. Okura , N. , Yamagishi , N. , Naito , Y. , Kanno , K. , and Koiwa , M. ( 2004 ), Technical note: 
Vaginal absorption of 1,25(OH)2D3 in cattle , J. Dairy Sci. , 87 , 2416 – 2419 . 
361. Otero , M. C. , Morelli , L. , and Nader - Macias , M. E. ( 2006 ), Probiotic properties of vaginal 
lactic acid bacteria to prevent metritis in cattle , Lett. Appl. Microbiol. , 43 , 91 – 97 . 
362. Gavini , E. , Sanna , V. , Juliano , C. , Bonferoni , M. C. , and Giunchedi , P. ( 2002 ), Mucoadhesive 
vaginal tablets as veterinary delivery system for the controlled release of an 
antimicrobial drug, acrifl avine , AAPS PharmSciTech , 3 , E20 . 
363. Romano , J. E. , and Benech , A. ( 1996 ), Effect of service and vaginal - cervical anesthesia 
on estrus duration in dairy goats , Theriogenology , 45 , 691 – 696 . 
364. Loehr , B. I. , Rankin , R. , Pontarollo , R. , King , T. , Willson , P. , Babiuk , L. A. , and van 
Drunen Littel - van den Hurk , S. ( 2001 ), Suppository - mediated DNA immunization 
induces mucosal immunity against bovine herpesvirus - 1 in cattle , Virology , 289 , 
327 – 333 . 
365. Grimmett , J. B. , Hanlon , D. W. , Duirs , G. F. , and Jochle , W. ( 2002 ), A new intra - vaginal 
progesterone - releasing device (Cue - Mare ™ ) for controlling the estrous cycle in mares , 
Theriogenology , 58 , 585 – 587 . 
366. Cross , P. S. , Kunnemeyer , R. , Bunt , C. R. , Carnegie , D. A. , and Rathbone , M. J. ( 2004 ), 
Control, communication and monitoring of intravaginal drug delivery in dairy cows , Int. 
J. Pharm. , 282 , 35 – 44 . 

TABLET PRODUCTION 
SECTION 6


881 
6.1 
PHARMACEUTICAL 
PREFORMULATION: 
PHYSICOCHEMICAL PROPERTIES OF 
EXCIPIENTS AND POWDERS AND 
TABLET CHARACTERIZATION 
Beom - Jin Lee 
Kangwon National University, Chuncheon, Korea 
Contents 
6.1.1 Introduction 
6.1.2 Selection of Pharmaceutical Excipients 
6.1.2.1 Defi nitions and Goals 
6.1.2.2 Types of Pharmaceutical Excipients 
6.1.2.3 Characteristics of Pharmaceutical Excipients 
6.1.2.4 Selection Guideline of Pharmaceutical Excipients in Tablet Formulation 
6.1.3 Drug – Excipient Compatibility 
6.1.3.1 Experimental Studies for Drug – Excipient Compatibility 
6.1.3.2 Analytical Methods for Drug – Excipient Compatibility 
6.1.3.3 Reaction Types and Stabilization Guidelines 
6.1.4 Powder Characteristics 
6.1.4.1 Crystal Form and Habit 
6.1.4.2 Particle Size Distribution 
6.1.4.3 Flow Characteristics 
6.1.4.4 Density and Bulkiness 
6.1.4.5 Hygroscopicity 
6.1.4.6 Mixing 
6.1.4.7 Particle Size Reduction (Micronization and Milling) 
6.1.4.8 Compaction (Compressibility) 
6.1.4.9 Surface Area and Other Properties 
6.1.5 Tablet Characterization 
6.1.5.1 Disintegration 
6.1.5.2 Dissolution 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

882 PHARMACEUTICAL PREFORMULATION 
6.1.5.3 Weight Variation 
6.1.5.4 Hardness or Breaking Strength 
6.1.5.5 Friability 
6.1.5.6 Content Uniformity 
6.1.5.7 Tablet Thickness 
6.1.5.8 Tablet Shape and Size 
References 
6.1.1 INTRODUCTION 
Tablet is a major category of solid dosage forms which are widely used worldwide. 
Extensive information is required to prepare tablets with good quality at high standards. 
Based on preformulation studies, the optimal dosage forms are generally 
decided. When given orally, the solid dosage form tablet undergoes in vitro disintegration 
and dissolution followed by absorption through the gastrointestinal tract 
(GI). The in vivo biodistribution of drug which enters the systemic circulation then 
occurrs (Figure 1 ). 
In this section, general preformulation approaches for tablet production are 
described (Figure 2 ). The physicochemical properties of drug and excipients, which 
are crucial factors at the beginning stages, are presented. The types and functions 
of excipients used for tablet formulation and drug – excipient incompatibility are also 
discussed. Because tablets are prepared by compression of the drug with powdered 
excipients, the physicochemical properties of excipients and their multiple functions 
under regulatory standards are very important. To prepare a drug substance for the 
fi nal dosage forms, pharmaceutical excipients should be added. The guidelines for 
selection of excipients are also given, based on the type and function of excipient, 
drug – excipient compatibility, type of tablet, and manufacturing parameters. Drug – 
excipient incompatibility is a very important issue before and after tablet prepara- 
FIGURE 1 In vitro and in vivo pathways of drug which enters systemic circulation. 
Preformulation 
Formulation 
(dosage forms) 
Release/dissolution Absorption 
Distribution 
(blood concentration) 
Metabolism 
Elimination 
Receptor sites 
(efficacy) 
In vitro In vivo

tion. According to global International Conferce on Harmonisation (ICH) guidelines, 
we should cautiously choose the excipients in tablet formulations because the quality 
and sources are variable by supplier and by batch. 
In general, a tablet is prepared by mixing, milling, and compression the drug – 
excipient mixtures using a tablet machine. Therefore, powder characteristics of drug 
and excipients, including particle size, fl ow characteristics, bulk density, hygroscopicity, 
mixing and milling, and compaction, are extensively discussed. If these physical 
properties are not fully understood, tablets with good quality at high standards are 
often impossible to produce. 
The prepared tablet must be validated according to regulatory guidelines. In 
general, disintegration, dissolution, friability, hardness, and weight are characterized 
for quality validation of fi nished tablet. This biopharmaceutical preformulation 
information for tablet production will guide us to design the optimal tablet very 
effi ciently in the laboratory and industrial companies. 
6.1.2 SELECTION OF PHARMACEUTICAL EXCIPIENTS 
6.1.2.1 Defi nitions and Goals 
A tablet contains active ingredients as well as other substances known as excipients, 
which have specifi c functions. The types and functions of various excipients which 
are incorporated into tablet formulations are discussed in many textbooks [1 – 3] . 
A pharmaceutical excipient is defi ned as an inactive ingredient or any component 
other than the active ingredient added intentionally to the medicinal formulation 
or everything in the formulation except the active drug. Pharmaceutical excipients 
are also called additives, pharmaceutical ingredients, or inactive pharmaceutical 
ingredients. There are many reasons for selecting and adding these pharmaceutical 
excipients in formulations. In the preparation of various dosage forms, it is essential 
to combine pharmaceutical excipients with model drugs as adjuvants to prepare the 
FIGURE 2 General preformulation approaches for tablet production. 
Physicochemical properties of excipients Physicochemical properties of drug 
Manufacturing process 
Tablet production 
Tablet validation 
Drug–excipient compatibility 
SELECTION OF PHARMACEUTICAL EXCIPIENTS 883

884 PHARMACEUTICAL PREFORMULATION 
solid dosage forms, mainly tablets. The pharmaceutical excipients make the drug 
into the fi nal dosage forms. Physicochemical properties such as solubility, stability, 
metabolism, and even bioavailability of drugs can be varied by the pharmaceutical 
excipients [4 – 6] . Figure 3 shows the correlation and functions of drugs combined 
with pharmaceutical excipients in dosage from designs. Pharmaceutical excipients 
are regarded as key ingredients not only to decide optimal dosage forms but also 
to change the physicochemical and biological parameters of drugs. With an aid of 
pharmaceutical excipients, drug effi cacy can be maintained. Changes of other types 
of dosage forms for different routes of administration are also achieved. The excipients 
can also function for the preparation of dosage formulation during the manufacturing 
processes. Patient compliance and modifi ed releases of drugs can also be 
achieved if the excipients are properly applied. The colorant makes the tablet distinguishable 
after the coating process. 
However, utilization of these pharmaceutical excipients is limited by the regulatory 
guidelines to be satisfi ed in the dosage formulations [3, 7] . In general, the regulatory 
guidelines require the following conditions for the use of excipients in the 
dosage formulations: (a) no harmful or toxicological effect and listed GRAS (generally 
recognized as safe), (b) good stability with no drug – excipient incompatibility 
and by any impurities in the excipients, (c) no interference in quality validation and 
analytical tests, (d) satisfaction of regulatory issues and guidelines in all countries 
where the product is to be marketed, (e) no instability with primary packing materials, 
(f) ease of accessibility, distribution, and economical cost, (g) satisfaction for 
environmental issues, (h) be physiologically inert, (i) be physically and chemically 
compatible with the active substance and the other excipients in the formulation, 
and (j) no unacceptable microbiological burden. 
The Handbook of Pharmaceutical Excipients [1] contains some details of functional 
tests carried out on a wide range of excipients. The excipients all have pharmacopeial 
monographs, but it is important to understand that compliance with a 
monograph does not indicate equivalence between different grades or suppliers. 
6.1.2.2 Types of Pharmaceutical Excipients 
To prepare a drug substance into a fi nal dosage form, pharmaceutical excipients 
should be added. The Handbook of Pharmaceutical Excipients presents more than 
FIGURE 3 Correlation and functions of drug combined with pharmaceutical excipients in 
dosage form design. 
+ 
Efficacy, 
Adverse effect 
Multiple 
Functionality 
Bioavailability 
Safety/Quality 
drug Pharmaceutical 
Excipients

TABLE 1 Pharmaceutical Excipients Used in Tablet Formulations 
Excipient Type Defi nition Examples 
Adsorbent Agent capable of holding other 
molecules onto its surface by 
physical or chemical 
(chemisorption) means 
Powdered cellulose, activated 
charcoal 
Antioxidant Agent that inhibits oxidation and 
thus is used to prevent 
deterioration of preparations by 
oxidative process 
Ascorbic acid, ascorbyl palmitate, 
butylated hydroxyanisole, 
butylated hydroxytoluene, 
hypophosphorus acid, 
monothioglycerol, propyl 
gallate, sodium ascorbate, 
sodium bisulfi te, sodium 
formaldehyde, sulfoxylate, 
sodium metabisulfi te 
Colorant Used to impart color to tablet FD & C red no. 3, no. 20, FD & C 
yellow no. 6, FD & C blue no. 2, 
D & C green no. 5, D & C 
orange no. 5, D & C red no. 8, 
caramel, ferric oxide, red 
Encapsulant Used to form thin shells for purpose 
of enclosing drug substance or 
drug formulation for ease of 
administration 
Gelatin, cellulose acetate 
phthalate 
Plasticizer Used as component of fi lm coating 
solutions to enhance spread of 
coat over tablets, beads, and 
granules 
Diethyl phthalate, glycerin 
230 monographs of excipients used in the dosage formulations [1] . The amount and 
type of pharmaceutical excipients are highly dependent on the fi nal dosage forms. 
In the preparation of tablets, diluents or fi llers are commonly added to increase the 
bulk of the formulation, binders to cause the adhesion of the powdered drug and 
pharmaceutical substances, antiadherents or lubricants to assist in the smooth tableting 
process, disintegrating agents to promote tablet break - up after administration, 
and coating agents to improve stability, control disintegration, or enhance appearance. 
Thus, the pharmaceutical excipients establish the primary features and physicochemical 
properties of the tablet, such as the physical form, stability, dissolution, 
taste, and overall appearance [3] . Table 1 presents examples of pharmaceutical 
excipients used in tablet formulations according to principal categories. 
6.1.2.3 Characteristics of Pharmaceutical Excipients 
Filler (Diluent) Powder mixtures should be compacted to achieve the appropriate 
strength at a low compaction pressure in the tablet preparation. The compaction 
properties of a formulation will largely be governed by its major components. For 
a high - dose drug the drug itself will strongly infl uence the compaction, while for 
low - dose drugs the bulk size needs to be increased with an inactive ingredient 
SELECTION OF PHARMACEUTICAL EXCIPIENTS 885

886 PHARMACEUTICAL PREFORMULATION 
Excipient Type Defi nition Examples 
Surfactant Substance that adsorbs to surfaces 
or interfaces to reduce surface or 
interfacial tension; may be used 
as wetting agent, detergent, or 
emulsifying agents 
Benzalkonium chloride, 
nonoxynol 10, oxtoxynol 9, 
polysorbate 80, sodium 
lauryl sulfate, sorbitan 
monopalmitate 
Tablet 
antiadherent 
Agent that prevents the sticking of 
tablet formulation ingredients to 
punches and dies during tablet 
production 
Magnesium stearate, talc 
Tablet binder Substance used to cause adhesion 
of powder particles in tablet 
granulations 
Acacia, alginic acid, 
carboxymethylcellulose 
sodium compressible sugar, 
ethylcellulose gelatin, liquid 
glucose, metylcellulose 
povidone, pregelatinized starch 
Tablet diluent Inert substance used as fi ller to 
create desired bulk, fl ow 
properties, and compression 
characteristics in preparation of 
tablets 
Dibasic calcium phosphate, 
kaolin, lactose, mannitol, 
microcrystalline cellulose, 
powdered cellulose, 
precipitated calcim carbonate, 
sorbitol, starch 
Tablet sugar 
and fi lm 
coating 
excipient 
Used to coat a formed tablet for 
purpose of protecting against drug 
decomposition by atmospheric 
oxygen or humidity, to provide 
desired release pattern for drug 
substance after administration, 
to mask taste or odor of drug 
substance, or for aesthetic 
purposes 
Sugar: liquid glucose, 
sucroseFilm: hydroxyethyl 
cellulose, hydroxypropyl 
methylcellulose, 
methylcellulose, ethylcellulose 
Enteric: cellulose acetate 
phthalate, shellac (35% in 
alcohol) 
Tablet direct - 
compression 
excipient 
Used in direct - compression tablet 
formulations 
Dibasic calcium phosphate 
Tablet 
disintegrant 
Used in solid dosage forms to 
promote disruption of solid mass 
into smaller particles which are 
more readily dispersed or 
dissolved 
Alginic acid, 
carboxymethylcellulose 
calcium, microcrystalline 
cellulose, polacrilin potassium, 
sodium alginate, sodium starch 
glycollate, starch 
Tablet glidant Agent used in tablet and capsule 
formulations to improve fl ow 
properties of powder mixture 
Colloidal silica, corn starch, talc 
Tablet 
lubricant 
Substance used in tablet 
formulations to reduce friction 
during tablet compression 
Calcium stearate, magnesium 
stearate, mineral oil, stearic 
acid, zinc stearate 
Tablet 
opaquant 
Used to render tablet coating 
opaque; may be used alone or in 
combination with colorant 
Titanium dioxide 
Tablet - 
polishing 
agent 
Used to impart an attractive sheen 
to coated tablets 
Carnauba wax, white wax 
TABLE 1 Continued

termed a diluent (or fi ller). High - dose formulations may also use a diluent to overcome 
compaction problems experienced with an active pharmaceutical substance. 
There are a number of general rules for selecting a diluent. The selection of the 
diluent will depend on the type of processing and plasticity of materials to be used. 
A direct - compression formulation will require a diluent with good fl ow and compaction 
properties. If the material is extremely plastic, it is appropriated to add a diluent 
that compacts by brittle fracture; similarly, a brittle drug substance should be combined 
with a plastic fi ller. The solubility of the drug substance should also be considered. 
A soluble drug is normally formulated with an insoluble fi ller to optimize 
the disintegration and dissolution process. The hydrophilic excipients added in the 
formulation may also change drug solubility. Table 2 lists the more commonly used 
diluents in tablet formulation. 
Binder (Granulating Agent) Before tableting the powder mixture via direct compression, 
generally powders are granulated simply by adding water or an organic 
solvent to form liquid bridges followed by the drying process. This granulation 
process can make powders of larger particle size and that are more free fl owing for 
tablet production. The most common method of adding binders is as a solution in 
the granulating fl uid. It is also possible add synthetic polymers such as PVP and 
HPMC as powders and use water as the granulating agent. When the granulate dries, 
the crystallization of any solids that had dissolved in the liquid will form solid bonds 
between the particles [8] . Inclusion of granulating agents or binders to increase 
granule strength is necessary. Granulating agents are usually hydrophilic polymers 
that have cohesive properties that both aid the granulation process and impart 
strength to the dried granulate. 
For a granulating agent to be effective, it must form a fi lm on the particle surface 
and be selected on the basis of its spreading coeffi cient. The spreading coeffi cient 
is defi ned as the difference between the work of adhesion of the binder and the 
substrate and the work of cohesion of the binder. Commonly used granulating 
agents are listed in Table 3 . The binder may vary the disintegration and dissolution. 
Binders form hydrophilic fi lms on the surface of the granules, which can aid in the 
TABLE 2 Commonly Used Tablet Diluents 
Diluent Comments 
Lactose Available as anhydrous and monohydrate; anhydrous material 
used for direct compression due to superior compressibility 
Microcrystalline cellulose Originally direct - compression excipient, now often included in 
granulations due to its excellent compressibility 
Dextrose, glucose Direct - compression diluent, often used in chewable tablets 
Sucrose Was widely used as sweetener/fi ller in effervescent tablets and 
chewable tablets; less popular nowadays due to cariogenicity 
Starch and derivatives Versatile material that can be used as diluent binder, and 
disintegtant 
Calcium carbonate Brittle material 
Dicalcium phosphate Excellent fl ow properties; brittle material 
Magnesium carbonate Direct - compression diluent 
SELECTION OF PHARMACEUTICAL EXCIPIENTS 887

888 PHARMACEUTICAL PREFORMULATION 
wetting of hydrophobic drugs. However, if added at too great concentrations, the 
fi lms can form viscous gels on the granule surface and will retard dissolution. 
Disintegrant Tablets must have suffi cient strength to withstand the stresses of 
subsequent manufacturing operations, such as the coating, packaging, and distribution 
process. However, once the tablet is taken by the patient, it must break up 
rapidly to ensure rapid dissolution of the active ingredient in immediate - release 
preparations. To overcome the cohesive strength produced by the compression 
process and to break down the tablet into the primary particles as rapidly as possible, 
the disintegrants are combined with other excipients during the tableting process. 
Starch was the fi rst disintegrant used in tablet manufacture. Recently, so - called 
superdisintegrants, including croscarmellose sodium, sodium starch glycolate, and 
crospovidone, display excellent disintegration activity at low concentrations and 
have better compression properties than starches. Traditionally, swelling and rate of 
swelling have been regarded as the most important characteristics of disintegrants. 
With the aid of these superdisintegrants, sustained - release acetaminophen tablets 
with biphasic patterns were successfully established to mimic the bilayered Tylenol 
ER tablet [9]. As a general rule, soluble drugs are formulated with insoluble fi llers 
to maximize the effect of disintegrants. The positioning of disintegrants within the 
intragranular and extragranular portions of granulated formulations can affect their 
water uptake and disintegration time. 
Commonly used disintegrants are listed in Table 4 . The greater the level of disintegrant, 
the faster the tablet will disintegrate. The compaction properties of many 
disintegrants, including starch, are not satisfactory and use of high concentration 
could also reduce tablet strength. Disintegrants are hygroscopic materials and will 
absorb moisture from the atmosphere, which could negatively affect the stability of 
moisture - sensitive drugs if the packaging does not provide adequate protection 
from the environment. Disintegrant activity can be affected by mixing with hydrophobic 
lubricants so that care needs to be taken to optimize the manufacturing 
TABLE 3 Commonly Used Granulating (Binding) Agents 
Granulating Agent 
Normal 
Concentration 
(%) Comments 
Starch 5 – 25 Was once the most commonly used binder; 
starch has to be prepared as paste, which 
is time consuming 
Pregelatinized starch 0.1 – 0.5 Cold - water soluble so easier to prepare 
than starch 
Acacia 1 – 5 Requires preparation of past prior to use; 
can lead to prolonged disintegration 
times if used at too high a concentration 
Polyvinylpyrrolidone 
(PVP) 
2 – 8 Available in range of molecular weight/ 
viscosities; soluble in water and ethanol 
Hydroxypropyl 
methylcellulose (HPMC) 
2 – 8 Low - viscosity grades most widely used 
Methylcellulose (MC) 1 – 5 — 

process as well as the formulation. If the tablet contains a high proportion of a 
hydrophobic drug that has a high contact angle, a wetting agent or surfactant should 
be added to the formulation to modify disintegration time and subsequent dissolution 
of the drug from tablet. The most commonly used wetting agents are sodium 
lauryl sulfate and the polysorbates. 
Most pharmacopeias include a disintegration test which can be applied to tablets 
and capsules and the detailed monograph is given in the pharmacopeias (see Section 
6.1.5.1 ). 
Lubricant The use of a lubricant is essential to increase the free fl ow of powders 
and to prevent manufacturing disorders in the tablet production. The type and 
amount of lubricant are cautiously selected in the formulation. The order of addition 
and mixing time is also considered in the tableting process. 
There are three types of lubricants employed in solid dosage form manufacture. 
The fi rst class of lubricant is the glidant . The fl ow properties of a powder can be 
enhanced by the inclusion of a glidant. These are added to overcome powder cohesiveness. 
The two other classes of lubricant are antiadherent excipients, which reduce 
the friction between the tablet punch faces and tablet punches, and die wall lubricant 
excipients, which reduce the friction between the tablet surface and the die wall 
during and after compaction to enable easy ejection of the tablet. The level of a 
lubricant required in a tablet is formulation dependent and can be optimized using 
an instrumented tableting machine. 
Commonly used lubricants are listed in Table 5 . Talc is traditionally one of the 
most commonly used glidants, having the additional benefi t of being an excellent 
antiadherent. The level of talc that can be added to a formulation is restricted by 
its hydrophobic nature, too high levels resulting in decreased wetting of the tablet 
and a subsequent reduction in the rate of dissolution. Fumed silicon dioxides are 
TABLE 4 Commonly Used Disintegrants 
Disintegrant 
Normal 
Concentration 
(%) Comments 
Starch 5 – 10 Probably works by wicking; swelling 
minimal at body temperature 
Microcrystalline cellulose Strong wicking action; loses disintegrant 
action when highly compressed 
Insoluble ion exchange resins Strong wicking tendencies with some 
swelling action 
Sodium starch glycolate a 2 – 8 Free - fl owing powder that swells rapidly 
on contact with water 
Croscarmellose sodium a 1 – 5 Swells on contact with water 
Gums — agar, guar, xanthan < 5 Swell on contact with water; form viscous 
gels that can retard dissolution, thus 
limiting concentration that can be used 
Alginic acid, sodium alginate 4 – 6 Swell like gums but form less viscous gels 
Crospovidone a 1 – 5 High wicking activity 
a Often mentioned as superdisintegrant. 
SELECTION OF PHARMACEUTICAL EXCIPIENTS 889

890 PHARMACEUTICAL PREFORMULATION 
TABLE 5 Lubricants Commonly Used in Formulations 
Lubricant 
Typical 
Percent Comments 
Glidants 
Talc 1–5 Fine, crystalline powder; Widely used as lubricant 
and diluent 
Fumed silicon dioxide: 
Aerosil, Cab - O - Sil, 
Syloid 
0.1–0.5 Has small particle size and large surface area for 
good fl owability; used for adsorbent, antitacking 
agent disintegrant, and glidant 
Starch 1 – 10 Mainly used for binder, disintegrant, and diluent but 
also used for glidant 
Sodium lauryl sulfate 0.2 – 2 Anionic surfactant, luricant and wetting agent 
Boundary Lubricants 
Magnesium stearate 0.2 – 2 Hydrophobic, variable properties between suppliers 
Calcium silicate 0.5 – 4 Hydrophobic 
Sodium stearyl fumarate 0.5 – 2 Less hydrophobic than metallic stearates, partially 
soluble 
Polyethylene glycol 4000 
and 6000 
2 – 20 Soluble, poorer lubricant activity than fatty acid 
ester salts 
Sodium lauryl sulfate 1 – 3 Soluble, also acts as wetting agent 
Magnesium lauryl sulfate 1 – 3 Acts as wetting agent 
Sodium benzoate 2 – 5 Soluble 
Fluid Lubricants 
Light mineral oil 1 – 3 Hydrophobic, can be applied to either formulation 
or tooling 
Hydrogenated vegetable 
oil 
1 – 5 Hydrophobic, used at higher concentrations as 
controlled - release agents 
Stearic acid 0.25 – 2 Hydrophobic 
Glyceryl behenate 0.5 – 4 Hydrophobic, also used as controlled - release agent 
perhaps the most effective glidants. These are materials with very small (10 - nm) 
spherical particles that may achieve their glidant properties. They are available in a 
number of grades with a range of hydrophobic and hydrophilic forms and also commercially 
available under diverse brand names. Starch has also been used as a 
glidant. The use of large amounts of starch has also aided the disintegration 
properties. 
Die - wall lubricants can be dived into two classes, fl uid and boundary lubricants . 
Fluid lubricants work by separating moving surfaces completely with a layer of 
lubricant. These are typically mineral oils or vegetable oils, and they may be either 
added to the mix or applied directly to the die wall by means of wicked punches. 
The oily lubricants may have a mottled appearance in the tablet due to uneven distribution, 
poor powder fl ow due to their tacky nature, and reduced tablet strength. 
Fluid lubricants include stearic acid, mineral oils, hydrogenated vegetable oils, glyceryl 
behenate, paraffi ns, and waxes. Boundary lubricants work by forming a thin 
solid fi lm at the interface of the die and the tablet. Metallic stearates are the most 

widely used boundary lubricants. Such lubricants should have low shear strength 
and form interparticulate fi lms. 
Magnesium stearate is the most widely used lubricant. The magnesium stearate 
used in the pharmaceutical industry is not a pure substance but a mixture of magnesium 
salts of fatty acids, though predominantly magnesium stearate and magnesium 
palmitate. Despite its popularity, which is a refl ection of its excellent lubricant 
properties, it has some problems associated with product consistency: its effect on 
tablet strength and its hydrophobicity. The U.S. Pharmacopeia (USP) requires that 
the stearate content should account for not less than 40% of the fatty acid content 
of the material, and the stearate and palmitate combined should account for not 
less than 90%. Within this defi nition, there are a range of materials to be supplied 
as magnesium stearate. For a given formulation, it is important that a single source 
of magnesium stearate be used for all batches to get product reliability. 
The lubricant activity of magnesium stearate is related to its readiness to form 
fi lms on the die wall surface. As a result, it has two consequences: a reduction in the 
ability of the powder to form strong compacts and, due to its hydrophobicity, a deleterious 
effect on the dissolution rate of the tablets. The hydrophobic surfaces 
created by magnesium stearate have been shown to reduce the rate of dissolution 
and bioavailability of several tablet formulations. When both lubricant and disintegrant 
are being added to a granulated formulation, the disintegrant should be 
blended with the granules prior to the addition of the lubricant to minimize the risk 
of forming a hydrophobic fi lm around the disintegrant. 
The third class of lubricant activity is the antiadherent. Some materials have 
adhesive properties and can adhere to the punch surfaces during compression. This 
will induce tablet disorders: sticking, with a fi lm forming on the surface of the tablets, 
or picking, where solid particles from the tablet stick to the punch surface. Most die 
wall lubricants also have antiadherent actions, and in many formulations, the addition 
of a specifi c antiadherent will not be required separately. The antiadherent 
includes talc, maize starch, and microcrystalline cellulose. 
Coating Materials The core compressed tablet can be used by itself, but are additional 
coating process of the compressed tablet can be applied for several reasons: 
(a) protection of the drug from the environment (moisture, air, light) for stability 
reasons, (b) taste masking, (c) minimizing patient/operator contact with drug substance, 
particularly for skin sensitizers, (d) improving product identity and appearance, 
(e) improving ease of swallowing, (f) improving mechanical resistance and 
reducing abrasion and attrition during handling, and mostly (g) modifying release 
properties. 
There are three main methods used to coat pharmaceutical tablets: sugar coating, 
fi lm coating , and compression coating . Sugar coating has been the most commonly 
used method and involves coating tablets with sucrose. A sugar - coated tablet is 
water based and generally starts to break up in the stomach. This is a highly skilled 
and multistep process that is very labour intensive. This coating process results in a 
50% increase of the fi nal tablet weight and in a signifi cant increase in tablet size. 
Traditionally, sugar coating has been performed in coating pans in which the tablets 
are tumbled in a three - dimensional direction. The pan is supplied with a source of 
warm air for drying and an extraction system to remove moist air and dust. The 
coating solution is distributed around the tablets by their tumbling action. A dusting 
SELECTION OF PHARMACEUTICAL EXCIPIENTS 891

892 PHARMACEUTICAL PREFORMULATION 
powder may be sprinkled onto the surface of the tablets during the drying phase to 
prevent the tablets from sticking together. The cycle of wetting and drying is continued 
until the desired amount of coating has been applied to the tablets. Typically, 
a sugar coating will consist of three types of coats: a sealing coat, a subcoat, and a 
smoothing coat. Traditionally, a seal coating of shellac dissolved in ethanol or synthetic 
water - resistant polymers such as cellulose acetate phthalate or polyvinylacetate 
phthalate is used. The subcoat is an adhesive coat on which the smoothing coat 
of the sharp corners of the tablet can be built. The subcoat is a mixture of a sucrose 
solution and an adhesive gum, such as acacia or gelatin, which rapidly distributes 
over the tablet surface. A dusting follows each application of solution with a subcoat 
powder containing materials such as calcium carbonate, calcium sulfate, acacia, talc, 
and kaolin. The smoothing coat consists of the majority of the tablet bulk and provides 
the tablet with a smooth fi nish. A colorant is also applied if needed. The coat 
consists of sucrose syrup which may contain starch or calcium carbonate. The coated 
tablets are usually transferred to a polishing pan and coated with a beeswax – 
carnauba wax mixture to provide a glossy fi nish to the surface. 
Film coating involves the application of a polymer fi lm to the surface of the tablet, 
gelatin capsules, and multiparticulate systems with a negligible increase in tablet 
size. The method of application of the coat differs from the sugar coating in that the 
coating suspension is sprayed directly onto the surface of the tablets, and drying 
occurs as soon as the coat hits the tablet surface. The tablet only receives a small 
quantity of coating solution at a time. The fi lm coat can generally be affected by the 
following properties: a method of atomizing the coating suspension, the ability to 
heat large volumes of air (which heat the tablets and facilitate the rapid drying of 
the applied coat), and a method of moving the tablets that ensures all tablets are 
evenly sprayed. 
The main methods of coating are modifi ed conventional coating pans, side - vented 
pans, and fl uid bed coating. The side - vented pan, now the most commonly used 
equipment for fi lm coating, was designed to maximize the interaction between the 
tablet bed and the drying air. The mixing effi ciency of the table, granules, or pellets 
is achieved by using appropriately designed baffl es on the pan surface. Fluid - bed 
coating offers an alternative to pan coating and is particularly popular for coating 
multiparticulate systems. There are three methods by which the coating can be 
applied: top spraying, bottom spraying, and tangential coating. The pellets, granules, 
or tablets being coated are suspoended in an upward stream of air, maximizing the 
surface available for coating. The coating is applied by an atomizer, and this is dried 
by the fl uidizing air. 
Table 6 gives commonly used polymers for fi lm coating of core tablet. With the 
exception of HPMC, the polymers are rarely used alone but are combined with 
other polymers to optimize the fi lm - forming properties. A polymer for fi lm coating 
will ideally meet the following criteria: 
1. Solubility in the solvent selected for application: Currently the organic solvent 
are replaced with water as a suspension system, although certain types of fi lm 
coatings may require organic solvents to be used. Commonly used solvents 
include alcohols (methanol, ethanol, and isopropanol), esters (ethyl acetate 
and ethyl lactate), ketones (acetone), and chlorinated hydrocarbons (dichloromethane 
and trichloroethane). 

2. Solubility in GI fl uids: The solubility of polymers is dependent on its physicochemical 
nature and pH. Unless the coating is being applied for enteric coating, 
it should ideally be soluble across the range of pH values encountered in the 
GI tract. 
3. Capacity to produce an elegant fi lm even in the presence of additives such as 
plasticizers, pigments, and colorants. 
4. Compatibility with fi lm - coating additives and the tablet being coated. 
5. Stability in the environment under normal storage conditions. 
6. Freedom from undesirable taste or odor. 
7. Lack of toxicity. 
Enteric coating materials are also used to prevent release of the drug substance 
in the stomach if the drug is either an irritant to the gastric mucosa or unstable in 
gastric juice. Table 7 lists enteric coating polymers commonly used in tablet formulations. 
The choice of of enteric coating material depends on its solubility. 
The third type of tablet coating is multiple - compression coating to make a bilayered, 
multilayered tablet (a layered tablet of two drugs) or a tablet within a tablet 
(a core of one drug and a shell of another). The multilayered tablet is prepared by 
initial compaction of a portion of the fi ll materials in a die followed by additional 
fi ll material and compression, depending on the number of fi ll materials. The layered 
tablet can provide some advantages. Each layer has different drug in a separate 
layer. The incompatible drug can be compressed simultaneously at different layers. 
TABLE 6 Commonly Used Polymers for Film Coating of Core Tablet 
Polymer Comments 
Methylcellulose (MC) Soluble in cold water, GI fl uids, and a range of organic 
solvents 
Ethylcellulose (EC) Soluble in organic solvents, insoluble in water and GI 
fl uids; used alone in modifi ed - release formulations 
and in combination with water - soluble cellulose for 
immediate - release formulations 
Hydroxyethylcellulose (HEC) Soluble in water and GI fl uids 
Methyl hydroxyethylcellulose 
(MHEC) 
Soluble in water and GI fl uids; has similar fi lm - forming 
properties to HPMC but is less soluble in organic 
solvents, which limited its popularity when solvent 
coating was the norm 
Hydroxypropyl cellulose (HPC) Soluble in cold water, GI fl uids, and polar solvents; 
becomes tacky when dried, so is unsuitable for use 
alone, often used in combination with other polymers 
to optimize adhesion of coat 
Hydroxypropyl methylcellulose 
(HPMC) 
Soluble in cold water, GI fl uids, alcohols, and 
halogenated hydrocarbons; excellent fi lm former and 
the most widely used polymer; can be used with 
lactose to improve adhesiveness 
Sodium carboxymethylcellulose 
(NaCMC) 
Soluble in water and polar solvents 
SELECTION OF PHARMACEUTICAL EXCIPIENTS 893

894 PHARMACEUTICAL PREFORMULATION 
The staged release and improved appearance from the layered tablet are also possible. 
In the preparation of a tablet within a tablet, a special tableting machine is 
required to place the intended core tablet precisely within the die, which already 
contains some of the coating formulation to surround the fi ll materials. This multilayered 
tablet or tablet within a tablet is also useful to get the modifi ed release, 
either immediately or in a sustained manner. 
Auxiliary Excipients Plasticizers are added during the tablet coating process. The 
fi lm coating process involves two important stages: droplet formation and fi lm formation. 
Film formation is a multistage process that involves wetting the tablet 
surface followed by spreading the fi lm and eventually coalescence of the individual 
fi lm particles into a continuous fi lm. Most fi lm - forming polymers have glass transition 
temperatures in excess of the temperatures reached during the coating process 
(typically 40 – 50 ° C), so it is necessary to add plasticizers to the formulations which 
reduce the glass transition temperature. The choice of plasticizer is dependent on 
the type of polymer and its permanence and compatibility. Permanence is the duration 
of the plasticizer effect; the plasticizer should remain within the polymer fi lm 
to retain its effect, so it should have a low vapor pressure and diffusion rate. Compatibility 
requires the plasticizer to be miscible with the polymer. Commonly used 
plasticizers include phthalate esters, citrate esters, triacetin, propylene glycol, polyethylene 
glycols (PEGs), and glycerol. 
The pigments or opacifi ers are also combined with the coating solution to get 
colored tablets. Insoluble pigments are normally preferred to soluble dyes for a 
number of reasons. Solid pigments produce a more opaque coat than dyes, protecting 
the tablet from light. The presence of insoluble particles in the suspension allows 
TABLE 7 Enteric Coating Polymers Commonly Used in Tablet Formulations 
Polymer Solubility Profi le Comments 
Shellac Above pH 7 Original enteric coating material, originally 
used in sugar - coated tablets; high pH 
required for dissolution may delay drug 
release; natural product which exhibits 
batch - to - batch variability 
Cellulose acetate 
phthalate (CAP) 
Above pH 6 High pH required for dissolution a 
disadvantage; forms brittle fi lms, so must be 
combined with other polymers 
Polyvinylacetate 
phthalate (PVAP) 
Above pH 5 — 
Hydroxypropyl 
methylcellulose 
phthalate (HPMCP) 
Above pH 4.5 Optimal dissolution profi le for enteric coating 
Polymers of 
methacrylic acid 
and its esters 
Various grades 
available with 
dissolution 
occurring 
above pH 6 
— 

the rate of solid application to the tablet to be increased without having an adverse 
effect on the viscosity of the coating suspension, improving productivity. 
6.1.2.4 Selection Guideline of Pharmaceutical Excipients in Tablet Formulation 
In the proper selection of pharmaceutical excipients for the formulation, there are 
numerous factors to be considered [7] . The type of excipient is highly dependent on 
the model compound and its intended dosage form. The preparation method of the 
dosage form and proper dosage regime are also factors. 
The formulation parameters of the tablet are essential. The amount and type of 
excipients relative to drug contents should be considered to determine the size of the 
dosage form. The bulk density of excipients and drug and their fi lling doses in the die 
are important factors to be considered in the tablet formulation . Uniform distribution 
of drug or excipient is in trouble if its contents are low in the formulation, while the 
use of high contents are diffi cult to fi ll in the die and the tablet size is also larger. 
Excipients that are potentially incompatible with the drug should be avoided. The 
amount and type of reactive impurities in the excipients should also be established. 
Batch - to - batch and supplier - to - supplier variation in impurity levels is possible. 
Excipients with potential adverse interaction and any unwanted incompatibility with 
drug should be avoided in the formulation. For an ideal formulation, it is helpful to 
consider these para meters as well as drug – excipient incompatibility. 
The global harmonization and standardization of pharmaceutical excipients are 
nowadays necessary in the formulation studies [3, 7] . Since all excipients in the market 
worldwide are supplied from many countries or companies with facilities in more 
than a single country, the quality of the excipients must be well documented and validated. 
Otherwise, the quality of the excipients should be varied batch to batch, factory 
to factory, and country to country. There are also hundreds of different brands and 
grades of excipients available, but it would be unrealistic for the formulator to expect 
to have a totally free choice. Most manufacturing companies select the excipients 
used in their factory on the basis of cost, availability, and performance. To establish 
the equivalency of excipients obtained from different sources, it is also necessary to 
perform some kind of functionality testing. Since the sources, origin, and manufacturer 
of the excipients are different, regulatory approval for the product is required 
in each country. The standards for each drug substance and excipient are contained 
in pharmacopeia. The four pharmacopeia with the largest international use are the 
United States Pharmacopeia (USP) and National Formulary (NF), British Pharmacopoeia 
(BP), European Pharmacopoeia (EP), and Japanese Pharmacopoeia (JP). 
Unless global harmonization is established, analytical methods, testing criteria, 
and specifi cation limits must be variable according to monographs of pharmacopeia 
from the different countries. For example, Table 8 lists the different specifi cation 
limits for the viscosity of cellulose ether among three pharmacopeias. If global harmonization 
and standardization of pharmaceutical excipients are established, the 
marketing and sales of a single formulation are more facilitated worldwide. The 
manufacturing cost and research - and - development (R & D) cycles are also reduced. 
Most of all, the quality and bioequivalence of the drug products with the same formulation 
can be more validated since the regulatory approval of the pharmaceutical 
product is enhanced. The global harmonization is an ongoing effort by corporate 
representatives and international regulatory authorities. 
SELECTION OF PHARMACEUTICAL EXCIPIENTS 895

896 PHARMACEUTICAL PREFORMULATION 
In the selection of excipients, formulation scientists should also remember that 
every excipient is not limited by a single function but rather can be used for many 
pharmaceutical applications in dosage forms. For example, cellulose and its derivatives 
(HPMC, EC, HPC) have been widely used as fi llers, binders, suspending agents, 
and mainly controlled - release agents. Povidone, polymethacrylate, and cyclodextrins 
are also multifunctional excipients in formulations. Table 9 gives the physical properties 
of some directly compressible fi llers. The grades of physical properties are 
variable among fi llers used in tablet formulation. The simple and optimal formulation 
of drugs with multifunctional excipients can provide some advantages in tablet 
preparations. 
6.1.3 DRUG – EXCIPIENT COMPATIBILITY 
The potential for excipients to cause chemical and physical instability in drugs has 
been recognized for over 30 years. Drug compatibility studies have been used as an 
TABLE 8 Specifi cation Limits for Viscosity of Cellulose Ether 
Parameter EP USP XXII JP XII 
Concentration of 
solution, % 
2.0 2.0 2.0 
Temperature, ° C 20 ± 0.1 20 ± 0.1 20 ± 0.1 
Type of viscometer Rotating 
viscometer 
Capillary type, 
Ubbelohde 
Capillary type, 
Ubbelohde 
Shear rate, S . 1 10 — — 
Unit cP (Pa · s) cP (Pa · s) cSt 
Sample preparation a 
a Very similar in all three pharmacopeia. 
TABLE 9 Comparative Properties of Some Directly Compressible Fillers a 
Filler Compactibility Flowability Solubility Disintegration Hygroscopicity Lubricity Stability 
Dextrose 3 2 4 2 1 2 3 
Spray dried 
lactose 
3 5 4 3 1 2 4 
Fast - Flo 
lactose 
4 4 4 4 1 2 4 
Anhydrous 
lactose 
2 3 4 4 5 2 4 
Emdex 
(dextrates) 
5 4 5 3 1 2 3 
Sucrose 4 3 5 4 4 1 4 
Starch 2 1 0 4 3 3 3 
Starch 1500 3 2 2 4 3 2 4 
Dicalcium 
phosphate 
3 4 1 2 1 2 5 
Avicel 
(MCC) 
5 1 0 2 2 4 5 
a Graded on a scale from 5 (good/high) to 1 (poor/low); 0 means none. 

approach for accepting/rejecting excipients for use in pharmaceutical formulations 
[1, 10 – 12] . In general, drug stability can be investigated under the stress condition 
according to the guideline of accelerated stability testing. The factors for stress 
condition usually include temperature, pH, light, moisture, agitation, gravity, packaging, 
and method of manufacture [13, 14] . Despite this fact, approaches for excipient 
selection in drug formulation are often empirical. The stability issues in the development 
of a drug must be considered at the early and late formulation stages. Nowadays, 
advanced analytical instrumentation makes it possible to more rapidly identify 
potential excipient - induced instability to select excipients. Excipients that exhibit 
incompatibility with the drug are “ rejected ” and not included in subsequent tablet 
formulation studies. 
6.1.3.1 Experimental Studies for Drug – Excipients Compatibility 
Compatibility studies are carried out by mixing drug with one or more excipients 
under some type of stress condition. It has also been suggested that aqueous suspensions 
of the drug and excipients or drug – excipient complexes provide a better model 
for tablet formulations. It has been recommended that small - scale formulations 
using the selected excipients (this may eventually be used for the eventual formulation) 
be prepared using experimental processes. 
Table 10 provides examples of binary and factorial design for drug – excipient 
compatibility studies. The two conditions of each potential parameter can be included 
as a binary system. All potential parameters can also be combined for factorial 
design. For example, water is added to the drug – excipient mixture at reasonable 
temperatures (50 ° C or less) and then both intact drug and degradation products 
are measured. The high - temperature and high - humidity conditions are usually used 
to obtain more rapid stability assessment of drug and excipients. Generally, physicochemical 
properties such as drug content, color, taste, related substances, thermal 
analysis, and high - performance liquid chromatography (PLC) studies of evaluated 
for drug – excipient compatibility studies using conventional analytical techniques. 
6.1.3.2 Analytical Methods for Drug – Excipient Incompatibility 
The key to the early assessment of instability in formulations is the availability of 
analytical methods to detect low levels of degradation products, generally less than 
2%. With the aid of thermal analysis and chromatographic methods [HPLC and 
TABLE 10 Examples of Binary and Factorial Designs for Drug-Excipient Compatibility 
Studies 
Binary Design Factorial Design 
Drug – excipient ratio 1 : 1 or 1 : 10 Total number of formulation = a . b . c . d . e 
Water addition = yes/no Type of drug – excipient ( a ) 
Storing temperature 25 ° C or 40 ° C/75% Amount of drug – excipient ( b ) 
Storing time 1 or 4 weeks Water addition ( c ) 
Storing temperature ( d ) 
Storing time ( e ) 
DRUG–EXCIPIENT COMPATIBILITY 897

898 PHARMACEUTICAL PREFORMULATION 
liquid chromatography/mass spectrometry (LC/MS)], it can assign at least tentative 
structures for most degradation products as well as the intact drug contents [15] . 
Excipient compatibility screening must provide more rapid identifi cation of excipient 
- mediated instability that is detected in complete formulations. 
Early compatibility studies relied on color change as an indication of incompatibility. 
Subsequently, there were numerous reports on the use of thermal analysis 
techniques such as differential thermal analysis (DTA) or differential scanning calorimetry 
(DSC) to detect drug – excipient incompatibilities. DTA and DSC have the 
advantage of rapid analysis. Generally, formation of new peaks and disappearance 
of drug peak by the endothermic or exothermic reaction are carefully investigated. 
Diffuse refl ectance spectroscopy (DRS) can be also used to determine the ultraviolet 
(UV) absorption of drug on the surface of drug – excipient mixtures. 
6.1.3.3 Reaction Types and Stabilization Guidelines 
Understanding the degradation chemistry of drug with excipients is essential to 
select proper excipients in the formulation stages [16, pp 101 – 151]. Drug - excipient 
compatibility studies are crucial to decide optimal tablet formulation and to understand 
the possible mechanism in many cases [10, 12, 14] . Drug instability occurs by 
three types of reactions: hydrolysis, oxidation, and aldehyde – amine addition. Table 
11 gives reaction types of chemical and physical instability. 
Hydrolysis is the most common mechanism to induce drug – excipient incompatibility. 
Most hydrolysis reactions are catalyzed by acids and/or bases. The microscopic 
pH between drug and excipients is critical. The pH degradation rate profi les are 
good predictors for hydrolysis reactions in solid dosage forms. The pH of optimal 
stability in solution is similar to the “ microscopic pH ” in solid dosage forms. A shift 
TABLE 11 Reaction Types of Chemical and Physical Instability 
Type of Instability Order of Frequency 
Chemical instability 
pH - dependent hydrolysis 12 
Oxidation 
Air oxidation 5 
Metal - catalyzed oxidation 3 
Peroxides in excipients 2 
Aldehyde – amine addition 
Aldose excipient 2 
Formaldehyde from excipients 1 
Michael addition with maleate salt 1 
Intramolecular cyclization 1 
Dimerization (Diels – Alder) 1 
Racemization 1 
Addition of ammonia residue from gelatin capsule 1 
Esterifi cation 1 
Physical instability 
Evaporation of volatile free base 2 
Reaction of methanesulfonic acid with disintegrants 1 
Gelatin cross - linking by excipient impurities 1 

in only one pH unit can increase or decrease the reaction rate by a factor of 10. 
Although solid - state compatibility studies are commonly used, solution kinetic 
studies are generally conducted to select excipients since it is possible to study all 
three of these reactions in solution. Often solution kinetic studies allow the identi- 
fi cation of more potential degradation products and hence the development of 
better stability - indicating assays. The pH of optimal stability is also important for 
selecting the appropriate salt form of the compound and excipients in the solid 
formulation. Acetylsalicylic acid (ASA) is a readily hydrolyzable drug and there are 
literature references to its rate of hydrolysis in aqueous solution. 
The moisture content of the drug and excipients plays a critical role in their 
incompatibility by hydrolysis. Excipients such as starch and povidone have particularly 
high water contents (povidone contains about 28% equilibrium moisture at 
75% relative humidity), which can increase the possibility of drug contact. Magnesium 
trisilicate causes increased hydrolysis of aspirin in tablet form because, it is 
thought, of its high water content. For these reasons, some scientists recommend 
inclusion of water in the samples for compatibility studies. Depending on the degree 
of hydrolytic susceptibility, different approaches for tablet formulation can be used 
to minimize hydrolysis. For compounds such as ASA that are readily hydrolyzable, 
direct compression or dry granulation is preferable rather than wet granulation. 
However, drug – excipient incompatibility still occurrs. Chemical interaction between 
moieties of drug and excipients may lead to increased decomposition. The transacetylation 
reaction between aspirin and paracetamol and also possible direct 
hydrolysis of the paracetamol can occur. The amount of free salicyclic acid at 37 ° C 
in the tablets containing paracetamol increases by the addition of talc (0.5 – 1%). The 
stearate salts should be avoided as tablet lubricants if the active component is 
subject to hydroxide ion – catalyzed degradation. The degradative effect of the alkali 
stearates is inhibited in the presence of malic, hexamic, or maleic acid. 
As a general rule, in selecting excipients, it is probably best to avoid hygroscopic 
excipients when formulating hydrolytically labile compounds. One of the most effective 
ways to stabilize a pH - sensitive drug is through adjustment of the microscopic 
pH of the formulation. Excipients with high pH stability and buffering agents are 
recommended. The equilibrium moisture content (hygroscopicity) at different relative 
humidities for a variety of drug and excipients would be a clue to selecting 
optimal formulations. If dry processing and the use of nonhygroscopic excipients 
still result in unacceptable rates of hydrolysis, use of a dessicant and/or moisture 
protective packaging can further increase stability against drug hydrolysis. The 
manufacturing process should be conducted under low - humidity conditions and not 
during the hot summer season to improve drug – excipient compatibility. 
Oxidation reactions are complex and it is diffi cult to understand the reaction 
mechanism. The best approach is to avoid excipients containing oxidative reactants 
such as peroxides and metal ions. The air oxidation or metal ion – catalyzed oxidation 
can be tested after storing the samples in the solutions. The impurities in excipients 
such as povidone or as degradation products in PEGs are organic peroxides and 
are typically more reactive than hydrogen peroxide. A commercially available 
organic peroxide such as tert - butyl hydroperoxide is better to evaluate the susceptibility 
of a compound to peroxide oxidation rather than the hydrogen peroxide. 
Reactive impurities such as peroxides and ionic chemicals (talc and titanium oxide) 
in the excipients commonly may act as catalysts for oxidation of the drug. 
DRUG–EXCIPIENT COMPATIBILITY 899

900 PHARMACEUTICAL PREFORMULATION 
Therefore, it is essential to remove any oxidative peroxides and ionic chemicals. 
Use of free - radical scavengers such as butylated hydroxyanisole (BHA) or butylated 
hydroxytoluene (BHT) can stabilize the oxidation reaction of the drug with 
excipients via a free - radical mechanism. However, uniform distribution of the low 
levels of these antioxidants in solid formulations is quite diffi cult. The BHA is added 
to both lovastatin and simvastatin tablets. If an oxidation reaction is catalyzed by 
metal ions or excipients with high transition metal ion contents (talc), chelating 
agents can also be used to bind trace metal ions. However, citrate is often the agent 
of choice because of the potential toxicity of many chelating agents. Oxidation is 
less likely to occur if the oxidizable group of phenols/catechols and secondary and 
tertiary amines is protonated. If some oxidation reactions are catalyzed by light, the 
formulation can be stabilized by a light - absorbing coating. 
Aldehyde – amine addition is also a typical reaction type. The potential aldehyde 
interaction also makes sense to screen out a compound as part of the preformulation 
evaluation. All aldoses such as lactose or excipients such as starch and microcrystalline 
cellulose that have terminal aldose groups should be avoided with the excipients 
having primary/secondary amines. 5 - (Hydroxymethyl) - 2 - furaldehyde (HMF) would 
be a good choice since it is a degradation product of many sugars (lactose) and celluloses, 
at least in trace levels. The Schiff ’ s base[ – CH=N – ] is formed when the sugar 
aldehyde (lactone) and the primary/secondary amines are mixed. The isoniazide 
and trace level of HMF from lactose can also readily form the Schiff ’ s base. It has 
been reported that the reaction of hydrazine hydrochloride and starch to form 
high - molecular - weight addition products gives high - molecular - weight products. 
The reaction of fl uoxetine hydrochloride is more rapid with spray - dried lactose 
monohydrate. 
Many excipients are acids or bases or have acidic or basic impurities. For this 
reason, the reaction of amines with aldehydes requires that the amine exist in the 
nucleophilic - free base, rather than the protonated cationic form. For example, the 
reaction of fl uoxetine hydrochloride with lactose was much more rapid as potassium 
hydroxide was added to neutralize the hydrochloride salt. Michael addition between 
seproxetine and maleic acid from a tablet formulation following dissociation of the 
hydrochloride salt occurs because of the conversion of a salt to a free acid/base. 
Finally, the physical instability that can occur is the cross - linking of gelatin or its 
capsules. Low levels of aldehyde impurities in excipients (starch, polysorbate 80, 
PEG, and rayon coils) and packaging materials have been reported to cause cross - 
linking of gelatin. Dissolution slowing is also more pronounced for wet granulation 
tablets than direct - compression tablets. Dissolution slowing appears to be due to 
hardening of the water - soluble excipients and a reduction of disintegration time. 
Once suspect excipients have been eliminated from consideration, small - scale 
formulations with manufacturing processes such as granulation, drying, and compression 
can be used to assess whether interactions between excipients or processing 
conditions result in any unpredicted instability. The ratio of drug and excipients is 
also a critical factor to consider in incompatibility studies. In addition, the lower the 
drug dose, the greater the possibility of degradation by low - level impurities in the 
excipients. 
In conclusion, drug – excipient compatibility studies have a key role at the early 
preformulation stages to select excipients or after formulation to help identify the 
mechanism of any detected instability [14] . An understanding of the potential physicochemical 
interactions of drug with known chemical reactivities of excipients and 

POWDER CHARACTERISTICS 901 
their impurities will aid in the proper selection of excipients. Knowledge of trace 
impurities/additives in excipients and the consistency of these levels from batch to 
batch and supplies to supplier is also essential to select proper pharmaceutical 
excipients. Although the best solution kinetics and drug – excipient compatibility 
studies may be established, the possibility of unexpected instability because tablets 
and capsules are complex multicomponent systems must be remembered. 
6.1.4 POWDER CHARACTERISTICS 
Most drug and inactive excipients used in tablet formulation are in the solid state 
as amorphous powder or crystals of various morphological structures. There may 
be substantial differences in particle size, surface area, crystal morphology, wetting, 
and fl owability as well as many physical properties of drug, excipients, and their 
blends [16] . Table 12 describes common micromeritic topics important to pharmaceutical 
preformulation. 
Before their use in the solid dosage forms, it is necessary to understand and characterize 
the physical and chemical properties of drug, excipient, and their powder 
mixtures , including crystal habit, particles size, shape, fl ow characteristics, density, 
hygroscopicity, and compressibility and compaction [2, 3] . Hiestand noted that successful 
tableting operations require the selection of excipients that balance desirable 
physical, fl ow, and mechanical properties for tablet manufacturing [17] . The quantifi - 
cation of these properies using a unifi ed approach is essential to the design and potimization 
of solid dosage formulations [1] . Instrumental analyses such as scanning 
electron microscopy (SEM), DSC, and powder X - ray diffraction (PXRD) can be very 
useful to characterize powder properties such as purity, polymorphism, salvation, 
degradation, drug – excipient compatibility, and other desirable characteristics. 
6.1.4.1 Crystal Form and Habit 
The morphology of a pharmaceutical solid is of importance since this property can 
infl uence the bulk powder properties. The six crystal systems are cubic, hexagonal, 
tetragonal, orthorhombic, monoclinic, and triclinic crystals. The observed overall 
TABLE 12 Micromeritic Topics Important to 
Pharmaceutical Preformulation 
Particle shape 
Particle size distribution 
Solid geometry (packing, density, porosity, void) 
Surface characteristics (adsorption, area, surface energy, 
solubility) 
Methods of determination 
Chemical stability 
Dynamics (fl ow rate, transport) 
Particle separation 
Processing (sieving, sedimentation, grinding, mixing, 
compaction) 
Sampling 
Drug release 

902 PHARMACEUTICAL PREFORMULATION 
shape of crystal habits comprises plate, tabular, equant, columnar, blade, and avicular 
as well as dendrites, treelike pattern, and spherulites, tiny crystals radiating from 
a center [2, 18, 19] . Crystal form (crystal habit) as well as a noncrystalline amorphous 
form may affect drug stability, dissolution rate, fl ow, mechanical properties, and 
ability to mix with excipients. The amorphous form of a drug has the lowest melting 
point and usually the fastest dissolution rate, but it is most likely to react or degrade. 
Mechanical properties such as fl owability, miscibility, particle strength, and cohesiveness 
often vary among the polymorphs, crystal shapes, or habits. Cohesiveness, the 
surface free energy effect that results in particle aggregation, may also be different 
for various polymorphs and habits. 
6.1.4.2 Particle Size Distribution 
The determination and control of particle size distribution are often very important 
in pharmaceutical preformlation since the drug safety, stability, and viability of the 
dosage form and manufacturing process can be signifi cantly infl uenced [19] . Particle 
shape is also important in determining particle size. The particle size of materials is 
readily expressed in terms of its diameter according to the defi nition of particle size. 
As the degree of asymmetry of particles increases, however, so does the diffi culty 
of expressing size in terms of a meaningful diameter [20] . Figure 4 shows different 
ways of defi ning a diameter. The “ diameter ” can be simply the longest or the shortest 
linear dimension of the crystal. If one can calculate the area of the particle, one 
may obtain the diameter d a , which is the diameter of a circle with the same area as 
the particle. 
If all the particles in a sample are of the same size, then the powder is monodisperse. 
Truly, the particles have more than one size in polydisperse samples. The 
monodisperse particle size distribution is more desirable than the polydisperse one. 
Therefore, the shape and surface area of the individual particles, the size ranges 
based on number or weight of particles, as well as the total surface area are variable. 
The commonly illustrated particle forms are sphere, rod, fi ber, granular, cubical, 
fl ake, condensation fl oc, and aggregate. 
The fi neness of the powder is characterized by a number (e.g., a diameter d ). 
Particles, of course, will have different shapes so that there are different ways of 
defi ning a diameter. The technique for obtaining d a given above has been used 
microscopically. More conventional is the so - called surface mean diameter, which, 
is the diameter of a sphere that has the same surface area as the particle. The so - 
called single - particle volume mean diameter is possible if there are instruments that 
can measure the volume of an odd - shaped particle. If the shape factor is indepen- 
FIGURE 4 Different ways of defi ning a diameter. 
Irregular shape Sphere with area (A) and volume (V) 
d (small) 
d (A), d(V) d (large) 

POWDER CHARACTERISTICS 903 
dent of the size of the particle, then the shape is called isometric. Examples of isometric 
shapes are cube, sphere, and cylinder. 
Suppose a sample of 12 particles of corn starch with fairly narrow particle size 
distribution (25 – 35 . m) were measured. In this case, the distribution of particles is 
very narrow and approximately distributed as normal or Gaussian pattern. To 
convert these numbers into frequencies, it is noted that there are 12 particles in 
total; that is, dividing each number by 12 and multiplying this by 100 will give the 
percent frequency, as shown in Table 13 . 
If the frequency is plotted as a function of the midpoint of the diameter ranges, 
then a frequency histogram is obtained. It is noted that for the noncumulative curve 
the midpoints of the intervals are used, but for the cumulative curve the interval 
endpoint is used. When the number or weight of particles lying within a certain size 
range is plotted against the size range or mean particle size, a frequency distribution 
curve is obtained However, due to the deviation from the normal distribution of 
particles, a lognormal distribution of the particle size is statistically plotted against 
the cumulative percent frequency on a probability scale, and a linear relationship is 
observed (Figure 5 ). Probability paper is a type of paper that straightens out this 
type of S - shaped curve. The logarithm of the particle size is equivalent to 50% on 
the probability scale, that is, the 50% size is known as the geometric mean diameter 
and the slope is given by the geometric standard deviation .g , which is also the quotient 
of the ratio: (84% undersize or 16% oversize)/(50% size) or (50% size)/(16% 
undersize or 84% oversize). 
Many methods are available for determining particle size in pharmaceutical 
practice, including microscopy, sieving, sedimentation, and determination of particle 
volume [19] . Sieve analysis with U.S. standard sieves is widely used to determine the 
particle size distribution based on powder weight. Sieves are classifi ed according to 
the number of openings (Table 14 ) and are generally made of wire cloth woven 
from brass, bronze, or other suitable wire. 
The USP uses descriptive terms to characterize the particle size of a given 
powder, which are related to the proportion of powder that is capable of passing 
through the openings of standardized sieves of varying dimensions in a specifi ed 
time period under the mechanical sieve shaker as follows: 
Coarse (or a no. 20) powder: All particles pass through a no. 20 sieve and not 
more than 40% through a no. 60 sieve. 
Moderately coarse (or a no. 40) powder: All particles pass through a no. 40 sieve 
and not more than 40% through a no. 60 sieve. 
TABLE 13 Particle Size Distribution of Powdered Cornstarch Samples 
Particle Size 
Range ( . m) 
Number of 
Occurrences ( n ) 
Percent 
Frequency 
Cumulative 
Frequency (%) 
25 – 27 1 8.3 8.3 
27 – 29 3 25.0 33.3 
29 – 31 4 33.3 (33.4) 66.7 
31 – 33 3 25 91.7 
33 – 35 1 8.3 100 
Total 12 99.9 (100) — 

904 PHARMACEUTICAL PREFORMULATION 
Fine (or a no. 80) powder: All particles pass through a no. 80 sieve. There is no 
limit as to greater fi neness. 
Very fi ne (or a no. 120) powder: All particles pass through a no. 120 sieve. There 
is no limit as to greater fi neness. 
1 10 100 
0.01 
0.1
1 
10 
30 
50 
70 
90 
99 
99.9 
Weight 
Number 
Cumulative % undersize 
Particle size (.m) 
FIGURE 5 Typical lognormal distribution of particles based on weight and number. 
TABLE 14 Openings of Standard Sieves 
Sieve Number Sieve Opening 
2 9.5 mm 
3.5 5.6 mm 
4 4.75 mm 
8 2.36 mm 
10 2.00 mm 
20 850 . m 
30 600 . m 
40 425 . m 
50 300 . m 
60 250 . m 
70 212 . m 
80 180 . m 
100 150 . m 
120 125 . m 
200 75 . m 
230 63 . m 
270 53 . m 
325 45 . m 
400 38 . m 

POWDER CHARACTERISTICS 905 
Microscopy, in which the particles are sized through the use of a calibrated grid 
background or other measuring devices (range 0.2 – 100 . m). SEM can also readily 
measure the smallest particle size. The microscope allows the observer to view the 
actual particles, but it gives two - dimensional views. With the sedimentation rate, 
particle size is determined by measuring the terminal settling velocity of particles 
through a liquid medium in a gravitational or centrifugal environment (range 
0.8 – 300 . m). Sedimentation rate may be calculated from the well - known Stokes 
equation. The sedimentation methods yield a particle size relative to the rate at 
which particles settle through a suspending medium, a measurement important in 
the development of emulsions and suspensions. With light energy diffraction (light 
scattering), particle size is determined by the reduction in light reaching the sensor 
as the particle, dispersed in a liquid or gas, passes through the sensing zone (range 
0.2 – 500 . m). On the other hand, laser scattering utilizes a H 3 – Ne laser, silicon 
photodiode detectors, and an ultrasonic probe for particle dispersion (range 0.02 – 
2000 . m). The measurement of particle volume using a Coulter counter allows one 
to calculate an equivalent volume diameter, but no information on shape of the 
particles is available. Laser holography, in which a pulsed laser is fi red through an 
aerosolized particle spray and photographed in three dimensions with a holographic 
camera, allows the particles to be individually imaged and sized (range 
1.4 – 100 . m). The above methods and others may be combined to provide greater 
assurance of size and shape parameters. Automated particle size analyzers linked 
with computers are commercially available for data processing, distribution analysis, 
and printout. 
Determination and control of particle size are often prerequisites in preformulation 
stages because the size distribution of excipients, drug, and their mixtures can 
infl uence safety, effi cacy, stability, viability of dosage form, and manufacturing processes. 
Furthermore, the particle size of pharmaceuticals can affect uniform mixing, 
fl ow characteristics, formulation characteristics, dose - to - dose content uniformity, 
dissolution rate, and bioavailability of drug. Tablet characteristics such as porosity 
and fl owability are highly affected by the particle size as well. The smallest particles 
induce electrostatic forces and aggregations while the larger particles show greater 
weight variations. The ideal size ranges of particles are usually 10 – 150 . m. Therefore, 
detailed information of the particle size of drug, excipients, and their blends should 
be required in tablet formulation as well as regulatory issues. Particle size is also 
important in the tableting fi eld, since it can be very important for good homogeneity 
in the fi nal tablet. The particle size should be consistent throughout the production 
to satisfy table formulation and regulatory demands. 
6.1.4.3 Flow Characteristics 
Good fl ow properties are a prerequisite for the successful manufacture of both 
tablets and powder - fi lled hard gelatin capsules. Proper fl uidity of materials is 
required to be transported through the hopper of a tableting machine. The elongated 
particle shape and small particle size could cause high tablet weight variation, 
strength, unacceptable blend uniformity, and diffi culty in fi lling containers and 
dies. Excipients with good fl ow characteristics and low cohesive powders 
should be more preferable in tablet production. Powder fl ow is affected by the 
numerous parameters, including purity, crystallinity, electrostatic forces, mechanical 

906 PHARMACEUTICAL PREFORMULATION 
properties (brittleness, elasticity), density, size, shape, surface area, moisture content, 
direction and rate of shear, storage container dimension, and particle – wall interaction 
[19] . 
It is a property of all powders to resist the differential movement between particles 
when subjected to external stresses. A bulk powder is somewhat analogous to 
a non - Newtonian liquid, which exhibits plastic fl ow and sometimes dilatancy if the 
particles being infl uenced by attractive forces. Accordingly, powders may be free 
fl owing or cohesive ( “ sticky ” ). The resistance is to free fl ow is due to the cohesive 
forces between particles [18] . Three principal types of interparticular forces are 
forces due to electrostatic changing, van der Waals forces, and forces due to moisture. 
Electrostatic forces are dependent on the nature of the particles, in particular 
their conductivity. Van der Waals forces are the most important forces for most 
pharmaceutical powders. These forces are inversely proportional to the square of 
the distance between the two particles and hence diminish rapidly as particle size 
and separation increase. Powders with particles below 50 . m will generally exhibit 
irregular or no fl ow due to van der Waals forces. Particle shape is also important; 
for example, the force between a sphere and a plane surface is about twice that 
between two equal - sized spheres. At low relative humidity, moisture produces a 
layer of adsorbed vapor on the surface of particles. Above a critical humidity, typically 
in the range 65 – 80%, it will form water liquid bridges between particles. Where 
a liquid bridge forms, it will give rise to an attractive force between the particles 
due to surface tension or capillary forces. The role of the formulator is to ensure 
that the fl ow properties of the powder are suffi cient to enable its use on modern 
pharmaceutical equipment, powder hoppers, and fl ow through orifi ces in the tablet 
production. 
It is important that the powder fl ows from the hopper to the fi lling station of the 
tablet machine at an appropriate rate and without segregation occurring. There are 
two types of fl ow that can occur from a powder hopper: core fl ow and mass fl ow [2] . 
Figure 6 shows the two different powder fl ow patterns in hoppers. When a small 
amount of powder is allowed to leave the hopper, there is a defi ned region in which 
downward movement takes place and the top surface begins to fall in the center. A 
core fl ow hopper is characterized by the existence of dead spaces during discharge. 
A mass fl ow hopper is one in which all the material is in motion during discharge, 
in particular the areas adjacent to the hopper wall. As a small amount of powder is 
discharged, the whole bulk of the powder will move downward. Whether core fl ow 
or mass fl ow is achieved is dependent on the design of the hopper (geometry and 
wall material) and the fl ow properties of the powder. 
Powder fl ow into orifi ces is also important when fi lling dies in tablet machines 
and in certain types of capsule - fi lling machines. For a given material, the fl ow into 
or through an orifi ce is dependent on the particle size (Figure 7 ). In general, as the 
particle size increases, the powder fl ow rate also increases. However, there is practically 
no fl ow if the particle size is below 50 . m or above 1200 . m. The Carr index 
gives us the guidance for powder fl owability. A lower Carr index of excipients is 
more desirable for acceptable powder fl ow. At the lower end of the particle size 
range, cohesive forces will result in poor fl ow. Powders with particles below 50 . m 
will generally exhibit irregular or no fl ow due to van der Waals forces. As the particle 
size increases, the fl ow rate increases until a maximum is achieved, at an orifi ce 
diameter – particle diameter ratio of 20 – 30. As the particle size continues to increase, 

POWDER CHARACTERISTICS 907 
FIGURE 6 Powder fl ow patterns in hoppers. 
(a) Core flow 
(b) Massflow 
FIGURE 7 Effect of particle size on rate of powder fl ow through orifi ce. 
Particle size 
Powder flow rate 
No flow below 50 .m 
No flow above 1200 .m 
the rate decreases due to mechanical blocking or obstruction of the orifi ce. Flow 
will stop completely when the orifi ce – particle ratio falls below 6 and if the size is 
above 1200 . m. 
There are several different methods available for determining the fl ow properties 
of powders. Shear cell methods provide an assessment of powder fl ow properties as 
a function of consolidation load and time. There are a number of types of shear cells 
available, the most common being the Jenike shear cell [21] . 

908 PHARMACEUTICAL PREFORMULATION 
Common indices of fl owability are the Hausner ratio and the Carr index (compressibility). 
The increase in bulk density of a powder is related to the cohesiveness 
of a powder. So measurement of the bulk density of a powder is essential to defi ne 
the fl ow characteristics. Ratios of poured - to - tapped bulk densities are expressed in 
two ways to give indices of fl owability: 
Hausner ratio 
tapped bulk density 
poured bulk density 
= 
Carr index (compressibility) 
(tapped bulkdensity poured bu = 
. . 100 lk density) 
poured bulk density 
The Hausner ratio varies from about 1.2 for a free - fl owing powder to 1.6 
for cohesive powders. Carr index classifi cations for fl owability [2] are listed in 
Table 15 . 
Compressibility indices are a measure of the tendency for arch formation and 
the ease with which the arches will fail and, as such, is a useful measure of fl ow. A 
limitation of the bulk density indices for fl ow characteristics is that they only 
measure the degree of consolidation; they do not describe how rapidly consolidation 
occurs. Angle of repose is a common method used to measure powder fl ow with 
small sample quantity. If powder is poured from a funnel onto a horizontal surface, 
it will form a cone due to gravitational forces. The angle between the sides of the 
cone and the horizontal is referred to as the angle of repose. So there is a correlation 
between powder fl ow and angle of repose. The relationship between the Carr 
index and angle of repose is now discussed. The angle of repose is a measure of the 
cohesiveness of the powder, as it represents the point at which the interparticle 
attraction exceeds the gravitational pull on a particle. A free - following powder will 
form a cone with shallow sides, and hence a low angle of repose, while a cohesive 
powder will form a cone with steeper sides. As a rough guide, angles less than 30 ° 
are usually indicative of good fl ow, while powders with angles grater than 40 ° are 
likely to be problematic. 
The avalanching behavior of powder is also a measure of fl owability. If a powder 
is rotated in a vertical disc, the cohesion between the particles and the adhesion of 
the powder to the surface of the disc will lead to the powder following the direction 
of rotation until it reaches an unstable situation where an avalanche will occur. After 
the avalanche, the powder will again follow the disc prior to a further avalanche. 
TABLE 15 Carr Index Classifi cation and Powder 
Flowability 
Carr Index (%) Flow 
5 – 12 Free fl owing 
12 – 16 Good 
18 – 21 Fair 
23 – 35 Poor 
33 – 38 Very poor 
> 40 Extremely poor 

POWDER CHARACTERISTICS 909 
Measurement of the time between avalanches and the variability in time is a measure 
of the fl ow properties of the powder. 
If a powder fl ows poorly, the vibrator can be used, but it also causes powder segregation 
and stratifi cation. The addition of glidant (occasionally lubricant) in the 
powder mixtures can readily increase fl owability at the low concerntration. Talc or 
fumed silicon dioxide is an example of a glidant. If this is not suffi cient to improve 
the fl ow, other means of fl ow improvement are necessary. There are two main factors 
that affect powder fl ow: particle size and particle shape. The more spherical a particle 
is, the better it fl ows. Small particles are very cohesive and hence do not fl ow 
well, but increasing the particle size will improve fl ow. With the aid of spray drying 
or spheronizers , particles become spherical. 
In general, powder below 50 . m is not very free fl owing because the cohesive 
forces below this size become stronger than the gravitational force, and fl ow through 
the orifi ce is prevented. This, of course, is a function of the size of the orifi ce, and 
fl ow might be possible in a larger orifi ce. 
Particle size enlargement of the drug substance can be brought about by manipulation 
of the recrystallization step in the synthesis of the drug. To increase powder 
fl ow, particle size enlargement by slugging, roller compaction, and wet granulation 
can be used. If a powder is compressible but does not fl ow well, then slugging may 
be employed. In slugging, tablets are made of the poorly fl owing substance on a 
high - duty, slowly operating machine into large dies. The dies are large so that the 
fl ow is suffi ciently increased, but now the compression forces must be increased 
because the larger area dictates a larger force to attain a given pressure (the elastic 
limit being in stress units). In roller compacting , the powder is processed between 
two heavy - duty rollers, compacted between the rolls, and emerges as a compressed 
sheet, which is then milled. These two methods are necessary if the drug substance 
(e.g., aspirin) is suffi ciently moisture sensitive and there are stability issues so it 
cannot be wet granulated. Otherwise, wet granulation is a frequently used method 
of particle enlargement for free - fl owing powder. 
6.1.4.4 Density and Bulkiness 
Density When a powder is poured into a container, the volume that it occupies 
depends on a number of factors, such as particle size, particle shape, and surface 
properties. In normal circumstances, it will consist of solid particles and interparticlulate 
air spaces (voids or pores). The particles themselves may also contain 
enclosed or intraparticulate pores. If the powder bed is subjected to vibration or 
pressure, the particles will move relative to one another to improve their packing 
arrangement. Ultimately, a condition is reached where further densifi cation is not 
possible without particle deformation. The density of a powder is therefore dependent 
on the handling conditions to which it has been subjected, and there are several 
defi nitions that can be applied either to the powder as a whole or to individual 
particles. 
Because particles may be hard and smooth in one case and rough and spongy in 
another, one must express densities with great care. Density is universally defi ned 
as weight per unit volume. Three types of densities — true density, particle density, 
and bulk density — can be defi ned, depending on the volume of particles containing 
microscopic cracks, internal pores, and capillary spaces. 

910 PHARMACEUTICAL PREFORMULATION 
The true density is the material itself exclusive of the voids and interparticular 
pores larger than molecular or atomic dimension in the crystal lattice. Particle 
(granular) density, determined by the displacement of mercury, which does not 
penetrate at ordinary pressure into pores smaller than 10 . m, is the mass of the 
particle divided by its volume. The different terms arise from the defi nition of 
volume: True particle density is when the volume measured excludes both open and 
closed pores and is a fundamental property of a material; apparent particle density 
is when the volume measured includes intraparticulate pores; effective particle 
density is the volume “ seen ” by a fl uid moving past the particles. Bulk density 
(powder density) is the volume in a graduated cyclinder including both the particulate 
volume and the pore volume. The bulk density will vary depending on the 
packing of the powder. Based on the defi nition of volume, minimum bulk density is 
when the volume of the powder is at a maximum, caused by aeration, just prior to 
complete breakup of the bulk. Poured bulk density is when the volume is measured 
after pouring powder into a cylinder, creating a relatively loose structure. Tapped 
bulk density is, in theory, the maximum bulk density that can be achieved without 
deformation of the particles. 
Based on the defi nition of density, two new terms are defi ned. Porosity is defi ned 
as the proportion of a powder bed or compact that is occupied by pores and is a 
measure of the packing effi ciency of a powder and relative density is the ratio of the 
measured bulk density and the true density: 
Porosity 
bulk density 
true density 
= . 1 
Relative density 
bulk density 
true density 
= 
Bulkiness The specifi c bulk volume, the reciprocal of bulk density, is often called 
bulkiness or bulk . It is an important consideration in the packaging and fi lling of 
powders for tablet production. The bulk density of calcium carbonate can vary from 
0.1 to 1.3, and the lightest or bulkiest type would require a container about 13 times 
larger than the heaviest type. Bulkiness increases with a decrease in particle size. In 
a mixture of materials of different sizes, however, the smaller particles sift between 
the larger ones and tend to reduce the bulkiness. 
To defi ne bulkiness in detail, the porosity and density of powders should be 
understood. Suppose a powder, such as zinc oxide, is placed in a graduated cylinder 
and the total volume is noted. The volume occupied is known as the bulk volume 
V b . If the powder is nonporous, that is, has no internal pores or capillary spaces, the 
bulk volume of the powder consists of the true volume of the solid particles plus 
the volume of the spaces between the particles. The volume of the spaces, known 
as the void volume v , is given by equation 
v V V = . b p 
where V p is the true volume of the particles. 
The porosity or voids ( . ) of a powder is also defi ned as the ratio of the void 
volume to the bulk volume of the packing given below. Porosity is frequently 
expressed in percent, . . 100:

POWDER CHARACTERISTICS 911 
. = 
. 
= . 
V V 
V 
V
V 
b p 
b 
p
b 
1 
6.1.4.5 Hygroscopicity 
The hygroscopicity of a drug and pharmaceutical substances is a potential parameter 
to be considered in tablet formulation. The moisture uptake rate is quite variable 
depending on the type of drug and excipients as well as the environmental conditions. 
So, a concise defi nition of hygroscopicity is not possible. Powders can absorb 
moisture by both capillary imbibition and swelling. The instantaneous water absorption 
prosperties of pharmaceutical excipients correlate with total surface area while 
the total absorption capacity correlates with powder porosity [22] . 
If drug and excipients are so hygroscopic, they can readily adsorb water until 
they deliquesce, or begin to dissolve. Moisture adsorption is important because 
adsorbed water can cause incorrect weighing and degradation of drug and/or excipients. 
The drug, excipients, and water reaction will continue as the amount of water 
increases. When a solid is placed in a room, moisture will condense onto it. If this 
occurs simply as a limited amount of adsorbed moisture, then the substance is not 
hygroscopic under these conditions. These conditions exist if the water vapor pressure 
in the surrounding atmosphere is lower than the water vapor pressure over a 
saturated solution of the solid in question. However, if the water vapor pressure in 
the atmosphere is higher than that of the saturated solution, there will be a thermodynamic 
tendency for water to condense upon the solid materials (drug and 
excipients). 
The drug and pharmaceutical excipients adsorb or lose the moisture depending 
on the relative humidity in the atmosphere. The nonhygroscopic materials are not 
affected by the moisture and are in a equilibrium state. In general, solid dosage 
forms such as tablets or capsules should be hydrophilic because the solid materials 
must dissolve after swallowing. However, solid dosage forms must also be stable 
against physical and chemical factors. 
The moisture uptake rates (MUR) can simply be obtained by weighing the 
sample after a given time (six days), but in such a case it is assumed that the moisture 
uptake is still in the linear phase. If, for instance, the weight gain is 5 mg per 10 - g 
sample in six days, then the MUR is 5/(10 . 6) = 0.083 mg/g/day. If the MUR values 
are plotted versus relative humidity (RH) and the curve that intercepts the x axis 
at 20% RH is obtained from a straight line, the compound can be stored without 
moisture pickup in atmospheres of less than 20% RH. In addition, the hygroscopicity 
of materials is indicated as follows: 
Loss of drying (LOD%) 
weight of water in sample 
total weight of wet sa 
= 
mple 
. 100 
Moisture content (MC%) 
weight of water in sample 
weight of dry sampl 
= 
e 
. 100 
Equilibrium moisture contents (EMC) 
%LOD 
LOD 
= 
+ 
. 
% 100 
100 
Relative humidity (RH) 
water vapor pressure in atmosphere 
saturat 
= 
ed water vapor pressure 
. 100 

912 PHARMACEUTICAL PREFORMULATION 
Depending on the hygroscopicity based on the EMC, various drug and excipients 
are classifi ed in four groups (Table 16 ). 
Figure 8 also shows an example of moisture uptake for four selected excipients 
as a function of relative humidity. Depending on the hygroscopicity of the exscipients, 
the uptake behaviors are quite variable. Excipients such as microcrystalline 
cellulose (MCC) and starch can pick up signifi cant amounts of water at relatively 
low relative humidity. Since this water is not present as a hydrate, it is potentially 
free to interact with a drug. 
TABLE 16 Classifi cation of Hygroscopicity and Example Pharmaceutical Excipients 
Type II: Type II: 
Type I: 
Nonhygroscopic 
Slightly 
Hygrocopic 
Moderately 
Hygroscopic 
Type IV: 
Very Hygroscopic 
No MC change 
below RH 90% or 
less than 20% MC 
changes at RH 
90% after 1 week 
storage 
No MC change 
below RH 
80% or less 
than 40% MC 
changes at RH 
80% after 1 
week storage 
Not more than 5% 
MC change below 
RH 60% or less 
than 50% MC 
changes at RH 
70% after 1 week 
storage 
Increase of MC at RH 
40 – 50% or more 
than 70% MC 
changes above RH 
40% after 1 week 
storage 
Examples 
Lactose USP MCC HPC Povidone 
Dicalcium phosphate Sucrose, dextrose HPMC Sodium starch glycolate 
Ethylcellulose Poloxamer 188 Bentonite Polyplasdone XL 
Magnesium stearate PEG 3350 Pregelatinized starch Sorbitol 
CAP Starch USP, Corn CMC Na 
Gelatin USP 
FIGURE 8 Profi les of moisture uptake for four selected excipients as function of relative 
humidity. 
Relative humidity(%) 
0 20 40 60 80 100 
Moisture content (%) 0 
10 
20 
30 
40 
50 
60 
Lactose USP, anhydrous 
Cellulose acetate phtahlate NF 
Magnesium aluminum silicate NF 
Povidone USP 

POWDER CHARACTERISTICS 913 
The moisture content of drug can affect the cohesiveness of particles through 
hydrogen bonding or by changing surface energy effects. Water also can act as a 
plasticizing agent and can lower the glass transition temperature of amorphous 
polymorphs, thus allowing a rubbery state to exist at a lower temperature. Water 
also can take up intra - and intergranular pore spaces (capillaries) of powders by 
acting as an interparticle bridge through surface adsorption mechanisms. 
The hygroscopicity of materials infl uences the fl owing characteristics of drug and 
excipients by forming adsorption fi lm with an aid of water as a solvent. Dissolution 
of drug and excipients from solid dosage forms can also occur. The crystallinity of 
troglitazone - PVP K30 solid disperisions can be changed by the water content [23] . 
During manufacturing operations, water ’ s ability to act as a bridge between particles 
can improve the compression capabilities of powder masses and affect the tensile 
strength and hardness of the tablet. The milling or blending is also changed. Most 
of all, the chemical hydrolysis of labile materials is more facilitated, resulting in low 
stability during storage. Adsorbed water also can act locally as a solvent, and in 
many cases drug – excipient incompatibility occurs by dissolving drug or excipients 
in granulation or blended powder mass, as observed when aspirin tablets liberate a 
strong odor of free salicyclic acid. Any free, unbound water in the mass can migrate 
throughout the material mass and act as a reagent. A waterproof packaging or 
container is used for many moisture - sensitive materials. 
6.1.4.6 Mixing 
The mixing of powders is a key step in the manufacture of virtually all solid dosage 
forms. In general, particle size, shape, and surface energy are important factors for 
blending of drug and excipients in unit operations. The perfect mixing of powder is 
desirable, that is, a mixture in which the probability of fi nding a particle of a given 
component is the same at all positions in the mixture, but the powder mixing has a 
maximum degree of randomness (Figure 9 ). To determine the degree of mixing 
obtained in a pharmaceutical operation, it is necessary to reasonably sample the 
mixture and determine the variation within the mix statistically [2] . 
Uniform mixing of powdered materials occurs if they have similar particle size 
distributions and particle shapes. Spherical particles mix least well while plate and 
fi ber shapes also do not mix well because they tend to clump. The more cohesive 
the material, the more diffi cult it is to mix that material with other materials. Similarly, 
cohesiveness between drug and excipient or among excipients may hinder the 
FIGURE 9 Comparison of powder mixing: perfect mixing and random mixing. 

914 PHARMACEUTICAL PREFORMULATION 
successful blending process. Ideally, the uniform mixing of powders should be such 
that the weight of sample taken is similar to the weight that the powder mix contributes 
to the fi nal dosage form. 
Figure 10 shows the effect of mixing time on the mechanical strength of powders. 
As the mixing time increases, the tensile strength of powders (or curshing strength 
of the tablet) gradually decreases [24, 25] . While the strength decreases with increasing 
mixing time for all materials tested, the effect is far more marked for materials 
that deform plastically. For example, the glidant tensile strength invariably decreases 
as the mixing time (2 min vs. 3 min) increases (Figure 11 ). There is no direct correlation 
of tensile strength with primary particle size of glidant (Figure 12 ). However, 
FIGURE 10 Effect of mixing time on mechanical strength of excipients. 
Mixing time 
Crushing strength or tensile strength 
FIGURE 11 Effect of mixing time glidant tensile strength. 
Mixing time (min) 
Aerosil 
R 812 
Aerosil 300 
Aerosil R 805 
Aerosil 200 
Aerosil OX 50 
Aluminum oxide C 
Printex 95 
Printex G 
Printex 25 
Titanium dioxide P 25 
Titanium dioxide T 805 
Tensile strength (Pa) 
0
2
4
6
8 
10 
12 
14 
2 min 
30 min 

POWDER CHARACTERISTICS 915 
this correlation was further improved when some outlying glidant (Aserosil 200 and 
300 and Printex 25) were excluded ( R 2 values of 0.1287 – 0.8616). 
If a powder consisting of two materials both having identical physical properties 
is mixed for a suffi cient time, random mixing will eventually be achieved. Unfortunately, 
most pharmaceutical powders consist of mixtures of materials with differing 
physical properties, such as size, shape, density, and surface area, leading to segregation 
among particles, where particles of similar properties tend to collect together 
in part of the powder. When segregating powders are mixed, as the mixing time is 
extended, the powders appear to unmix. The differences in particle size are the most 
important for segregation in pharmaceutical powders. One exception to overcome 
segregation is ordered mixing rather than random mixing. When one component of 
a powder mix has a very small particle size (less than 5 . m) and the other is relatively 
large, the fi ne powder may coat the surface of the larger particles, and the adhesive 
forces will prevent segregation, known as ordered mixing. This ordered mixing 
makes the powders produce greater homogeneity than by random mixing. The percolation 
of fi ne particles is also a factor. If the particles sizes are quite different, the 
smaller particles can drop easily and move to the bottom of powder, resulting in 
segregation. This segregation process can occur whenever movement of particles by 
vibration, shaking, and pouring takes place. 
6.1.4.7 Particle Size Reduction (Micronization and Milling) 
Mechanical attrition, that is , high - energy ball milling of powders, is a nonequilibrium 
processing method that has generated the reduced particle size and the formation 
of physically metastable materials. It can be used to modify materials by refi ning 
the microstructure, homogenizing the composition, extending solid solubility, creating 
matastable crystalline phases, or producing metallic glass. High - energy ball 
milling is both a processing method to reduce particle size and a route to the physical 
synthesis of metastable materials. Early in product development, when only 
small amounts of drug are available, comminution (grinding/mixing) may be carried 
out with a mortar and pestle. For lager batches, ball milling or micronization can be 
FIGURE 12 Correlation of tensile strength with primary particle size of glidant. 
Primary particle diameter (nm) 
0 10 20 30 40 50 60 
Tensile strength (Pa) 
0
2
4
6
8 
10 
12 
14 
Aerosil 300 
Aerosil R 812 
Printex 95 
Aerosil OX 50 
Aerosil 200 
Aerosil R 805 
Printex G 
Printex 25 
Titanium dioxide P 25 
Titanium dioxide T 805 
Aluminum oxide C

916 PHARMACEUTICAL PREFORMULATION 
used to reduce the particle size because comminution or grinding are not practical 
due to the length of time required [18] . 
There are numerous methods of particle size reduction, but their application is 
dependent on the intended particle size, particle distribution, cleaning convenience, 
operating cost, dust containment, temperature, and fl exibility. The type of milling 
machine includes slurry, fl uid energy (jet), universal, cone, and hammer. This size 
reduction process may reduce the risk of dissolution rate - limited bioavailability of 
drugs. Particle size reduction can be accomplished by using a hammer mall or a 
similar mill, but this process may only break up the larger crystal aggregates without 
signifi cantly changing the distribution of smaller particle sizes. On the other hand, 
air jet mills, which impinge two streams of particles at a right angle to each other 
in high - velocity air streams, reduce particle size signifi cantly within microsized 
ranges. 
Ball milling was the most commonly used at the preformulation stage to reduce 
the particle size of small amounts of a compound. Ball mills reduce the size of particles 
through a combined process of impact and attrition. Usually they consist of a 
hollow cylinder that contains balls of various sizes which is rotated to initiate the 
grinding process. Micronized particles are typically less than 10 . m in diameter. The 
effi ciency of the milling process is affected by rotation speed, number of balls, mill 
size, wet or dry milling, amount of powder, and length of time of milling. Although 
ball milling can effectively reduce the particle size of compounds, prolonged milling 
may be detrimental in terms of changes of compound crystal form from crystalline 
to polymorphic or amorphous form and stability. 
Although ball milling on a large scale is possible, hammer milling is more preferable 
in the pharmaceutical industry. Powder is bled into the mill house via the 
hopper, and the rotating hammers impact with the powder. When this is fi ne 
enough to pass the screen, the powder will exit. The powder exiting will have a 
maximum particle size of that of the screen. The average particle size of the milled 
powder will be smaller, the smaller the feed rate, the more rapid the milling 
speed (rpm ’ s), and the fi ner the screen. The knife has a blunt edge on one side and 
a knife edge on the other. Milling with the blunt edge forward gives rise to a smaller 
average particle size. The usual minimum particle range is about 50 . m. Large, 
heavy - duty hammer mills (micropulverizers) give much smaller particle size further 
down, typically to 20 . m. If particle sizes less than 5 . m are desired, micronizers are 
used. 
If particle sizes in the micrometer range are required, then the attrition mill can 
be used. Here particles are bled into a chamber in which great turbulence has been 
created by two inlets of air at different pressure. The particles hit one another and 
are removed by centrifugal means and collected in a cyclone setup or in a bag above 
the cyclone. Such particles become highly electrically charged during the operation 
and also become very cohesive. 
The milling process provides energy to the powders so that melting occurs if their 
melting point is suffi ciently low. There is no guarantee that the original polymorph 
and habit will be regained upon resolidifi cation. The milling and micronization 
process can also reduce the particle size of poorly soluble drugs so that the maximum 
surface area is exposed to enhance the solubility and dissolution properties. Although 
micronization of the drug offers the advantage of a small particle size and a lager 
surface area, it can also result in processing problems due to high dust, low density, 

POWDER CHARACTERISTICS 917 
and poor fl ow properties. Indeed, micronization may be counterproductive, since 
the micronized particles may aggregate, which may decrease the surface area. Milling 
and micronization process also induce the changes of crystallinity of drug into 
amorphous form. It has been shown that the amorphous change of the crystalline 
structure can be achieved by vapor condensation, supercooling of melt, rapid precipitation 
from solution, and mechanical applications of a crystalline mass by milling 
or compaction [26] . 
Particle size reduction of the powder has produced defects on the surface that, 
if enough energy is imparted, leads to amorphous regions on the surface. In turn, 
these regions are found to have a greater propensity to adsorb water. The dissolution 
rate increases as the particle size of drug powder decreases due to its greater 
surface area for wetting. 
It is known that ball milling and other types of milling can change the morphology 
of a solid, for example, make a crystalline compound amorphous, increase the surface 
energy of a solid, and distort the crystal lattice. For example, the crystalline solid state 
of sorbitol exhibits a complex polymorphism made of fi ve different forms called A - , 
B - , . - , . - , and E - sorbitol [27] . The structures of these polymorphs have been identifi ed 
by single - crystal X - ray diffraction. The structure of the . form is also the most stable 
and the most common polymorph of sorbitol. It was reported that the . form of sorbitol 
underwent a complete gransformation toward the A form upon ball milling and 
also was affected by milling time. The DSC and XRPD patterns are useful in identifying 
these phenomena, althought the low level of amorphous character cannot be 
detected by techniques such as XRPD and DSC. Crystal structures by the milling 
process as well as compaction forces have been shown [2] . 
6.1.4.8 Compaction(Compressibility) 
Compressibility is the property of forming a stable and intact mass when pressure 
is applied. The manufacture of tablets involves the process of powder compaction 
or compressibility, the purpose of which is to convert a loose incoherent mass of 
powder into a single solid object. A protocol to examine the compression properties 
of fl owing powder should be considered by the formulation scientist when selecting 
the excipient, the formulation type, and the manufacturing process, for example, 
direct compression or granulation for the intended solid dosage form. Acetaminophen 
is poorly compressible whereas lactose compresses well. In general, drug and 
excipients, including lubricant, are blended in a tumbler mixer for a period of time 
and then compressed into tablets in a hydraulic press . The crushing strength is also 
determined to test the compressibility of tablet at room temperature. 
The compression of drug powder may change the crystal structure into a polymorphic 
form — likewise in the milling process. The PXRD patterns and DSC of a 
drug can detect these phenomena. Knowledge of the crystal structure of a drug is 
a prerequisite for compaction. 
To fully understand the compaction behavior of a material, it is necessary to be 
able to quantify of its elasticity, plasticity, and brittleness. A powder in a container 
subjected to compressive force will undergo particle rearrangement [28] . The density 
of the bed will increase with increasing pressure at a characteristic rate. Brittle 
materials will undergo fragmentation, and the fi ne particles formed will percolate 
through the bed to give secondary packing. Plastically deforming materials will 

918 PHARMACEUTICAL PREFORMULATION 
distort to fi ll voids and may also exhibit void fi lling by percolation. When the limit 
of plastic deformation is reached , fracture occurs. 
Many of the basic principles of compaction and the test methodologies are currently 
employed in pharmaceutical formulation. To characterize the compaction 
properties of a material or formulation, it must be possible to measure the relationship 
between the force applied to a powder bed and the volume of the powder bed. 
There are two principal types of compaction studies used to characterize materials: 
pressure – volume relationships and pressure – strength relationships. While ultimately 
it is the strength of a tablet that is important, the pressure – volume relationships 
provide information about the compaction properties of a material that allows an 
appropriate formulation to be developed. The instrumented tableting machine provides 
information for compaction that is directly relevant to production conditions. 
The compression profi les differ from those of rotary tableting machines used for 
commercial production. The profi le of a single punch involves the powder bed being 
compressed between a moving upper punch and a stationary lower punch, while on 
a rotary machine, both punches move together simultaneously. A major advantage 
of instrumented machines is that they provide information not only on the compaction 
properties but also on fl ow and lubrication. The disadvantage of using instrumented 
rotary machines is the large quantity of materials required. 
A large number of equations have been proposed to describe the relationship 
between pressure and volume reduction during the compaction process. The most 
widely used equation to describe the compaction of powders is the Heckel equation. 
Pharmaceutical powders do not produce perfect straight lines, and the type of deviation 
provides information about the compaction behavior of the material. A typical 
Heckel plot for a pharmaceutical powder is obtained showing a straight - line portion 
over a certain pressure range with a negative deviation at low pressures and a positive 
deviation at high pressures. The strength of tablets has traditionally been determined 
in terms of the force required to fracture a specimen across its diameter. The 
fracture load obtained is usually reported as a hardness value. Initially, most materials 
will demonstrate an increase in tensile strength proportional to the compaction 
pressure applied. As the compaction pressure increases, the tablet approaches zero 
porosity, and large increases in pressure are required to achieve small volume reductions 
[28] . Some materials will attain a maximum strength, and subsequent increases 
in pressure will produce weaker tablets. Other materials will also display an initial 
increase in strength proportional to the applied pressure, but the strength reaches 
a maximum before falling off sharply, resulting in capping or lamination in tablet 
production [28] . Figure 13 shows the correlation of hardness with compression pressure. 
Capping is the partial or complete removal of the crown of a tablet from 
the main body, while lamination is the separation of a tablet into two or more distinct 
layers. If the compressibility and fl ow of powders are good, it is possible to 
directly compress the powders into tablet. If either compressibility or fl ow is not 
satisfactory, roller compact, slugging, or we granulation can be utilized to improve 
compressibility. 
6.1.4.9 Surface Area and Other Properties 
The surface area of particles is related to particles size, as discussed previously. 
The surface area of powders affects the drug dissolution rate, powder fl ow, cohesiveness, 
and adsorption. Furthermore, the surface area of solid materials may also 

infl uence the physicochemical properties, adsorption, dissolution, and bioavailability 
of drugs. 
The particle size and surface area distributions of pharmaceutical powders can be 
obtained by microcomputerized mercury porosimetry. Mercury porosimetry gives 
the volume of the pores of a powder, which is penetrated by mercury at each successive 
pressure; the pore volume is converted into a pore size distribution. Two other 
methods, adsorption and air permeability, are also available that permit direct calculation 
of surface area. In the adsorption method, the amount of a gas or liquid solute 
that is adsorbed onto the sample of powder to form a monolayer is a direct function 
of the surface area of the sample. The air permeability method depends on the fact 
that the rate at which a gas or liquid permeates a bed of powder is related, among 
other factors, to the surface area exposed to the permeant. The determination of 
surface area is well described by the BET (Brunauer, Emmett, and Teller) equation. 
The wetting behavior of powders is an also important factor for drug dissolution. 
If the wetting is not satisfactory, hydrophilic excipients (lactose) and surfactant 
(sodium lauryl sulfate or polysorbate) are combined in the powder mixtures. The 
contact angle is used as an index of wetting. The lower the contact angle, the better 
the wetting occurs. 
The general appearance of a tablet with good visual identity and overall 
“ elegance ” are essential for consumer acceptance, quality control of lot - to - lot uniformity, 
general tablet - to - tablet uniformity, and monitoring trouble - free manufacturing. 
Control of the general appearance of a tablet involves the measurement 
of a number of attributes, such as a tablet ’ s size, shape, color, presence or absence 
of and odor, taste, surface texture, physical fl aws and consistency, and legibility of 
any identifying markings. 
6.1.5 TABLET CHARACTERIZATION 
In order to formulate the optimal tablet, various properties should be considered, 
including drug – excipient compatibility, fl owability, lubricity, appearance, dissolution, 
and disintegration [2] . The prepared tablet must also meet physical specifi cation and 
FIGURE 13 Correlation of compression pressure with hardness. 
Pressure (kP) 
Hardness (kP) 
Capping pressure (kP) 
5000 7000 2000 2 
12 
6
TABLET CHARACTERIZATION 919

920 PHARMACEUTICAL PREFORMULATION 
quality standards according to the monograph of the pharmacopeia. In general, 
weight and its variation, content uniformity, thickness, hardness, friability disintegration, 
and dissolution should be considered for tablet validation [3] . These factors 
must be controlled during tablet production (in - process control) and are validated 
after the production to ensure the quality standards. 
6.1.5.1 Disintegration 
Immediate - release tablets should be readily disintegrated in the stomach when 
swallowed. This disintegration involves bursting apart the compact masses by 
aqueous fl uid penetrating the fi ne pore structure of tablet. Disintegration testing is 
an important part of in - process control testing during production to ensure batch - 
to - batch uniformity, but its role in end - product testing has largely been superseded 
by dissolution testing because recently modifi ed - release preparations are getting 
popular. It was recognized in the 1940s that tablets had to disintegrate in order for 
them to be bioavailable due to lack of biopharmaceutical information and primarily 
analytical limitations. Later, of course, in the 1950s and 1960s, the pharmaceutical 
scientist became aware of the importance of dissolution rates as well. 
In general, for the medicinal agent in a tablet to become fully available for 
absorption, the tablet must fi rst disintegrate and discharge the drug to body fl uids 
for dissolution. The general manner in which a tablet disintegrates is as follows: 
(a) the tablet wets down, (b) the dissolution liquid penetrates the pore space, (c) 
the disintegrant absorbs water and swells, and (d) this swelling causes the tablet to 
break down into granules. Figure 14 shows the disintegration pathways of solid 
dosage forms for the dissolution and absorption of drugs. After the disintegration 
process, the solid dosage forms change into granules or smaller and fi ne particles 
ready for dissolution and absorption in the fl uid. 
FIGURE 14 Disintegration and dissolution pathways of solid dosage forms for absorption 
of drug. 
Solid 
dosage form 
Drug in solution 
Granules 
or 
aggregates 
Primary 
drug particles 
Disintegration Deaggregation 
Dissolution 
(major) Dissolution 
(major) 
Dissolution 
(major) 
Absorption 
Drug in body fluids and tissues

Tablet disintegration is the important fi rst step in the dissolution of the drug 
substance contained in immediate - release tablets but dissolution is more meaningful 
in case of many modifi ed release products rather than disintegration . A number 
of formulation and manufacturing factors can affect the disintegration and dissolution 
of a tablet, including the particle size of the drug substance in the formulation; 
the solubility and hygroscopicity of the formulation; the type and concentration of 
the disintegrant, binder, and lubricant used; the manufacturing method, particularly 
the compactness of the granulation and the compression force used in tableting; and 
the in - process variables which may occur. Therefore, it is vitally important for batch - 
to - batch consistency to establish disintegration and dissolution standards and controls 
for both materials and processes. 
Tablet disintegration also is important for those tablets containing medicinal 
agents (such as antacids and antidiarrheals) that are not intended to be absorbed 
but rather to act locally within the GI tract. In these instances, tablet disintegration 
provides drug particles with an increased surface area for localized activity within 
the GI tract. 
It is evident that there are some correlations of physical parameters with tablet 
disintegration time. Figure 15 shows the correlation of water penetration force, disintegration 
force, disintegrant contents, and compression forces of the tablet with 
disintegration time of the tablet. As the water penetration force increases, the tablet 
disintegration force also increases, resulting in shorter disintegration time [29, 30] . 
The amount of disintegrant in the tablet also decreases the disintegration time. 
The disintegration time increases as tableting pressure increases below the critical 
capping pressure [18] . At very low pressures the penetration of liquid into the tablet 
is virtually unhindered (almost like pouring water into a breaker) but the pores will 
be too large to allow disintegrant swelling to cause stress and the disintegration time 
will decrease. Once the pores are suffi ciently small, penetration of the liquid into 
the disintegrant becomes the limiting step, and the disintegration time will increase 
FIGURE 15 Correlation of physical parameters on tablet disintegration time. 
Water penetration force or disintegration force Disintegration time 
Tablet compression force 
Amount of disintegrants 
TABLET CHARACTERIZATION 921

922 PHARMACEUTICAL PREFORMULATION 
with increasing pressure. Disintegrants and lubricants are added to wet - granulated 
products after the granulation has been dried. Disintegration time increases as the 
amount of hydrophobic lubricant increases. The mixing time for the lubricant must 
be kept short because otherwise the lubricant may fl uidize during the mixing 
step and lose part of the lubricant properties that are necessary for fl ow in the 
tablet die. If the disintegration is not satisfactory, numerous types of disintegrants 
are added in the tablet formulations, including starch, croscarmellose sodium, sodium 
starch glycolate and crospovidone known as superdisintegrants. In general, the 
swelling rate and water uptake are the most important properties of disintegrants. 
All USP tablets must pass a test for disintegration, which is conducted in vitro 
using a testing apparatus. The detailed monograph for disintegration testing is 
described in the many pharmacopeias [2, 3] . 
The apparatus consists of a basket - rack assembly containing dimensions held 
vertically upon a 10 - mesh stainless steel wire screen. During testing, a tablet is 
placed in each of the six tubes of the basket, and the mechanical device raises and 
lowers the basket in the immersion fl uid at a frequency of between 29 and 32 cycles 
per minute, the wire screen always maintained below the level of the fl uid. For 
uncoated tablets, buccal tablets, and sublingual tablets, water maintained at about 
37 ° C is used as the immersion fl uid unless another fl uid is specifi ed in the individual 
monograph. For these tests, complete disintegration is defi ned as that state in which 
any residue of the unit, except fragments of insoluble coating or capsule shell, 
remaining on the screen of the test apparatus is a soft mass having no palpably fi rm 
core. Buccal tablets must disintegrate within the time set forth in the individual 
monograph, usually 30 min, but varying from about 2 min for nitroglycerin tablets 
to up to 4 h. If one or more tablets fail to disintegrate, additional tests prescribed 
by the USP must be performed. Enteric - coated tablets are also similarly tested, 
except that the tablets are permitted to be tested in simulated gastric fl uid for 1 h 
with no sign of disintegration, cracking, or softening. They are then switched to the 
simulated intestinal fl uid for the time stated in the individual monograph during 
which time the tablets disintegrate completely. If 1 or 2 of the 6 tablets fails to disintegrate 
completely, disintegration testing is repeated on 12 additional tablets, and 
not less than 16 of the total 18 tablets tested must disintegrate to meet the 
standards. 
6.1.5.2 Dissolution 
Defi nitions Dissolution is the dynamic process by which drug is dissolved in a 
solvent (water) and is characterized by a rate (amount dissolved per unit time). In 
vitro dissolution testing of a tablet is very important for many reasons: It guides the 
formulation and product development process toward optimization of dosage forms 
for quality control and reliability. By conducting dissolution studies in the early 
stages of a product ’ s development, the formulation compositions and manufacturing 
parameters are also tuned and monitored about the old and newly advanced tablet. 
The U.S. Food and Drug Administration (FDA) allows manufacturers to examine 
scale - up batches of 10% of the proposed size of the actual production batch or 
100,000 dosage units, whichever is greater, by performing in vitro dissolution testing 
to assure bioequivalence from batch to batch and processing parameters. New drug 
applications (NDAs) submitted to the FDA contain in vitro dissolution data generally 
obtained from batches that have been used in pivotal clinical and/or bioavail

ability studies and from human studies conducted during product development. 
Once the specifi cations are established in an approved NDA, they become offi cial 
(USP) specifi cations for all subsequent batches and bioequivalent products. The 
dissolution testing is also used as a tool of SUPAC (scale - up postapproval change) 
and variation of equipment, location, and processing factors. In addition, dissolution 
testing is used as a tool to examine the short - and long - term stability of dosage 
forms. Release mechanism and parameters which change the dissolution are also 
studied. Most of all, one of main goals of in vitro dissolution testing is to provide 
reasonable prediction and correlation with the product ’ s in vivo bioavailability. Differentiations 
in the formulations and other related variables may cause deviations 
from in vivo bioavailability data. 
A system has been developed which relates combinations of a drug ’ s solubility 
(high or low) and its intestinal permeability (high or low) as a possible basis for 
predicting the likelihood of achieving a successful in vivo – vitro correlation (IVIVC). 
The four classes based on BCS are (I): high solubility and high permeability, (II): 
low solubility and high permeability, (III): high solubility and low permeability, and 
(IV): low solubility and low permeability. In class I, dissolution testing can be used 
as a prognostic tool to predict in vivo biovailability. 
Equations Many dissolution equations are well described in the text. Most of the 
equation are based on the well - known Fick ’ s law. Figure 16 shows a diagram of the 
concentration gradient between the matrix tablet and bulk fl uid for dissolution. 
From these gradient situations, the well - known Noyes – Whitney equation is 
given as 
dc 
dt 
DAK 
Vh 
C C = . ( ) s t 
where dc / dt = rate of drug dissolution, where dQ / dt = V dc / dt 
V = volume of dissolution fl uid 
D = diffusion rate constant 
A = surface area of dosage forms 
C s = concentration of drug in stagnant layer 
FIGURE 16 Concentration gradient from the tablet between matrix and bulk fl uid for 
dissolution. Cs, drug solubility; C, uniform concentration; h, thickness of stagnant fi lm; X = 
diffusional path length. 
Tablet Stagnant layer Bulk fluid 
Concentration 
Cs 
C 
Matrix 
X = 0 X = h 
TABLET CHARACTERIZATION 923

924 PHARMACEUTICAL PREFORMULATION 
C t = concentration of drug in bulk fl uid at given time 
K = partition coeffi cient 
h = thickness of stagnant layer 
If the bulk volume is large and the concentration of drug in the fl uid is much 
lower than the drug solubility ( C s > > C t ), it is regarded as a sink condition. In this 
case, the equation is much simpler and the dissolution behaviors continuously occur 
because the chemical potential ( C s . C t ) approximates drug solubility ( C s ). 
In a matrix tablet, the following Higuchi equations are given depending on the 
polymeric structures of the homogenous and porous matrix [20] : 
Q 
D A C C 
D A C C 
= 
. [ ] 
. [ ] 
( ) 
( ) 
2
2 
1 
1 
S S 
/2 
S S 
/2 
t for homogenousmatrix 
t / f . . . or porousmatrix 
... 
In the dissolution of granular powders, the Hixson – Crowell equation is also 
established as 
Q Q kt 0 
1 1/3 /3 for granular powders . = 
Testing Method In addition to formulation and manufacturing controls, the method 
of dissolution testing also must be controlled to minimize important variables such 
as paddle rotational speed, vibration, and disturbances by sampling probes. The 
USP includes seven apparatus designs for drug release and dissolution testing of 
immediate - release oral dosage forms, extended - release products, enteric - coated 
products, and transdermal drug delivery devices: 
Apparatus I: rotating basket method, 25 – 150 rpm (100 rpm) 
Apparatus II: rotating paddle method, 25 – 150 rpm (50 rpm) 
Apparatus III: reciprocating cylinder method, inner tube (5 – 40 dips/min), outer 
tube (300 mL) 
Apparatus IV: fl ow - through method: 4 – 16 mL/min 
Apparatus V: paddle over disc 
Apparatus VI: cylinder method 
Apparatus VII: reciprocating holder method 
Detailed guidelines for dissolution testing are described in monographs of many 
pharmacopeias. The USP apparatus I and USP apparatus II are used principally for 
tablet dissolution testing. In USP apparatus I, the dosage unit is placed inside the 
basket. In USP apparatus II, the dosage unit is placed on the bottom in the vessel. 
In each test, a volume of the dissolution medium (500 – 900 mL in general) is placed 
in the vessel and allowed to come to 37 ± 0.5 ° C. Then the stirrer is rotated at the 
specifi ed speed (50 – 200 rpm). The samples of the medium are withdrawn for analysis 
of the proportion of drug dissolved. The tablet or capsule must meet the stated 
monograph requirement for rate of dissolution. For example, “ not less than 85% of 
the labeled amount is dissolved in 30 minutes in case of immediate release tablet. ” 
In a fl oating tablet, the sinker can be used in the paddle method. 

Variables Affecting Dissolution In general, the dissolution profi les are highly 
dependent on the physicochemical properties, formulation, processing parameters, 
and testing conditions [2] . The physicochemical properties include particle size, 
surface area, crystal habit and polymorphism, solubility, molecular size, salt formation, 
p K a , hydration, wetting, and surface tension. Physical factors such as viscosity, 
density, fl occulation, and agglomeration are also considered. The formulation factors 
are also of importance. The amount and type of excipients and type of dosage forms 
play a key role in modifying dissolution behaviors. For example, Figure 17 gives the 
effect of lubricant and its mixing time on the dissolution rate of drugs. The presence 
of lubricant and its mixing time signifi cantly changed tablet dissolution [31] . For 
poorly soluble drug, numerous pharmaceutical methods have been utilized to 
increase the dissolution rate of drug, including micronization, amorphous crystallization, 
spray drying, inclusion complex, microemulsion, and solid dispersion. 
The processing parameters in the tablet preparation also change the dissolution, 
including temperature, mixing, milling, rotation speed, solvent, hardness, and surface 
area. The testing conditions are also important in modifying the dissolution of tablet. 
Therefore, the testing conditions are well defi ned by the regulations in the many 
pharmacopeias. The testing conditions include pH of the fl uid, temperature, ionic 
strength, common ion effect, type of apparatus, rotation speed, volume size of 
dissolution fl uid, analytical conditions, aeration, sample treatment, and mainly 
composition of dissolution media. Table 17 provides some of the media compositions 
suggested for in vitro dissolution testing of the tablet. These modifi ed 
dissolution media can be used to achieve dissolution of drug under simulated in 
vivo conditions. 
In Vitro – In Vivo Correlation In vitro dissolution testing can provide a reasonable 
prediction of the product ’ s in vivo bioavailability. For a high - solubility and high - 
permeability drug (class I), an IVIVC may be expected if the dissolution rate is 
slower than the rate of gastric emptying (the rate - limiting factor). In a low - solubility 
and high - permeability drug, drug dissolution may be the rate - limiting step for 
drug absorption and an IVIVC may be expected. In a high - solubility and low - 
FIGURE 17 Effect of lubricant and its mixing time on dissolution rate of drugs. 
Time 
Percent dissolved 
No lubricant 
Lubricant, 2 min 
Lubricant, 30 min 
TABLET CHARACTERIZATION 925

926 PHARMACEUTICAL PREFORMULATION 
permeability drug, permeability is the rate - controlling step and only a limited IVIVC 
may be possible. In a drug with low solubility and low permeability, signifi cant 
problems would be likely for oral drug delivery. 
However, the in vivo GI condition is very complicated in terms of complex physiology 
and absorption process and is not simulated by the simple in vitro dissolution 
conditions [32] . Moreover, in vivo conditions are also complicated by food composition, 
type and composition of dosage formulations, relative rates of permeation, GI 
transit time, site of absorption, complexity of GI fl uids such as pH, enzymes, bile 
and mucin, rate and capacity of metabolism by intestinal and hepatic enzymes, 
ethical difference, and patient conditions such as mood, disease, bed rest, and fasting 
volume of fl uid given. For these reasons, in vitro dissolution is not always correlated 
with in vivo absorption, especially low - soluble and low - bioavailable drugs. To use 
dissolution testing as a prognostic tool for in vivo bioavailability, the dissolution 
fl uid is simulated with in vivo GI condition as possible, compromising the biorelevant 
composition of dissolution fl uid (see Table 17 ), gradient pH, proper stirring 
rate, and addition of lipid, enzyme, and surfactants. 
For bioequivalence, dissolution profi les of two tablets are often compared. In 
this case, the difference factor and similarity factor are considered, defi ned by the 
equations 
f 
R T 
R
t t 
t 
1 100 = 
. 
. .
. 
f 
n 
R T t t 2 
2 
0 5 
50 
1 
100 = . ... 
... 
. ... 
... 
. 
. 
log 1+ ( ) 
. 
where R t and T t are the cumulative percents dissolved at each of the selected n time 
points. 
TABLE 17 Suggested Media Compositions for In Vitro Dissolution Testing 
Medium Type Codes Dissolution compositions 
Water W Purifi ed water 
Gastric fl uid 
(pH 1.2) 
SGF HCl – NaCl buffer 
SGF/TW HCl – NaCl buffer/Tween 80 (1%) 
SGF/SLS HCl – NaCl buffer/sodiun lauryl sulfate (1%) 
SGF/PEP HCl – NaCl buffer/pepsin (0.32%) 
Intestinal fl uid 
(pH 6.8) 
SIF KH 2 PO 4 buffer 
SIF/TW KH 2 PO 4 buffer/Tween 80 (1%) 
SIF/SLS KH 2 PO 4 buffer/sodiun lauryl sulfate (1%) 
SIF/Pan KH 2 PO 4 buffer/pancreatin (1%) 
SIF/fasted 0.2 M KH 2 PO 4 buffer (3.9 g)/3 m M Na - taurocholate, 75 m M 
lecithin/KCl (7.7 g)/NaOH (qs)/water (qs) 
Intestinal fl uid 
(pH 5.0) 
SIF/fed Acetic acid (8.65 g)/15 m M Na - taurocholate/3.75 m M , lecithin/ 
KCl (15.2 g)/NaOH (qs)/water (qs) 

The difference factor f1 is proportional to the average difference between the two 
profi les while the similarity factor f2 is inversely proportional to the average squared 
difference between the two profi les and measures the closeness between the two 
profi les. The two dissolution profi les are identical if f2 = 100. An average difference 
of 10% at all measured time points results in a f2 value of 50. The FDA guideline 
states that f2 values of 50 – 100 indicate similarity between two dissolution profi les. 
6.1.5.3 Weight Variation 
With a tablet designed to contain a specifi c amount of drug in a specifi c amount of 
tablet formula, the weight of the tablet being made is routinely measured to help 
ensure that the tablet contains the proper amount of drug. The quantity of fi ll placed 
in the die of a tableting press determines the weight of the resulting tablet. The 
volume of fi ll is adjusted to yield tablets of desired weight and content. The depth 
of fi ll in the tablet die must be adjusted to hold a predetermined volume of powder 
or granulation. Each tablet weight should be calculated if the amount of drug and 
other excipients such as diluent, disintegrant, and binder are decided. 
During production, sample tablets are periodically removed for visual inspection 
and automated physical measurement known as in - process control (IPC). The USP 
provides some guidelines for weight variation. In the test, 10 uncoated tablets are 
weighed individually and the average weight is calculated. The tablets are assayed 
and the content of active ingredient in each of the 10 tablets is calculated assuming 
homogeneous drug distribution. 
6.1.5.4 Hardness or Breaking Strength 
Tablets require a certain amount of strength, or hardness and resistance to friability, 
to withstand mechanical shocks of handling in manufacture, packaging, and shipping. 
Adequate tablet hardness and resistance to powdering and friability are necessary 
requisites for consumer acceptance while immediate - release tablets should 
readily disintegrate in the stomach as quick possible. For this reason, the relationship 
of harness to tablet disintegration and drug dissolution has been described. 
Tablet hardness has been defi ned as the force required to break a tablet in a 
diametric compression test. To perform this test, a tablet is placed between two 
anvils, and the crushing strength that just causes the tablet to break is recorded. 
Hardness is thus sometimes termed the tablet crushing strength. It is not unusual 
for a tableting press to exert as little as 3000 lb and as much as 40,000 lb of force in 
the production of tablets. Generally, the greater the pressure applied, the harder the 
tablets, although the formulation composition and manufacturing process may also 
change tablet hardness. In general, tablets should be suffi ciently hard to resist breaking 
during normal handling or transportation but have no problem in disintegrating 
and/or dissolving after swallowing. 
The hardness of a tablet, like its thickness, is a function of the die fi ll and compression 
force. At a constant die fi ll, the hardness values increase and thickness 
decreases as additional compression force is applied. This relationship holds up to 
a maximum value for hardness and a minimum value for thickness beyond which 
increases in pressure cause the tablet to laminate or cap, thus destroying the integrity 
of the tablet. At a constant compression force (fi xed distance between upper 
TABLET CHARACTERIZATION 927

928 PHARMACEUTICAL PREFORMULATION 
and lower punches), hardness increases with increasing die fi lls and decreases with 
lower die fi lls. The amount and mixing time of lubricants and excipients can affect 
tablet hardness. Large tablets require a greater force to cause fracture and are 
therefore “ harder ” than small tablets. 
Special dedicated hardness testers or multifunctional systems are used to measure 
the degree of force (in kilograms, pounds, or arbitrary units) required to break a 
tablet. Devices to test tablet hardness include the Monsanto tester, the Strong - Cobb 
tester, the Pfi zer tester, the Erweka tester, and the Schleuniger tester. A force of 
about 4 kg is considered the minimum requirement for a satisfactory tablet. Multifunctional 
automated equipment can determine tablet weight, hardness, thickness, 
and diameter. Unfortunately, these testers do not produce uniform results for the 
same tablet due to operator variation, lack of calibration, spring fatigue, and manufacturer 
variation. 
6.1.5.5 Friability 
Tablet hardness is not an absolute indicator of tablet strength since some formulations, 
when compressed into very hard tablets, tend to “ cap ” on attrition, losing 
their crown portions. Another measure of a tablet ’ s strength is tablet friability. 
More powders , chips, and fragments can be produced during friability test if the 
tablet lacks proper strength and is manufactured in dirty processes during coating 
and packaging. The high friability causes lacks of elegance and consumer acceptance 
and even weight variation or content uniformity problems. 
A tablet ’ s durability may be determined using friability tester like Varian Friabilator 
testng apparatus . This apparatus determines the tablet ’ s friability, or its tendency 
to crumble, by allowing it to roll and fall within the rotating apparatus. 
Normally, a preweighed tablet sample is placed in the friabilator, which is then operated 
for 100 revolutions. The tablets are then dusted and reweighed. Any loss in 
weight is determined. Resistance to loss of weight indicates the tablet ’ s ability to 
withstand abrasion in handling, packaging, coating, and shipment. Compressed 
tablets that lose a maximum of not more than 0.5 – 1% of their weight are generally 
considered acceptable [3] . 
6.1.5.6 Content Uniformity 
Tablet weight cannot be used as a potency indicator of its potency, except perhaps 
when the active ingredient is 90 – 95% of the total tablet weight. In tablets with 
smaller dosages, a good weight variation does not ensure good content uniformity, 
but a large weight variation precludes good content uniformity. The weight variation 
test would be a satisfactory method of determining the drug content uniformity of 
tablets. The content uniformity of the tablet is more important since the potency of 
tables is expressed on labels in terms of grams, milligrams, or micrograms. The 
content uniformity of tablets can be varied by three factors: (1) nonuniform distribution 
of the drug substance throughout the powder mixture or granulation, 
(2) segregation of the powder mixture or granulation during the various manufacturing 
processes, and (3) tablet weight variation. 
By the USP method, 10 dosage units are individually assayed for their content 
uniformity according to the assay method described in the individual monograph. 

The requirements for content uniformity are met if the amount of active ingredient 
in each dosage unit lies within the range of 85 – 115% of the label claim and the relative 
standard deviation is less than 6.0%. If one or more dosage units do not meet 
these criteria, additional tests as prescribed in the USP are required. Offi cial compendia 
or other standards provide an acceptable potency range around the label 
potency. For highly potent, low - dose drugs such as digoxin, this range is usually not 
less than 90% and not more than 110% of the labeled amount. For most other larger 
dose drugs in tablet form, the offi cial potency range that is permitted is not less than 
95% and not more than 105% of the labeled amount. 
6.1.5.7 Tablet Thickness 
At a constant compressive load, tablet thickness varies with changes in die fi ll, particle 
size distribution and packing of the particle mix being compressed, and tablet 
weight, while with a constant die fi ll, thickness varies with variations in compression 
forces. The thickness of individual tablets may be measured with a micrometer, 
which permits accurate measurement and provides information on the variation 
between tablets. Other techniques employed in production control involve placing 
5 or 10 tablets in a holding tray, where their total crown thickness may be measured 
with a sliding caliper scale. Tablet thickness should be controlled within ± 5% variation 
of a standard value. Any variation in tablet thickness within a particular lot of 
tablets or between manufacturer ’ s lots is not appropriate for consumer acceptance 
of the product. In addition, tablet thickness must be also controlled to facilitate 
packaging. 
6.1.5.8 Tablet Shape and Size 
The shape of the tablet alone can infl uence the choice of tableting machine used. 
Figure 18 shows some representative tablet shapes. The size, shape, and thickness 
FIGURE 18 Example tablet shapes. 
TABLET CHARACTERIZATION 929

930 PHARMACEUTICAL PREFORMULATION 
are very changeable. Shaped tablets requiring slotted and sophisticated punches 
must be run at slower speeds to avoid manufacturing disorder as compared with 
round tablets using conventional punches. Because of the nonuniform forces 
involved during compression, the more convex the tablet surface, the more likely it 
is to cause capping or laminating problems. The more complicated shaped tablet 
requires the use of a slower tableting machine or one with precompression capabilities. 
The size and shape of the tablet are governed by the choice of tableting 
machine, the best particle size for the granulation, production lot sizes, and the best 
type of tablet processing, packaging operation, and cost to produce the tablet. 
REFERENCES 
1. Kibbe , A. H. ( 2000 ), Handbook of Pharmaceutical Excipients , 3rd ed. , American Pharmaceutical 
Association and Pharmaceutical Press , London . 
2. Gibson , M. ( 2001 ), Pharmaceutical Preformulation and Formulation, A Practical Guide 
from Candidate Drug Selection to Commercial Dosage Form , HIS Health Group, Denver, 
CO . 
3. Allen , L. V. , Popovich , N. G. , and Angel , H. C. ( 2005 ), Pharmaceutical dosage forms and 
drug delivery systems , Lippincott Williams & Wilkins , Baltimore, MD, USA. 
4. Mountfi eld , R. J. , Senepin , S. , Schleimer , M. , Walter , I. , and Bittner , B. ( 2000 ), Potential 
inhibitory effects of formulation ingredients on intestinal cytochrome P450 , Int. J. Pharm. , 
211 , 89 – 92 . 
5. Cornaire , G. , Woodley , J. , Hermann , P. , Cloarec , A. , Arellano , C. , and Houin , G. ( 2004 ), 
Impact of excipients on the absorption of P - glycoprotein substrates in vitro and in vivo , 
Int. J. Pharm. , 278 , 119 – 131 . 
6. Wang , S. - W. , Monagle , J. , McNulty , C. , Putnam , D. , and Chen , H. ( 2004 ), Determination 
of P - glycoprotein inhibition by excipients and their combinations using an integrated 
high - throughput process , J. Pharm. Sci. , 93 , 2755 – 2767 . 
7. Karsa , D. R. , and Stephenson , R. A. ( 1995 ), Excipients and Delivery Systems for Pharmaceutical 
Formulations , The Royal Society of Chemistry , Cambridge , pp. 1 – 34 . 
8. Farber , L. , Tardos , G. I. , and Michaels , J. N. ( 2003 ), Evolution and structure of drying 
material bridges of pharmaceutical excipients: Studies on a microscope slide , Chem. Eng. 
Sci. , 58 , 4515 – 4525 . 
9. Cao , Q. - R. , Choi , Y. - W. , Cui , J. - H. , and Lee , B. - J. ( 2005 ), Formulation, release characteristics 
and bioavailability of novel monolithic hydroxypropylmethylcellulose matrix 
tablets containing acetaminophen , J. Controlled Release , 108 , 351 – 361 . 
10. Jackson , K. , Young , D. , and Pant , S. ( 2000 ), Drug - excipient interactions and their affect 
on absorption , PSTT , 3 ( 10 ), 336 – 345 . 
11. Patel , H. , Stalcup , A. , Dansereau , R. , and Sakr , A. ( 2003 ), The effect of excipients on the 
stability of levothyroxine sodium pentahydrate tablets , Int. J. Pharm. , 264 , 35 – 43 . 
12. Verma , R. K. , and Garg , S. ( 2004 ), Compatibility studies between isosorbide mononitrate 
and selected excipients used in the development of extended release formulations , 
J. Pharm. Biomed. Anal. , 35 , 449 – 458 . 
13. Young , W. R. ( 1990 ), Accelerated temperature pharmaceutical product stability determinations 
, Drug Dev. Ind. Pharm. , 16 ( 4 ), 551 – 569 . 
14. Waterman , K. C. , and Adami , R. C. ( 2005 ), Accelerated aging: Prediction of chemical 
stability of pharmaceuticals , Int. J. Pharm. , 293 , 101 – 125 . 

15. Simon , P. , Veverka , M. , and Okuliar , J. ( 2004 ), New screening method for the determination 
of stability of pharmaceuticals , Int. J. Pharm. , 270 , 21 – 26 . 
16. Florence , A. T. and Attwood , D. ( 1998 ), Physicochemical principles of pharmacy , 3rd ed. , 
Macmillan , London , pp. 5 – 35 , 101 – 151. 
17. Hiestand , E. N. ( 1989 ), The basis for practical applications of the tableting indices , Pharm. 
Technol. Int. , 8 , 54 – 66 . 
18. Carstensen , J. T. ( 1993 ), Pharmaceutical Principles of Solid Dosage Forms , Technomic 
Publishing , Lancaster, PA . 
19. Brittain , H. G. ( 1995 ), Physical Characterization of Pharmaceutical Solids , Marcel Dekker , 
New York . 
20. Sinko , P. J. ( 2006 ), Physical Pharmacy and Pharmaceutical Sciences , Lippincott Williams 
and Wilkins , Baltimore, MD . 
21. Carson , J. W. and Wilms , H. ( 2006 ), Development of an international standard for shear 
testing , Powder Technol. , 167 , 1 – 9 . 
22. Hedenus , P. , Mattsson , M. S. , Niklasson , G. A. , Camber , O. , and Ek , R. ( 2000 ), Characterization 
of instantaneous water absorption properties of pharmaceutical excipients , Int. J. 
Pharm. , 141 , 141 – 149 . 
23. Hasegawa , S. , Hamaura , T. , Furuyama , N. , Kusai , A. , Yonemochi , E. , and Terada , K. ( 2005 ), 
Effects of water content in physical mixture and heating temperature on crystallinity of 
troglitazone - PVP K30 solid dispersions prepared by closed melting method , Int. J. Pharm. , 
302 , 103 – 112 . 
24. Mullarney , M. P. , Hancock , B. C. , Carlson , G. T. , Ladipo D. D. , and Langdon , B. A. ( 2003 ), 
The powder fl ow and compact mechanical properties of sucrose and three high - intensity 
sweeteners used in chewable tablets , Int. J. Pharm. , 257 , 227 – 236 . 
25. Meyer , K. , and Zimmermann , I. ( 2004 ), Effect of glidants in binary powder mixtures , 
Powder Technol. , 139 , 40 – 54 . 
26. Hancock , B. C. , and Zografi , G. ( 1997 ), Characteristics and signifi cance of the amorphous 
state in pharmaceutical systems , J. Pharm. Sci. , 86 ( 1 ), 1 – 12 . 
27. Willart , J. , Lefebvre , J. , Dan e de , F. , Comini , S. , Looten , P. , and Descamps , M. ( 2005 ), Polymorphic 
transformation of the G - form of d - sorbitol upon milling: Structural and nanostructural 
analyses , Solid State Communi. , 135 ( 8 ), 519 – 524 . 
28. Adolfsson , A. , and Nystrom , C. ( 1996 ), Tablet strength, porosity, elasticity and solid state 
structure of tablets compressed at high loads , Int. J. Pharm. , 132 , 95 – 106 . 
29. Caramella , C. , Colombo , P. , Conte , U. , Ferrari , F. , and Manna , A. L. ( 1986 ), Water uptake 
and disintegration force measurements: Towards a general understanding of disintegration 
mechanism , Drug Dev. Ind. Pharm. , 12 , 1749 – 1766 . 
30. Pourkavoos , N. , and Peck , G. E. ( 1993 ), The effect of swelling characteristics of superdisintegrants 
on the aqueous coating solution penetration into the tablet matrix during the 
fi lm coating process , Pharm. Res. , 10 ( 9 ), 1363 – 1371 . 
31. Lerk , C. F. , Bolhuis , G. K. , Smallenbroek , A. J. , and Zuurman , K. ( 1982 ), Interaction of 
tablet disintegrations and magnesium stearate during mixing II. Effect on dissolution rate , 
Pharm. Acta Helv , 57 , 282 – 286 . 
32. Gundert - Remy , U. , and Moller , H. ( 1990 ), Oral Controlled Release Products, Therapeutic 
and Biopharmaceutical Assessment , Wissenschaftliche Verlagsgesellschaft mbH , Stuttgart, 
Germany , pp. 155 – 173 . 
REFERENCES 931


933 
6.2 
ROLE OF PREFORMULATION IN 
DEVELOPMENT OF SOLID 
DOSAGE FORMS 
Omathanu P. Perumal and Satheesh K. Podaralla 
South Dakota State University, Brookings, South Dakota 
Contents 
6.2.1 Introduction 
6.2.2 Physical/Bulk Characteristics 
6.2.2.1 Crystallinity and Polymorphism 
6.2.2.2 Hydrates/Solvates 
6.2.2.3 Amorphates 
6.2.2.4 Hygroscopicity 
6.2.2.5 Particle Characteristics 
6.2.2.6 Powder Flow and Compressibility 
6.2.3 Solubility Characteristics 
6.2.3.1 p Ka and Salt Selection 
6.2.3.2 Partition Coeffi cient 
6.2.3.3 Drug Dissolution 
6.2.3.4 Absorption Potential 
6.2.4 Stability Characteristics 
6.2.4.1 Solid - State Stability 
6.2.4.2 Solution - State Stability 
6.2.4.3 Drug – Excipient Compatibility 
6.2.5 Conclusions 
References 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

934 ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS 
6.2.1 INTRODUCTION 
The advent of combinatorial chemistry and high - throughput screening (HTS) has 
exponentially increased the number of compounds synthesized and screened during 
the drug discovery phase. However, the overall effi ciency of the drug discovery 
process is still exceedingly low (only 1 in 10,000 makes it to the market). Drug discovery 
is mostly driven by “ activity screens ” with little emphasis on “ property 
screens. ” This is exemplifi ed by the fact that 40% of attrition in drug discovery and 
development is attributed to poor biopharmaceutics and pharmacokinetic properties 
[1] , which in turn are related to poor physicochemical properties. As a result, 
pharmaceutical companies have started to redesign their strategies by including 
property screens quite early in the discovery stage [2] . Preformulation is the study 
of fundamental properties and derived properties of drug substances. In other 
words, preformulation is the fi rst opportunity to learn about the drug ’ s physicochemical 
properties from the perspective of transforming a biologically active molecule 
to a “ druggable ” molecule. The type and extent of preformulation activities 
vary in a discovery and generic setting. 
The main goal of a drug discovery program is to develop an orally deliverable 
molecule for obvious reasons of ease of manufacture and convenience of drug 
administration. More than 75% of the drug products in the market are oral formulations, 
of which more than half are solid dosage forms [3] . The “ rule of fi ve ” developed 
by Christopher Lipinski [4] is one of the “ physicochemical screens ” to weed 
out molecules with poor physicochemical properties very early in the drug discovery 
process. According to Lipinski ’ s rule, a drug will show poor oral absorption if it does 
not conform to any of the two physicochemical requirements listed in Table 1 . The 
rule of fi ve is applicable only to small molecules and it relates the chemical properties 
of the drug to its solubility and permeability characteristics. During the initial 
stages of drug discovery, the preformulation activities are mainly focused on developing 
a water - soluble compound for early activity studies and preclinical testing in 
animals. At this stage, the preformulation scientist is faced with the challenge of 
working with a limited quantity of compound (few milligrams) for testing a long list 
of physicochemical parameters. On the other hand, developing preclinical formulations 
can be quite a daunting task given the fact that toxicological studies require 
a high dose of drug (10 – 100 times above the effective dose) to be delivered in a 
small volume of the formulation. Preformulation activities increase as the molecule 
proceeds through the development phase. The “ discovery and development phar- 
TABLE 1 Lipinski Rule of Five for Orally Active Compounds 
Physicochemical Parameter Lipinski rule 
Molecular weight Not more than 500 Da 
log P Not more than 5 
Hydrogen bond donors Not more than 5 hydrogen bond donors expressed as the 
sum of OH ’ s and NH ’ s 
Hydrogen bond acceptors Not more than 10 expressed as the sum of OH ’ s and NH ’ s 

maceutics ” documentation forms a signifi cant portion of the investigational new 
drug application (IND) application and new drug application (NDA) fi led to the 
U.S. Food and Drug Administration (FDA). In a generic setting, preformulation 
studies are mainly focused on developing a formulation that is bioequivalent to the 
innovator ’ s product with the main objective of fi ling an abbreviated new drug application 
(ANDA). A strong preformulation team can generate intellectual property 
in the form of new salts, solid - state forms, or new stabilized formulations of the drug 
for an innovator and/or a generic manufacturer. 
In the present chapter, the discussion is mainly focused on preformulation testing 
for oral solid dosage forms in a drug discovery setting. The chapter address the following 
goals of preformulation: (i) to gain knowledge about the physicochemical 
characteristics of the drug, (ii) to defi ne the physical characteristics of the drug, (iii) 
to understand the stability characteristics of the drug, and (iv) to determine the 
compatibility of excipients with the drug. In this chapter, we have grouped all 
the parameters under three sections and discussed in a logical sequence for the 
convenience of the reader. 
6.2.2 PHYSICAL/BULK CHARACTERISTICS 
The bulk or physical characteristics of a drug substance are mainly dictated by its 
solid - state properties. Purity of the drug substance is a fundamental property that 
is characterized at the beginning of preformulation studies. In the initial stages of 
drug development, the drug is usually not very pure. Nevertheless, it is essential to 
know the purity of the material at hand using simple measurements such as melting 
point. This would serve to set drug specifi cations during later stages of drug development. 
Differential scanning calorimetry (DSC) requires very little sample (1 – 5 mg) 
and is a useful tool to estimate the purity of the compound. The drug sample is 
heated in a crucible, where the difference in heat between the sample and a reference 
crucible is seen as an endotherm or exotherm in the thermogram depending 
on whether heat is taken up or given up, respectively, by the sample. The integrated 
area under the endotherm or exotherm gives a measure of the heat or enthalpy 
involved in this process. Melting is seen as an endothermic event and the purity of 
the sample will govern the peak position, shape, sharpness, and heat of fusion ( .Hf ). 
DSC is sensitive in detecting impurities to the extent of 0.002 mol % [5] . The DSC 
fi ndings should be substantiated by a stability - indicating high - performance liquid 
chromatography (HPLC) assay. On the other hand, thin - layer chromato graphy 
(TLC) may be used to qualitatively detect the number of impurities in the drug 
sample. Impurity profi ling is an important aspect of the drug development process, 
particularly for optimizing the synthetic process. The impurities can originate from 
many sources, including starting materials, intermediates, synthetic processes, or 
degradation reactions [6] . The regulatory guidelines stipulate that any impurity 
> 0.05% of total daily dose (for drugs with a dose < 2 g/day) or > 0.15% of total daily 
dose (for drugs with a dose > 2 g/day) should be evaluated for its safety [6] . Organoleptic 
properties such as color, taste, and odor are assessed qualitatively to set bulk 
drug specifi cations. If the drug has an unacceptable taste or odor, the chemistry 
group is advised to make a suitable salt form of the drug. 
PHYSICAL/BULK CHARACTERISTICS 935

936 ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS 
6.2.2.1 Crystallinity and Polymorphism 
The majority of the drugs exist in crystalline form and are characterized by their 
crystal habit and crystal lattice. The crystal habit describes the external morphology 
of the crystal, including shape and size, while the crystal lattice describes the internal 
arrangement of molecules in the crystal (Figures 1 a and b ). Drug molecules arrange 
in more than one way in a crystal, and this difference in the internal arrangement 
of crystals is known as polymorphism. The polymorphs have the same elemental 
composition but differ in their physical, chemical, thermodynamic, stability, and 
spectroscopic properties. A crystal lattice represents the space in which molecules 
arrange in different ways. Organic molecules arrange in one or more of the seven 
possible crystal systems: triclinic, monoclinic, orthorhombic, tetragonal, rhombohedral, 
hexagonal, and cubic [7] . Each crystal system is characterized by its three - 
dimensional geometry and angles between the different crystal faces. The crystal 
lattice geometry is obtained using single - crystal X - ray diffractometry (XRD) and 
the details can be found elsewhere [7] . The difference in the crystal lattice of a drug 
arises as a result of the difference in packing of the molecules if the molecules are 
conformationally rigid (e.g., chlordiazepoxide) or due to the differences in conformation 
for fl exible drug molecules (e.g., piroxicam). Although polymorphs differ in 
their internal crystal lattice, it may not be necessarily refl ected in their external 
crystal habit (Figure 1 b ). In other words, a drug can exist in different crystal habits 
without any change in the internal crystal lattice (isomorphs). 
Crystal habit is mainly dependent on crystal growth conditions [8] . For example, 
Figure 1 a shows two different crystal habits for a given crystal lattice. A prismatic 
crystal habit will result if the growth is equal in all directions, while plates are formed 
if the growth is slow in one direction. Alternatively, needle - shaped crystals (acicular) 
are formed when the growth is slow in two directions. Thus, the crystal habits can 
FIGURE 1 Schematic of crystal habits, polymorphs and amorphous drug forms. ( a ) Two 
crystal habits are shown. The internal crystal lattice is the same while the external morphology 
is different. ( b ) In a crystal the molecules are arranged in a regular fashion. However, 
the arrangement may vary depending on how the molecules orient themselves in the internal 
crystal lattice. The internal crystal lattice is different in all the three polymorphic forms. The 
polymorphs may or may not differ in their external morphology. ( c ) Random arrangement 
of molecules in amorphous form. 
(a) 
(c) 
(b)

be altered without any change in the internal crystal lattice by varying the crystallization 
conditions. The polarity of the crystallizing solvent mainly infl uences the 
crystal habit by preferentially adsorbing to one surface of the crystal face. Similarly, 
surfactants or additives are added to the crystallization medium to prevent or 
promote the growth of a specifi c crystal habit [8] . Crystal habits mainly differ in 
physicomechanical properties such as packing, fl ow property, compressibility, and 
tablettability. Acetaminophen crystallizes as polyhedral crystals when crystallized 
from water and as plates when crystallized from ethanol – water (Figure 2 a ). Both 
these crystal habits are isomorphic [9] , that is, have the same internal crystal arrangement, 
since their melting points and heats of fusion were similar (melting point 
178 ° C and . H f = 177 kcal/mol). The polyhedral crystals have better fl ow and 
FIGURE 2 Difference in crystal habit of acetaminophen and resultant difference in compressibility 
( a ) Acetaminophen crystallizes as either platy crystals or polyhedral crystals 
depending on the solvent of crystallization. Both crystal habits have the same internal crystal 
lattice since they showed the same melting point. ( b ) Difference in compression behavior of 
two crystal habits. The x axis represents the compression pressure while the y axis represents 
the densifi cation of the drug sample on compression. This plot is known as Heckel plot. The 
polyhedral crystal habit shows a higher densifi cation implying better compressibility than 
plate like crystals. [From Garekani, H. A., Ford, J. L., Rubinstein, M. H., and Rajabi - Sahboomi, 
A. R., International Journal of Pharmaceutics , 187, 77 – 89, 1999. Reproduced with permission 
from Elsevier. ) 
(a)
(b) 
0.0 
0.5 
1.0 
1.5 
2.0 
2.5 
3.0 
3.5 
4.0 
ln [1/(1–D)] 
Polyhedral crystals 
Thin platelike erystals 
0 10 20 30 40 50 60 70 80 
Compression pressure (MPa) 
PHYSICAL/BULK CHARACTERISTICS 937

938 ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS 
compression properties than platy crystals, which were brittle and fragmented 
during tableting (Figure 2 b ). Crystal habits are characterized using optical and 
electron microscopy, but their internal crystal lattice should be confi rmed using 
DSC, XRD, and spectroscopic techniques. 
Polymorphs are generated by crystallizing the drug from various solvents. The 
solvents are usually those that are used in the synthesis and purifi cation of the bulk 
drug but may also include solvents used in drug formulations [10] . By convention, 
polymorphs are named based on their order of discovery, such as forms I, II or A, 
B or . , . . In general form I is considered the most stable and least soluble form, 
while form II is considered the more soluble and least stable form. The least stable 
and more soluble polymorphic form is usually called the metastable form. They are 
not “ unstable ” but “ metastable, ” because the least stable form can remain stable 
provided the conditions are controlled to prevent its conversion to the more stable 
polymorphic form. Polymorphs are characterized by their solubility and stability 
differences with respect to temperature (Figure 3 ). Thermodynamically, polymorphs 
are classifi ed as enantiotropic or monotropic depending on their thermal reversibility 
from one form to another [11] . Enantiotropic polymorphs are reversible 
polymorphs, where one form (form I) is more stable at higher temperature, while 
the other form (form II) is stable at lower temperature. They are characterized by 
a transition point ( Ts ) below the melting points of both forms (Figure 3 a ). The 
transition point represents the temperature at which one form converts to another. 
In the temperature – solubility curve, this is represented by the intersection of the 
solubility curves of both forms; that is, at the transition temperature, both polymorphs 
have the same solubility. As shown in Figure 3 a , form II can convert to form 
I at a temperature above Ts , while form I can convert to form II at a temperature 
below Ts . On the other hand, monotropic polymorphs are not reversible but can 
only convert from the metastable form to the stable form. Here, Ts is higher than 
the melting point of both forms (Figure 3 c ). Both forms are stable in the entire 
temperature range below Ts . 
The different polymorphs are generated based on their solubility differences in 
a given solvent. According to Ostwald ’ s rule [12] , the least stable or highly energetic 
form (form II, or metastable) will precipitate out fi rst from a supersaturated solution 
followed by the stable or less energetic form (form I). Supersaturation is 
achieved by antisolvent addition or by altering the temperature. So, if the initial 
precipitate is separated rapidly, it would have predominantly the metastable form. 
Alternatively, the stable form can be melted and rapidly cooled to crystallize the 
metastable form. A stable or metastable polymorphic form is also used as a “ seed ” 
to preferentially grow and isolate the desired form during drug crystallization [13] . 
Several rules have been proposed to differentiate enantiotropic and monotropic 
polymorphs [11, 13] . A simple way to differentiate enantiotropic and monotropic 
polymorphs is the use of the heat – cool cycle in DSC [11] . As shown in Figure 3 b , 
the enantiotropic polymorph is characterized by the appearance of solid – solid 
endothermic transition of form II to I followed by melting of form I. On cooling 
the melt of form I followed by reheating, the same thermogram is regenerated, 
proving the reversibility of the polymorphs. In monotropic polymorphs (Figure 3 d ), 
the thermogram is characterized by melting of metastable (form II) and recrystallization 
to form I followed by melting of form I. On cooling and reheating the 
sample, the transition and recrystallization peaks are not seen, indicating the irre

versible nature of these polymorphs. The heating rate in DSC is critical for characterizing 
the polymorphs, as a faster heating rate may not be able to identify the 
transition temperature, while a lower heating rate may lead to lower resolution of 
peaks. Therefore, it is a usual practice to generate DSC thermograms under different 
heating rates during polymorph characterization [11] . Also it is important to 
note that the sample preparation, particle size, and crucible type can affect the 
quality of the thermogram [5] . XRD is also another indispensable tool in identifying 
polymorphs. This is based on the differential scattering of X rays when passed 
FIGURE 3 Difference between enantiotropic and monotropic polymorphs. ( a ) Solubility 
of enantiotropic polymorphs as function of temperature. The dotted line indicates the melting 
curve. Form I is less soluble below the transition temperature ( T s), while form II is more 
soluble above T s. Form I has a higher melting point ( T m,I) than form II ( T m,II). Below T s, 
form I is converted to form II and above T s form II coverts to form I. ( b ) Thermogram generated 
from heat – cool – heat cycle in DSC. In the fi rst heating cycle two endotherms are seen 
corresponding to conversion of form II to form I and melting of form I, respectively. On 
cooling both events show up as exotherms and on second heating cycle both endotherms 
reappear, indicating thermal reversibility of enantiotropic polymorphic pairs. ( c ). Solubility 
of monotropic pairs as a function of temperature. The T s is above melting point of both forms. 
Forms I and II are stable in entire temperature range and their corresponding melting points 
are shown. ( d ) On heating in DSC, form II melts ( T m,II) followed by recrystallization ( T crys) 
and subsequent melting of form I ( T m,I). In cooling cycle only melting of form I is seen as 
an exotherm and on reheating only one endotherm corresponding to form I is seen. This is 
typical of monotropic polymorphs which converts from form II to stable form I and not vice 
versa. 
Second heat 
cycle 
First heat cycle 
Endotherm 
Temperature (°C) 
Temperature (°C) 
Solubility Heat flow 
Temperature (°C) 
Temperature (°C) 
Solubility Heat flow 
Ts Ts 
Ts 
Tm,I 
Tm,II 
Tm,I Tm,I Tm,II 
Tm,II 
Tm,I 
I 
I II 
II 
Cool cycle 
Second heat 
cycle 
First heat cycle 
Cool cycle 
Endotherm 
Tcrys 
(a) (c) 
(b) (d) 
PHYSICAL/BULK CHARACTERISTICS 939

940 ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS 
through a powder sample. Typically, on passing through a powder sample, X rays 
will tend to get diffracted at various angles, and at some angle of detection, the X 
rays diffracted from the different planes of the crystal converge to form an ampli- 
fi ed signal, which is detected by a photomultiplier tube. The angles at which the 
XRD peaks are obtained are characteristic for a polymorph (Figures 4 a & b ). The 
sample should be uniformly spread to get a good X - ray diffractogram, as an 
improper sample preparation may lead to variation in intensities due to the preferred 
orientation of a crystal in the XRD sample holder [14] . Other techniques, 
such as infrared (IR) spectroscopy and solid - state nuclear magnetic resonauce 
(NMR), are also used to characterize the polymorphs and are listed in Table 2 . 
FIGURE 4 Schematic X - ray diffractograms of two polymorphic forms of a hypothetical 
drug. The x axis represents the detection angle and the y axis represents the intensity of the 
peak. As can be seen, there is a difference in the diffractograms due to the difference in the 
internal crystal lattice of polymorphs. The different internal arrangement in a crystal defl ects 
the X ray at different angles. 
10 20 30 30 40 50 
50 
100 
2 . 
2 . 
10 20 30 30 40 50 
Intensity 
50 
100 
Intensity 
(b) 
(a)

Polymorphism has signifi cant implications in the solubility, bioavailability stability, 
processing, packaging, and storage of solid drug substances [15 – 17] . The metstable 
polymorphic form may be used to improve the solubility of drug substances. 
Many drugs are known to exhibit polymorphism, particularly, steroids, barbiturates, 
anti - infl ammatory drugs, and sulfonamides, which have a high probability of exhibiting 
polymorphism [15] . The existing knowledge on drug polymorphism is a good 
starting point for a preformulation scientist to anticipate polymorphs based on the 
drug chemistry. In some cases, polymorphism may provide an opportunity to improve 
the solubility of a drug. For example, form II of chloramphenicol palmitate has a 
higher dissolution rate resulting in signifi cantly higher plasma concentration than 
form I when administered orally [15] . However, in many cases [16] the difference 
in solubility may not be signifi cant enough to cause differences in oral bioavailability 
(Table 3 ). Although the polymorphs differ in their dissolution rates, it should be 
realized that once the drug goes into solution, they do not differ in their properties. 
If a drug ’ s absorption is limited by its poor membrane permeability, then the difference 
in solubility of polymorphs may not impact its bioavailability. Similarly, if the 
drug dissolution is rapid in comparison to the gastrointestinal (GI) transit time, then 
the difference in polymorph solubility will not infl uence its bioavailability [16] . 
TABLE 2 Techniques to Characterize Different Crystalline Forms 
Technique Applications 
Thermal analysis 
Differential scanning 
calorimetry 
Melting point, enthalpy of fusion, and crystallization; solid - state 
transformations 
Thermogravimetric 
analysis 
Stoichiometry of solvates and hydrates; identifying vaporization 
and volatilization 
Hot - stage microscopy Visualization of solid - state transformations and desolvation 
events 
X - ray diffractometry Identifying polymorphs; quantifi cation of degree of crystallinity; 
crystal lattice geometry and solid - state transformations 
Spectroscopy 
Infrared Characterization of polymorphs based on functional groups; 
characterization of hydrates and solvates 
Near infrared In situ analysis of solid - state conversions; identifi cation and 
quantifi cation of polymorphs in dosage forms 
Nuclear magnetic 
resonance 
Useful to understand difference in molecular arrangement of 
polymorphs, hydrates, and solvates 
TABLE 3 Difference in Solubilities of Polymorphs 
Drug Melting Point ( ° C) Solubility Ratio a 
Indomethacin 157, 163 1.1 
Sulfathiazole 177, 202 1.7 
Piroxicam 136, 154 1.3 
a Indicates ratio of solubility of low - melting polymorphic form to solubility 
of high - melting polymorphic form of drug. 
PHYSICAL/BULK CHARACTERISTICS 941

942 ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS 
More than the presence of metastable polymorph, it is the conversion of the 
metastable to the stable form during processing, storage, or use that is of great 
concern to the pharmaceutical scientist [17] . The unpredictability in “ conditions ” 
that result in the generation and conversion of one polymorphic form to another 
mainly aggravates such a situation [18] . This is exemplifi ed by ritonavir, which is a 
classical case of appearance of a “ new polymorphic form ” after the drug was marketed 
[19] . Ritonavir, an anti - retroviral, drug was introduced in the market as a 
polymorphic form I in soft gelatin capsule in 1996. Two years later, a new polymorphic 
form II appeared in the formulation due to some unknown reasons causing the 
drug to be less soluble. The drug manufacturer withdrew the product due to failure 
of the batches in dissolution tests. After extensive investigation and reformulation, 
the drug was reintroduced in the market in 1999. 
Nonetheless, in spite of their unpredictability, a good preformulation team will 
be able to anticipate the different polymorphs during drug development. It should 
not be an impossible task given the recent advancements in high - throughput generation 
and characterization of polymorphs [20] . From an innovator ’ s perspective, the 
identifi cation and thorough characterization of multiple polymorphs during drug 
discovery can extend the patent life and delay the market entry of generic manufacturers. 
On the other hand, it is also an opportunity for the generic manufacturers 
to generate new polymorphs with better solubility and stability for gaining market 
entry. The patent dispute on ranitidine polymorphs is a good example in this regard 
[21] . Two polymorphic forms of ranitidine were patented by the innovator company 
and the generic manufacturers had to fi nd an appropriate method to manufacture 
the desired polymorph without the accompanying impurity of the other polymorph. 
This provided an edge for the innovator to extend the drug ’ s market exclusivity for 
a little longer than they would otherwise have had. If a pure polymorph cannot be 
generated, the extent of polymorphic impurity should be quantifi ed and ensured 
from batch to batch. The preformulation scientist closely works with the synthetic 
chemist in setting specifi cations for polymorphs. 
6.2.2.2 Hydrates/Solvates 
In addition to drug molecules, solvent molecules also get incorporated in the crystal 
lattice, resulting in altered physicochemical properties. When the solvent is water, 
they are known as hydrates, while if it is any other solvent, they are known as 
solvates. They are also known as pseudopolymorphs or solvatomorphs. Hydrates 
are important in this regard as one - third of all marketed drugs are hydrates [13] . 
Depending on how the water is arranged inside the crystals, they are classifi ed as 
isolated hydrates, channel hydrates, and ion - associated hydrates [13] . In isolated 
hydrates, the water molecules are separated from each other by the intervening drug 
molecules in the crystal lattice (e.g., cephadrine dihydrate). Channel hydrates result 
when water molecules are linked to one another forming a channel (e.g., theophylline 
monohydrate). The water molecules may be present either stoichometrically or 
nonstoichometrically within the crystal lattice. Ion - associated hydrates are typically 
seen when the water is metal ion coordinated (e.g., nedocromil zinc). Nonstoichometric 
channel hydrates are problematic due to the presence of diffusible water, 
which can easily move in and out of the crystal lattices [13, 22] . 
Hydrates or solvates are formed by crystallizing the drug in the presence of water 
or solvents. The hydrate formation is dictated by water activity in a given solvent 

[22] . Hydrates are characterized using gravimetric methods such as thermogravimetric 
analysis (TGA) or by Karl Fischer titrometry [23] . In TGA, the loss of 
water/solvate on heating a sample is recorded as a thermogram (Figure 5 ). The mass 
change due to dehydration is seen as a step loss in the TGA thermogram. Based on 
the weight of the initial sample and its elemental composition, the number of water 
molecules can be calculated. The TGA curve in combination with a DSC thermogram 
helps to differentiate hydrates from other thermal transitions. In Figure 5 , 
the endotherm in the DSC thermogram corresponds to water loss as indicated 
by the TGA curve. The TGA can be coupled to an IR or mass spectro meter 
to characterize solvates. Thermal microscopy is a useful qualitative tool to 
visualize the release of water from the drug crystals as a function of temperature 
[23] . 
Hydrate formation and dehydration signifi cantly infl uences the processing and 
storage of drug products [17] . Hydrates may take up further water or dehydrate to 
lose water. Dehydration of hydrates leads to several possibilities [24] , as shown in 
Figure 6 . Hydrates on dehydration can form isomorphic desolvates retaining the 
same crystal lattice as the hydrate but without the water. Alternatively, hydrates can 
lose water and become anhydrous crystals. They can also lose water, forming amorphates 
with the loss of crystal lattice. Higher hydrates can lose water to form lower 
hydrates, for example, pentahydrate converting to di - or monohydrate. The hydrates 
also can exhibit polymorphism or on dehydration can convert to a different polymorphic 
form [13, 17] . Such solid - state transformations are possible during processing, 
such as granulation, tableting, and storage [17] . In general, hydrates are less 
soluble than anhydrous forms while solvates are more soluble than ansolvates in 
water. Ampicillin trihydrate is a classical example which shows lower solubility and 
lower plasma concentration than anhydrous ampicillin [15] . Preformulation studies 
FIGURE 5 Characterization of hydrate. ( a ) TGA thermogram of monohydrate. The thermogram 
shows weight loss as a function of temperature. The step in the thermogram shows 
weight loss due to dehydration of a hydrate. ( b ) DSC thermogram showing endotherm at 
corresponding temperature ( T dehyd). The second endotherm indicates the melting point of 
the hydrate ( T m). 
Percent weight loss Heat flow 
Tm Tdehyd 
Temperature (°C) 
(a) 
(b) 
PHYSICAL/BULK CHARACTERISTICS 943

944 ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS 
provide valuable inputs to the formulator in selecting a suitable form of the drug. 
For example, ampicillin is hygroscopic and hence can be used in suspension dosage 
forms, while ampicillin trihydrate, which is non - hygroscopic, is used in solid dosage 
forms. 
6.2.2.3 Amorphate 
Unlike a crystalline drug, an amorphous form of the drug does not have a regular 
crystal lattice arrangement and the molecules are arranged in random order (Figure 
1 c ). Glass is a typical amorphous substance and so amorphous drugs are also known 
as glasses [25] . Amorphous form is prepared through milling, rapid cooling of a melt, 
rapid precipitation using an antisolvent, rapid dehydration of a hydrate, spray drying, 
or freeze drying [26] . Some of the above methods may also unintentionally produce 
an amorphous form during processing of the crystalline form of the drug [17] . For 
instance, milling during dosage form manufacture may produce an amorphous form 
unintentionally, as in the case of indomethacin. The amorphous form does not show 
a melting point but is characterized by a glass transition temperature ( T g ). This 
temperature indicates the conversion of the amorphous form from a rigid glassy 
state to a more mobile rubbery state. Above T g the amorphous form will tend to 
recrystallize and convert to the crystalline form, which then undergoes melting, as 
shown in Figure 7 a . The T g for an amorphous drug can vary depending on the 
storage conditions and thermal history of the sample and is sensitive to moisture, 
pressure, and temperature [26] . The T g is seen only as a slight shift in the baseline 
due to a change in the specifi c heat capacity of the sample and is infl uenced by the 
heating rate in DSC [25] . In XRD, the amorphous form shows a shallow peak 
or halo, as opposed to sharp and intense peaks for a crystalline drug compound 
(Figure 7 b ). 
The main advantage of amorphous form of the drug substance state is its signifi - 
cantly higher solubility than the crystalline form of the drug, primarily due to the 
excess surface energy [16, 27] . Therefore, conversion of a crystalline drug into an 
amorphous form is one of the strategies to increase drug solubility. Table 4 compares 
FIGURE 6 Various possibilities that arise from dehydration of hydrate. A hydrate can 
dehydrate reversibly into various solid - state forms. It can dehydrate to form an anhydrous 
form of the drug or to a lower hydrate. Hydrate can also dehydrate to form an isomorphic 
desolvate where the crystal lattice is retained except for the absence of water. The crystal 
structure may also collapse on dehydration to form an amorphous form. Hydrates on dehydration 
can also result in different polymorphs. 
Amorphous Lower hydrate 
Polymorphs 
Anhydrous Isomorphic 
desolvate 
Hydrate

FIGURE 7 Characterization of amorphous form. ( a ) DSC thermogram of amorphous 
substance. Thermogram is characterized by a glass transition temperature ( T g) above which 
the amorphous form is mobile and recrystallizes ( T crys) into a crystalline form which fi nally 
melts ( T m). ( b ) Amorphous form that does not show any peaks in XRD as it does not have 
regular arrangement of molecules. Shallow peaks are indicative of an amorphous drug 
substance. 
(a) 
(b) 
10 30 20 30 40 50 
100 
50 
Temperature (°C) 
Tm Tg Tcrys 
Heat flow 
Endotherm 
2 . 
Intensity 
the solubility of amorphous and crystalline forms of a few drugs. However, the 
biggest challenge lies in the stabilization of the amorphous form to prevent it from 
converting to the less soluble crystalline form during storage and use. It should be 
noted that they can take up moisture to convert to a crystalline form or to a hydrate, 
resulting in decreased drug solubility. The moisture uptake can also lead to chemical 
degradation [26] . The stabilization of drug amorphates is usually accomplished by 
increasing the T g using polymers and thus restricting their molecular mobility and 
PHYSICAL/BULK CHARACTERISTICS 945

946 ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS 
chemical reactivity [26] . Particle size reduction, which is a very common processing 
step in dosage form manufacturing, can result in varying degrees of amorphous and 
crystalline forms in the drug. In such cases it is essential to quantify the degree of 
crystallinity using various analytical techniques such as DSC and XRD [28] . 
6.2.2.4 Hygroscopicity 
Some solid drug substances have a tendency to absorb moisture from the atmosphere 
leading to physical and/or chemical instabilities. Hygroscopicity is the rate 
and extent of moisture adsorbed/absorbed by a solid substance. Solid drug substances 
may vary in their behavior to moisture and are classifi ed as deliquescent, 
effl orescent, and effervescent. Deliquescent materials such as hydrochloride salts 
absorb moisture and become a liquid. Effervescent substances (e.g., a mixture of 
citric/tartaric acid and sodium bicarbonate) absorb moisture and release carbon 
dioxide. On the other hand, effl orescent substances such as hydrates may lose moisture 
depending on the relative humidity (RH). Therefore, it is important to study 
the moisture absorption behavior of drugs to choose the processing and storage 
conditions for the drug. 
Hygroscopicity is measured by exposing the drug sample to various RH in a dessicator. 
The RH is maintained at a constant level by using salt solutions of varying 
concentrations (e.g., KNO 3 , KCl) and the humidity is expressed with respect to the 
humidity of a saturated salt solution. Moisture sorption and desorption curves are 
generated to study the moisture uptake. The moisture absorption profi le is generated 
by noting the increase in mass on exposure to varying RH and the desorption 
profi le is generated by recording the change in weight with decreasing RH [23, 29] . 
This can be measured using a dynamic vapor sorption instrument. A typical sorption/
desorption profi le is shown in Figure 8 for an anhydrous and hydrate form of 
a hypothetical drug. As can been, for a hydrate the sorption and desorption profi le 
is superimposable and is hence non - hygroscopic. On the other hand, the anhydrous 
form of the drug is hygroscopic and shows hysteresis on the sorption and desorption 
profi le, indicating signifi cant moisture uptake. Such profi les give useful clues to the 
preformulation scientist. Signifi cant hysteresis is indicative of hydrate formation and 
can be used as a guide to evaluate the potential of hydrate formation. Further the 
profi le also helps to differentiate hygroscopic and non - hygroscopic salts [29] . The 
profi le also gives information on processing and storage conditions that can overcome 
solid - state transformations. For example, in Figure 8 , it is seen that the anhydrous 
form does not take up moisture if RH is below 80% and it retains the moisture 
TABLE 4 Comparative Solubilities of Amorphous and 
Crystalline Forms of Drugs 
Drug Solubility Ratio a 
Carbamazepine 1.5 – 1.7 
Griseofulvin 38 – 441 
Glibenclamide 14 
a Indicates ratio of solubility of amorphous form to solubility of crystalline 
form of drug. 

FIGURE 8 Sorption and desorption profi le of hydrate and anhydrous form of hypothetical 
drug. The solid lines represent the profi le for an anhydrous form of the drug, while the broken 
lines represent the profi le for a hydrate form of the drug. Anhydrous form of the drug does 
not take up moisture until it reaches 80% RH, and on reducing the RH, it does not lose 
moisture until it reaches RH of 20%. The hysteresis is indicative of hygroscopicity and 
signifi cant moisture uptake. The hydrate form of the drug does not show hysteresis but 
both the sorption and desorption curve superimpose on each other indicating that it is 
non - hygroscopic. 
20 40 60 80 
Change in mass (%) 
Relative humidity (%) 
until the RH is reduced below 20%. The study can also be used to extract kinetic 
and temperature information on moisture uptake by drug substances. The studies 
conducted during preformulation testing should be representative of the anticipated 
processing and storage conditions of the drug. 
6.2.2.5 Particle Characteristics 
Drug particle characteristics such as size, shape, and surface area impact the drug ’ s 
processability and product performance. Particle size is the most infl uential among 
these as the other two properties can be related to it. When suffi cient drug is available, 
the preformulation scientist characterizes the particle size and size distribution 
to set specifi cations for formulation and future drug lots. Table 5 lists the various 
methods used to measure particle size. The methods differ in their principle of 
operation and also in the range of particle sizes they can measure [30] . Usually, the 
gross particle morphology is characterized using a simple optical microscope and if 
required is further characterized using a scanning electron microscope. Light - 
scattering methods are commonly used to measure particle size due to their low 
sample requirements and ease of measurement. The instrument readout is in the 
form of a graph where the particle size is plotted against the percent frequency of 
particles in different particular size ranges. The results are used to set particle size 
specifi cations and understand polydispersity or multimodal particle size distribution 
in powders. Especially if the drug is potent, a narrow size distribution is desired to 
ensure drug homogeneity during formulation. The surface area of a powder bed is 
determined using the Brunauer, Emmett, and Teller (BET) method. In this method, 
PHYSICAL/BULK CHARACTERISTICS 947

948 ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS 
N2 gas is passed through a powder bed and the surface area is calculated based on 
the volume of gas coming out in the absence and presence of the powder [31] . 
Particle size reduction is an initial step in the development of any dosage form. 
The data generated during preformulation testing guides the formulator in deciding 
about size reduction. Particles with size > 100 . m generally require size reduction 
and particles in the size range of 10 – 40 . m are generally acceptable for solid dosage 
forms [32] . As mentioned in the earlier section, particle size reduction may lead to 
partial or complete amorphization of a powder, and this factor should be taken into 
consideration. Also worth mentioning are the other solid - state transformations that 
may take place during milling, such as the conversion of one polymorphic form to 
another or the desolvation of a hydrate [17] . It is important to maintain the particle 
size distribution within a narrow range to avoid powder stratifi cation and avoid 
fl owability issues during capsule fi lling or tablet compression. Particle size reduction 
increases drug solubility due to the enormous increase in surface area with 
decreasing particle size. Griseofulvin is often widely quoted in the literature in this 
regard, where the bioavailability of this water - insoluble drug is increased 10 times 
on reducing the particle size [32] . On the other hand, particle size reduction may 
be counterproductive for some drugs such as nitrofurantoin. Particle size reduction 
of nitrofurantoin causes rapid drug absorption with an associated increase in its 
adverse effects. In contrast, the slowly dissolving macrocrystals of nitrofurantoin do 
not cause adverse effects [32] . Excessive particle size reduction ( < 30 . m) may also 
lead to static charge buildup on the surface, resulting in agglomeration and reduced 
powder fl ow or reduced drug solubility. If the drug is sensitive to moisture or 
oxygen, then the increased surface area associated with particle size reduction 
may accelerate degradative reactions [32] . Therefore, it is important to study the 
infl uence of particle size on drug solubility and stability during preformualtion 
testing. 
TABLE 5 Methods for Particle Size Analysis 
Method Principle 
Sieve Sieve analysis utilizes a series or stack, or nest of electro brass or 
stainless steel sieves that have smaller mesh at the bottom followed by 
meshes that become progressively coarser toward the top of the series. 
Useful for measuring particles in size range 10 – 50,000 . m 
Microscopy Analysis is carried out on two - dimensional images (projected diameter) 
of particles which are assumed to be randomly oriented in three 
dimensions. Can measure particles in the size range 1 – 1000 . m. 
Electron microscopy is useful for analysis of particles in 
submicrometer range (0.01 – 100 . m). It also gives information on 
surface morphology and shape of the powder. 
Sedimentation Size analysis is based on sedimentation of particles as a function of their 
size due to gravitational pull or by using a centrifugal force. Can 
measure particles in the size range 0.01 – 100 . m. 
Light 
scattering 
This is based on the principle of light scattering of particles as a function 
of their hydrodynamic radius. Commonly used, as it requires a small 
sample size and is a rapid method for particle size measurement. It can 
be used to measure particles varying in size from 0.001 to 100 . m. 

6.2.2.6 Powder Flow and Compressibility 
The derived properties of a drug substance play a critical role in deciding about the 
feasibility of a solid dosage form. These include the bulk density, fl ow properties, 
and compressibility of the drug powder. Powder fl ow is infl uenced by many solid - 
state properties, including crystal habit, bulk density, particle size, and shape. Bulk 
density is an important determinant of powder fl ow. It is the ratio of a known weight 
of the powder and its bulk volume. This is determined by pouring a weighed amount 
of the powder into a graduated cylinder and measuring its volume. Bulk density is 
particularly important for high - dose drugs, where the drug would occupy a major 
portion of the tablet or capsule dosage form. The true density of a powder is determined 
using a helium densitometer [31] . The volume of helium gas that passes 
through an empty tube is compared with the volume of helium passing through the 
tube when fi lled with a defi ned weight of the powder. The difference in the volume 
gives the true volume of the powder, which is then used to calculate the true density 
of the powder. This information can be used to calculate the porosity: 
Porosity 
bulk volume true volume 
bulk volume 
= . . 100 
(1) 
The porosity of a powder depends on particle size and shape; pharmaceutical 
powders vary in porosity from 30 to 50% [31] . Powder with varying particle size will 
give a porosity of less than 30%, where the small particles may occupy the pores in 
between the larger particles. On the other hand, powder aggregates lead to increased 
porosity and poor fl ow properties. 
The powder fl ow is determined using the angle of repose or Carr ’ s index (Figures 
9 a and b ). The angle of repose is the angle that the powder makes with the horizontal 
surface when allowed to fl ow through a funnel (Figure 9 a ). This is based on the 
principle that powder fl ow is infl uenced by the relative infl uence of interparticle 
friction and the gravitational pull on the powder. Pharmaceutical powders have an 
angle of repose of 25 ° – 40 ° [33] , and in general, a good fl owing powder will have a 
lower angle of repose (Figure 9 c ). The angle of repose using the funnel provides a 
good estimate of the infl uence of particle size, shape, and electrostatic interaction 
between the particles when the powder fl ows through the hopper in a tableting 
machine [34] . When there is a limited drug sample, Carr ’ s index is used to estimate 
powder fl ow and compressibility. In this method, a small quantity of the powder is 
used to determine its bulk density and this is followed by determining the tapped 
density of the powder. After fi lling, the graduated cylinder is tapped on a hard 
surface (3 – 30 taps) until the powder consolidates and gives a constant volume 
(Figure 9 b ). Carr ’ s index is calculated using the equation 
Carr s index 
tapped poured density 
tapped density 
’ = . . 100 
(2) 
This index is a good measure of powder consolidation and compressibility for predicting 
the feasibility of developing a tablet dosage form. A lower Carr ’ s index is 
indicative of a good fl owing powder. There is a good correlation between angle of 
repose and Carr ’ s index [Figure 9 c ], and depending on the quantity of the drug 
PHYSICAL/BULK CHARACTERISTICS 949

950 ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS 
available for preformulation testing, either method may be used to estimate the 
powder fl ow property. Poor fl owing powders may require glidants to improve their 
fl ow property. Compressibility is studied by compressing the drug in a hydraulic or 
IR press, and such experiments give early warnings to the formulator about capping 
and lamination issues in tablets. Altogether, these preformulation tests give valuable 
information to the formulator in deciding excipients and processes. 
6.2.3 SOLUBILITY CHARACTERISTICS 
Drug solubility is one of the physicochemical parameters that receives lots of attention 
during preformulation testing. In the initial stages, solubility studies are usually 
kinetically determined, where the drug is placed in contact with the solvents and 
then the solubility is assessed using turbidometric methods almost instantaneously. 
FIGURE 9 Measurement techniques for powder fl ow. ( a ) Angle of repose is determined 
by pouring a powder through a funnel and noting the angle that the powder heap makes with 
the horizontal surface. ( b ) Carr ’ s index is measured by pouring a known weight of the powder 
into a graduated cylinder and tapping it on a hard surface until the powder is consolidated. 
( c ) Carr ’ s index is measured as percent, while angle of repose is measured in degrees, 
and both methods show good correlation. Powder fl ow is classifi ed based on either of the 
measurement methods. 
Poor 
Good 
Bulk volume 
Volume after tapping 
(a) (b) 
(c) 
Carr’s index (%) 
5–15 
12–16 
18–21 
23–35 
33–38 
>40 
<20 
20–30 
30–34 
— 
>40 
— 
Angle of repose (deg) Powder flow 
Excellent 
Fair to 
passable 
Very poor 
Extremely 
poor

SOLUBILITY CHARACTERISTICS 951 
A high - throughput solubility screen consists of dissolving the drug in a minimal 
volume (few microliters) of dimethyl sulfoxide (DMSO) and then adding different 
volumes of water until turbidity is observed. The appearance of turbidity is considered 
as a rough estimate of the drug ’ s water solubility and turbidity is measured in 
a 96 - well plate format in a turbidometer [35] . Solubility is qualitatively described in 
terms of how much solvent is required to solubilize 1 g of the drug and is shown in 
Figure 10 . Kinetic solubility measurements are only rough estimates, as they do not 
take into account the solid - state transitions and should be followed up with equilibrium 
solubility studies later when more drug is available. 
Equilibrium solubility is determined by placing an excess solid drug in a few 
milliliters of the solvent and shaking at 37 ° C for 60 – 72 h until equilibrium is reached. 
One to three samples are withdrawn, fi ltered, and assayed for drug content. Equilibrium 
solubility helps to identify polymorphic or amorphous forms of the drug, 
which shows an apparently higher solubility in kinetic studies. Intrinsic solubility 
measurements are measured for ionic compounds to determine the inherent solubility 
of the un - ionized form of the drug. This would mean that the solubility of a 
weakly acidic drug is tested in an acid medium and a weakly basic drug is tested in 
an alkaline medium, where the drug would remain completely un - ionized. The 
studies are followed by determining drug solubility at different pH and are discussed 
later in this section. 
FIGURE 10 Terminologies for drug solubility. The drug solubility is qualitatively described 
depending on how much solvent is required to solubilize 1 g of the drug. 
Practically insoluble 
>1000 mL 
Very slightly soluble 
100–1000 mL 
Slightly soluble 
100–300 mL 
Sparingly soluble 
30–100 mL 
Soluble 
10–30 mL 
Freely soluble 
1–10 mL 
Very soluble 
<1 mL 
Solvent 
required for 
1 gm of solid

952 ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS 
A drug solubility of 1 mg/mL is usually considered acceptable to avoid dissolution 
rate – limited absorption in vivo. If the drug solubility is < 1 mg/mL, salts should be 
considered, if the drug is ionic and cosolvents should be considered if the drug is 
nonionic [33] . This brings us to a discussion of the factors that infl uence drug solubility: 
temperature, pH, crystal form, and solvents. The effect of temperature on drug 
solubility depends on the heat of solution ( . H s ), which is the amount of heat given 
up or taken up when a drug goes into solution. The relationship between drug solubility 
S and temperature T is given by the van ’ t Hoff equation, where R and C are 
constants: 
ln S 
H 
RT 
C = + . s 
(3) 
Drugs which have . ve . H s (exothermic) generally decrease in solubility with 
increase in temperature (e.g., lithium salts and hydrochloride salts), while drugs 
which have + ve . H s (endothermic) usually increase in solubility with increase 
in temperature (e.g., most organic drugs). Nonelectrolytes have a . H s of 4 – 8 kcal/ 
mol and show signifi cant increase in solubility when the temperature is increased. 
On the other hand, salts have a . H s of . 2 – 2 kcal/mol and are therefore less 
sensitive to temperature [34] . Solubility studies are conducted at 25, 37, 
and 50 ° C. In addition to providing information on the drug solubility at the 
body or processing temperatures, it is also useful to understand polymorphic 
interconversions. 
With respect to selecting cosolvents, one should consider drug polarity and 
solvent polarity. Usually the solvents include glycerol, propylene glycol, and ethanol. 
Other solubilization techniques such as complexation and surfactants can also be 
used to enhance the solubility of the drug [36] . However, the solubilization techniques 
used in preclinical testing may not be same as the fi nal formulation used in 
clinical studies and marketing. 
For ionic substances, salt formation is the preferred strategy for drug solubilization. 
The salt selection is crucial during early discovery, as any change will require 
repeating some of the earlier preclinical studies. Salts provide wider fl exibility in 
modulating the drug properties without changing its activity. The salt formation is 
used to address drug solubility, stability, and processing issues [37] . Sometimes, salt 
formation may be used to deliberately reduce the solubility of drug to overcome 
unpleasant taste or stability of the drug. For example, clindamycin pamoate is used 
in place of hydrochloride salt to overcome the unpleasant taste of the latter. The 
various factors to be considered in salt selection are discussed in the next section. 
In addition to improving the solubility of the drug, solubility data guides the formulator 
to choose a suitable granulating solvent for a tablet dosage form. 
6.2.3.1 p K a and Salt Selection 
The majority of the pharmaceutical drugs are weak bases or weak acids. Among the 
marketed drugs, more than 75% are weak bases, 25% are weak acids, and 5% are 
nonionic [38] . Therefore, knowledge of p K a is useful for enhancing drug solubility 
and stability. The Henderson – Hasselbalch equation is used to describe the ionization 
of a weak acid or base:

SOLUBILITY CHARACTERISTICS 953 
Percent ionized 
100 
1+10 
for weak acid 
fo 
(p pH 
pH p 
a 
a 
= 
+ 
.
. 
K 
K
) 
( ) 
100 
1 10 
r weak base 
.
. . 
. . 
(5) 
The equations theoretically predict the ionization and solubility of a drug at a given 
pH. The p K a , which is a characteristic property of an ionizable drug, defi nes the pH 
at which the drug is half ionized and half un - ionized. A weakly acidic drug will be 
predominantly in the un - ionized form two pH units below its p K a and predominantly 
ionized at two pH units above its p K a . In a weakly basic drug, it is exactly 
the opposite of what is seen with an acidic drug. The p K a of a drug can be measured 
using potentiometry, solubility, conductometry, and spectroscopic techniques [35] . 
Potentiometry measures the change in potential when the drug is titrated with an 
acid or base and is suitable for drugs which have p K a of 3 – 10 [33] . If the drug is not 
water soluble, it is usually dissolved in a water - miscible solvent such as DMSO or 
methanol. In order to nullify the effect of cosolvents, measurements are made with 
various cosolvent concentrations and are plotted against p K a . The intercept on the 
y axis gives the p K a at zero cosolvent concentration. Some drugs have more than 
one ionizable group, such as ampholytes, and are characterized by more than one 
p K a value. They are classifi ed as ordinary or zwitterionic ampholytes [39] . In ordinary 
ampholytes, p K a,acidic > p K a,basic (e.g., chlorambucil), while in zwitterionic ampholytes, 
p K a,acidic < p K a,basic ampholytes (e.g., amino acids and proteins). The amino acids 
and proteins are characterized by their isoelectric point (pI), which is the pH at 
which the net charge is zero and is calculated using the formula 
pI p p a,acid a,base = + 1
2
( ) K K 
(6) 
Drugs may also have more than two p K a values, such as polyprotic or polybasic 
compounds (e.g., minocycline), and such drugs exhibit a complex pH solubility 
profi le. It is essential to know per se pH of the drug solution during preformulation 
studies. The pH is measured or theoretically calculated if the p K a and drug concentration 
C are known. The pH of a weak acid or the salt of a weak base and a strong 
acid can be calculated using the equation 
pH 
p a = . ( log ) K C 
2 
(7) 
Similarly, the pH of a weak base or a salt of weak acid – strong base can be calculated 
using the following equation, where p K w is the ionization constant of water: 
pH 
p p w a = + + ( log ) K K C 
2 
(8) 
Solubility of a drug is directly proportional to the extent of ionization of a drug in 
water. Therefore, p K a of the drug may also be determined by noting the change in 
solubility of a drug as a function of pH. It is customary to check the drug solubility 
(4)

954 ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS 
in the pH range usually encountered in the gastrointestinal tract (GIT; pH 1 – 8). The 
infl uence of pH on drug solubility can be estimated from the equations 
S 
S K 
S K 
= 
+ . 
+ . {u a 
u a 
pH p for weak acid 
p pH for weak base 
[ ( )] 
[ ( )] 
1 10 
1 10 
(10) 
In this equation S represents the total solubility of ionized and un - ionized forms of 
the drug, while S u represents the intrinsic solubility of un - ionized form of the drug. 
The pH solubility profi le and the drug p K a are important for salt selection. In addition, 
the salt selection is governed by various factors as outlined in Table 6 . As a 
rule of thumb, a strong acid is used to form a salt with a weakly basic drug and a 
strong base is used to form a salt with a weakly acidic drug. The probability of salt 
formation can be predicted from the relative p K a values of the drug and the counterions 
by using the equation [40] 
.p p p a a,drug a,counterion K K K = . (11) 
The probability of salt formation is high for a weak acid if . p K a is negative, that is, 
p K a,drug < p K a,counterion . Sodium phenytoin is an example, where phenytoin has a p K a 
of 8.3 and sodium has a p K a of 16. Similarly, a weak base will form a salt with acid 
counterion if . p K a is positive, that is, p K a,drug > p K a,counterion . In atropine sulfate, atropine 
has a p K a of 9.9 and sulfuric acid has a p K a of . 3. It is important to mention 
that salts do not alter the intrinsic p K a of the drug but increase drug solubility by 
keeping pH on the ionization side of the drug ’ s p K a (Figures 11 a and b ). The salt 
formation is a futile exercise if the p K a of a drug is < 3 or > 10 and other solubilization 
strategies have to be pursued [33] . 
Inorganic ions such as hydrochloride and sodium are the most frequently encountered 
species in pharmaceutical salts, primarily because of the ease of salt formation 
and their low molecular weights. They provide signifi cant increase in drug solubility 
and at the same time strong acids/bases may also be hygroscopic, posing problems 
during processing and storage. In those cases, salts are formed using weaker organic 
anions or cations such as mesylate, besylate, and choline. Table 7 lists some of the 
commonly used inorganic and organic counterions used in pharmaceutical salts. The 
pH provided by the salts signifi cantly infl uences the drug dissolution and subsequent 
drug absorption from the GIT. Though salts increase drug ionization and aqueous 
drug solubility, it is the un - ionized form of the drug that is absorbed through the 
membrane. According to the well - known pH partition hypothesis [33] , a weakly 
TABLE 6 Factors to Consider during Salt Selection 
Relative p K a of the drug and the counterion 
Common - ion effects 
Crystal habit and crystallinity 
Polymorphic conversions 
Hygroscopicity 
Chemical stability 
Manufacturability 
Toxicity 
(9)

SOLUBILITY CHARACTERISTICS 955 
TABLE 7 Various Counterions Used to Form Drug Salts 
Chemical class Salt - Forming Counterions 
Inorganic Hydrochloride, hydrobromide, sulfate, nitrate, phosphate, sodium, 
potassium, calcium, and zinc 
Organic Triethanolamine, ethanolamine, lactic acid, maleic acid, citric acid, acetic 
acid, choline, ethanesulfonic acid, oleate, and stearate 
FIGURE 11 Relative p K a of drug and salt - forming counterion. ( a ) For a weak acid, the 
p K a of the salt - forming counterion should be higher than the drug p K a to keep the pH on 
the ionization side. ( b ) For a weak base, the salt - forming counterion should have a p K a less 
than the drug ’ s p K a to keep the pH in the ionization side. 
(a) 
(b) 
pH 
pKa of counterion pKa of drug 
Percent Ionization Percent Ionization 
pH 
pKa of 
counterion 
pKa of drug

956 ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS 
acidic drug is primarily absorbed from the stomach (pH 1 – 3), where it remains in 
un - ionized form. A basic drug is expected to be absorbed from the intestines (pH 
6.5 – 8), where it would be in the un - ionized form. But in many cases, the absorption 
of drugs cannot be satisfactorily explained by this theory. This is understandable if 
one considers the fact that drug dissolution from a salt is mainly infl uenced by the 
surface pH of the dissolving drug (microenvironment) rather than the bulk pH of 
the GI fl uids. For instance, the surface pH of weakly acidic drug is 1.5 times higher 
than the bulk pH in the stomach, and therefore the dissolution of the salt of a weak 
acid will be 100 times faster than the free - acid form of the drug in stomach [40] . 
Similarly, it is the free base rather than its salt form which will dissolve faster in the 
gastric pH. So it is possible to modulate the drug solubility of a pharmaceutical salt 
irrespective of its location in the GIT. Considering the fact that most of the drugs 
are absorbed from the intestine (due to large surface area), it is desirable to select 
a salt that is not completely ionized or unionized at the intestinal pH to have optimal 
dissolution and absorption. 
An important phenomenon that is often overlooked during the salt selection 
process is the suppression of salt ionization in GI fl uids due to the common - ion 
effect. This is particularly important with inorganic salts, where salt ionization can 
be suppressed by ions such as chloride and sodium which are abundant in GI fl uids. 
A hydrochloride salt will ionize in solution, as shown in the Equation (12) , but in 
gastric fl uid, the presence of chloride ions suppresses the drug ionization [as shown 
by the thicker arrow in the reverse direction in Equation (13) ] to maintain an equilibrium 
between the ionized and un - ionized form of the drug: 
DH Cl DH Cl + . + . +  (12) 
DH Cl DH > Cl + . + . + (13) 
Hence it is important to study this effect during preformulation by testing the solubility 
of the salt in the presence and absence of sodium chloride. Although salts do 
not alter the pharmacological activity of the drug, safety is an important consideration 
in the selection of salts. From this perspective, salts are treated as a new 
molecule by the FDA. The safety of the salt is evaluated with respect to its route 
of administration and dose of the drug [37] . 
6.2.3.2 Partition Coeffi cient 
In simple terms, the partition coeffi cient represents the relative solubility of a drug 
in a hydrophobic and a hydrophilic solvent. The hydrophilic solvent is usually water 
or buffer (pH 7.4), while the hydrophobic phase is usually n -octanol. The partition 
coeffi cient ( K o/w ) is defi ned as the ratio of concentration of the drug in the organic 
phase ( C o ) to drug concentration in the aqueous phase ( C w ): 
K 
C
C o/w 
o
w 
= 
(14) 
The choice of n - octanol is based on its ability to mimic the lipophilicity of the biological 
membranes [33] and further its solubility parameter ( . = 10.24; solubility 

SOLUBILITY CHARACTERISTICS 957 
parameter is a measure of internal cohesive energy) falls within the solubility 
parameter range of most drugs (8 – 12.4). The partition coeffi cient is determined by 
dissolving the drug in one of the phases and shaking both the phases together in a 
fl ask for 30 min to achieve equilibration. Then the drug concentration is determined 
from one of the phases, usually the aqueous phase, and the drug concentration in 
the oil phase is determined by subtracting the drug concentration in the aqueous 
phase from the total drug that was added. This value when expressed in logarithmic 
form is known as log P . The phase volume of the two phases is 1 : 1 but, if the drug 
is less soluble in the aqueous phase, the ratio (water – octanol) is changed to 1 : 10 or 
1 : 20 to have a measurable drug concentration in the aqueous phase [33] . It is important 
to saturate the phases with respect to the other solvent before starting the 
experiment to rule out the infl uence of solvent partitioning on drug distribution 
between the two phases. 
Another important factor is the infl uence of drug ionization on the partition 
coeffi cient and this is particularly important when a buffer is used instead of water. 
The partition coeffi cient determined from Equation (14) is an apparent value rather 
than a true partition coeffi cient for ionic drugs. However, the true partition coeffi - 
cient can be calculated from the apparent partition coeffi cient if the drug ’ s p K a and 
the pH of the drug solution are known [39] , as shown in the equations 
log 
log log 
log log 
( ) 
P 
P
P 
K 
= 
. 
+ ( ) 
. 
+ 
. app pH p 
app 
a 
for weak acid 
1 
1 10
1 
1 10( ) p pH a 
for weak base K . ( ) 
.
. . 
. . 
(16) 
During the initial stages of drug screening, the partition coeffi cient is calculated 
based on the chemical structure ( C log P ). This is done by assigning values to different 
fragments in the chemical structure [41] . The calculated values are only estimates, 
but they are useful to rank order a homologous series of compounds based 
on their lipophilicity for further lead optimization. Given the fact that drugs have 
to cross many biological membranes before reaching the site of action, the log P 
value has a signifi cant infl uence on drug absorption, drug pharmacokinetics, and 
pharmacology. This is exemplifi ed from the numerous structure – activity and structure 
– property relationships using log P [41] . The log P is important in assessing the 
oral absorption potential of a drug. If a drug has a low log P ( < 1), it is expected to 
have poor membrane permeability, while if a drug has a large log P ( > 5), it will be 
trapped in the lipophilic membrane. A log P of 1 – 5 is usually considered optimal 
for oral drug absorption [3] . For an ionic drug, the un - ionized form of the drug will 
be more lipophilic than the ionized form of the drug. Therefore, at any given pH in 
vivo, the relative proportion of ionized versus un - ionized form of the drug dictates 
drug dissolution and absorption through the membrane. 
6.2.3.3 Drug Dissolution 
Dissolution is the rate at which a solid drug goes into solution and is a critical 
determinant in the absorption of drugs from solid dosage forms. A drug has to go 
into solution before it can be absorbed. In vitro dissolution studies are a valuable 
(15)

958 ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS 
tool for determining the infl uence of different solid - state properties on drug dissolution 
and vis - a - vis predict in vivo drug dissolution and absorption. It is used to screen 
drugs that show dissolution rate – limited absorption. The factors that infl uence drug 
dissolution rate ( dc / dt ) can be understood from the well - established Noyes – Whitney 
equation [42] 
dC 
dt 
kSC 
V 
= s 
(17) 
where k is the dissolution rate constant (cm/s), C s is the saturated solubility of the 
drug, S is surface area of solid exposed to the solvent, and V is the volume of the 
dissolution medium. The dissolution rate constant is a function of the diffusion coef- 
fi cient D of the drug through a stagnant aqueous layer adjacent to the dissolving 
surface and thickness h . Powder or particulate dissolution is carried out in a dissolution 
vessel in a specifi c volume of dissolution fl uid (900 mL) which is stirred and 
maintained at 37 ° C. Several confi gurations are available to study the dissolution of 
various dosage forms (Figures 12 a and 12 b ). Various simulated physiological media 
are used to understand the in vivo behavior of drug and dosage forms [43] . The 
usual dissolution media include water, 0.1 N HCl, and pH 7.4 buffers. It is important 
to maintain sink conditions in the dissolution medium by keeping the drug concentration 
at 10% of saturated solubility of drug (to mimic in vivo). For poorly water 
soluble drugs, surfactants are often added to the dissolution medium for this purpose. 
During preformulation testing, particulate dissolution studies reveal the infl uence 
FIGURE 12 Various drug dissolution methodologies. ( a ) In the paddle method, the tablet 
is placed in the dissolution vessel containing dissolution medium and the paddle is rotated 
at defi ned rpm, while the dissolution vessel is maintained at body temperature. ( b ) In the 
basket method, the tablet is kept inside a meshed basket and rotated. ( c ) For IDR studies, 
the tablet is kept inside a die cavity and only one face of the tablet is exposed to the dissolution 
medium. 
(a) (b) (c) 
Tablet 
Tablet 
Tablet

SOLUBILITY CHARACTERISTICS 959 
of particle size, crystal habit, and wettability of a drug substance. A formulator, on 
the other hand, routinely uses dissolution testing as a quality control tool in the 
design of dosage forms. Various models have been developed to describe the release 
kinetics of conventional and modifi ed release dosage forms [44] . 
Since powder dissolution is infl uenced by changing surface area, it is not useful 
for delineating the effects of polymorphs, hydrates, and pharmaceutical salts. Instead, 
the intrinsic dissolution rate (IDR) is used. The IDR studies are conducted at a 
constant surface area and hence the dissolution rate observed is only a function of 
the intrinsic solubility of the drug. The Noyes – Whitney equation is modifi ed for 
IDR, where the surface area is kept constant, and Equation (17) reduces to 
d
d 
k C 
C
t s = 1 
(18) 
where k 1 is the intrinsic dissolution rate constant and dC / dt is the intrinsic dissolution 
rate (mg · cm 2 /s). For IDR studies, the drug (500 mg) is compressed in a hydraulic 
press (at 500 mPa) to a 13 - mm disc. This disc is then loaded onto a holder which 
exposes only one surface of the disc to the dissolution medium (Figure 12 c ). The 
IDR is obtained by dividing the slope of the plot between the amount of drug dissolved 
and time by the area of solid exposed to the dissolution medium. The IDR 
predicts the infl uence of drug solubility on in vivo drug dissolution and absorption. 
A drug which has an IDR of 1 mg · cm 2 /s will not generally show dissolution rate – 
limited absorption in vivo. However, if the IDR falls between 0.1 and 1 mg · cm 2 /s, 
then further studies may be required to make a decision. Drugs with IDR < 
0.1 mg · cm 2 /s show dissolution rate – limited absorption in vivo, necessitating drug 
solubilization strategies [45] . With respect to pharmaceutical salts, IDR is used to 
understand the infl uence of surface pH on drug dissolution and absorption. The 
common - ion effect can be studied by including 0.1 – 0.15 M NaCl in the dissolution 
medium. Also IDR is useful to understand the difference in solubility of polymorph 
and amorphous forms. However, in some cases, the compression force used in 
making the IDR disk may by itself induce solid - state transformations [45] . DSC, IR, 
and XRD must be used to identify the drug ’ s solid state before and after compression 
as well as at the end of the dissolution studies. A well - designed dissolution study 
is used as an early warning for drug molecules that would pose absorption problems 
in vivo. 
6.2.3.4 Absorption Potential 
The ultimate goal of any drug development program is to develop an orally absorbable 
compound. Solubility and permeability are the two most critical parameters 
that dictate oral absorbability of a molecule. All other parameters are directly or 
indirectly related to these two physicochemical properties. As can be seen from 
Figure 13 , there are several barriers that a drug needs to overcome before reaching 
the systemic circulation. The oral absorption of a drug is mainly limited by drug 
dissolution and/or by the drug permeation across the GI membrane. Considering 
their importance, drug solubility and permeability are screened very early in the 
drug discovery process. Solubility studies are typically run in a high - throughput 
format using a turbidometric method as discussed earlier. Based on this primary 

960 ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS 
screen, detailed solubility and dissolution studies are carried out as the compound 
moves through different development phases (Figure 14 a ). A usual target for drug 
solubility during lead selection is 1 mg/mL for avoiding dissolution rate – limited 
absorption [46] . 
Drug permeability is commonly screened in the early discovery phase using 
CaCo - 2 cell lines, which is a human colon carcinoma cell line. Using a 96 - well format, 
the cell lines are used as a primary screen to rank order compounds based on permeability 
[4] . If designed properly, the cell culture studies can be used to understand 
the drug absorption mechanisms [47] . To avoid permeability - limited absorption in 
vivo, a drug should have a permeability coeffi cient of 2 . 10 . 4 – 4 . 10 . 4 cm/s [48] . 
Inputs from the drug metabolism team (based on liver microsomal studies) can give 
clues on the drug ’ s susceptibility to fi rst - pass metabolism. Following cell culture 
studies, a select group of compounds are studied using isolated rat intestine and 
segmental absorption studies to understand the drug absorption mechanism and the 
site of drug absorption in the GIT (Figure 15 b and 15 a ). This is further substantiated 
using in situ perfusion experiments in rats [49] . The details of the studies are 
depicted in Figure 15 b . Some of these compounds are studied in whole animals of 
which a few may make it to clinical trials in humans. 
FIGURE 13 Barriers to drug absorption. Drug from the dosage form should be soluble and 
dissolve in GI fl uids before it can be absorbed. Drug dissolution is one of the major rate - 
limiting steps in drug absorption. Drug absorption may be affected if the drug is unstable in 
GI fl uids. Furthermore, drug absorption will also depend on how long the drug resides in a 
particular region of the GIT. The drug has to diffuse through the highly viscous mucous layer 
before getting absorbed through the membrane. Membrane permeability is one of major the 
rate - limiting steps in absorption. After absorption the drug may be subject to fi rst - pass 
metabolism (FPM) in the liver before reaching the systemic circulation. Dark arrows indicate 
that solubility and permeability are the most infl uential factors. 
Permeability 
FPM 
Systemic 
circulation 
GI 
membrane 
GI transit time 
Dosage form Drug in 
solution 
Solubility and 
dissolution 
Drug stability 
Mucous layer

SOLUBILITY CHARACTERISTICS 961 
FIGURE 14 Different methods to determine solubility and permeability during various 
stages of drug discovery and development. ( a ) In the initial stages the drugs are screened 
using kinetic solubility studies, which are later followed by equilibrium solubility studies, pH 
solubility profi le, and dissolution studies during development phase. ( b ) Drug permeability 
is initially screened using CaCo - 2 cell lines followed by rat intestinal studies. This is followed 
by pharmacokinetic studies in animals and fi nally the potential drug molecules are tested in 
humans in clinical trials. 
(a) (b) 
Drug discovery and development 
Human studies 
Whole-animal studies 
Isolated rat 
intestine and In Situ 
perfusion in rats 
CaCo-2 
cells 
Kinetic 
solubility 
Equilibrium 
solubility 
pH solubility profile 
Dissolution studies 
Thus, the preformulation team in coordination with other discovery teams gets 
useful estimates on in vivo drug absorption. In addition to drug solubility and permeability, 
it is also important to consider the anticipated dose while assessing the 
absorption potential. A useful tool to optimize the drug ’ s physicochemical properties 
is the maximum absorbed dose (MAD) model [46] . The model predicts the dose 
that would be absorbed based on the drug ’ s solubility ( C s ; solubility in intestinal pH 
of 6.5), absorption rate constant ( k a ; usually obtained from rat permeability studies), 
physiological factors such as gastric transit time ( T i ; approximated as 4.5 h), and 
intestinal fl uid volume ( V int ; approximated as 250 mL): 
MAD s a i =C k V T int (19) 
Using this model, the required solubility or permeability can be estimated for a 
given dose of the drug. For example, a drug with an anticipated human dose of 
1 mg/kg (70 mg for a normal 70 - kg subject) will require a solubility of 0.05 mg/mL 
provided the drug shows good permeability. Similarly, the absorption rate required 
to achieve the same dose for a drug with good solubility (1 mg/mL) is 0.001 min . 1 . 
The MAD model is helpful in guiding development teams on optimizing drug solubility 
and/or permeability. 
A further refi nement of this model led to the evolution of the biopharmaceutics 
classifi cation system (BCS), which classifi es the drugs into four classes depending 
on their solubility and permeability (Figure 16 ). The BCS is applicable only to the 
oral route of administration, and according to this classifi cation, a drug is considered 
to be highly soluble if the highest dose of the drug is soluble in a glass of water 
(250 mL) covering the entire pH range in GIT from 1 to 7.5, and a drug is considered 
to be highly permeable if the drug has more than 90% oral bioavailability [48] . The 
model has been developed based on the solubility and permeability characteristics 

962 ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS 
of marketed drugs. However, BCS is bound to become an important biopharmaceutical 
tool in lead optimization in drug discovery setting and also at the same time 
serving as a useful guide to develop new formulations in a generic setting. From a 
regulatory perspective, BCS provides a scientifi c basis to grant biowaivers. As per 
BCS, the dissolution test can serve as a surrogate tool for costly and time - intensive 
bioequivalence studies for generic drugs which are highly soluble. This requires the 
establishment of a good in vitro – in vivo correlation. At present, the FDA grants 
biowaivers to generic manufacturers of immediate - release dosage forms for drugs 
in class I (highly soluble and highly permeable) provided they can prove the “ dissolution 
equivalence ” of their product to that of the innovator ’ s drug product [50] . 
FIGURE 15 Intestinal permeability studies. ( a ) Everted rat intestine is used to study the 
mechanism of drug absorption. Isolated rat intestine is fi lled with drug solution and tied at 
both ends and the drug permeation into the external medium is measured (left). In another 
set of experiment, the rat intestine is everted with the internal mucosal membrane facing 
outside (right) and the serosal side facing inward. If the drug is passively absorbed, then there 
would not be any differences in permeability in these two experiments. If the drug is transported 
by carriers, then drug permeation would be seen only in the fi rst experiment, as the 
carriers are present only on the mucosal side. ( b ) In situ intestinal perfusion studies are conducted 
in an anesthetized animal and the drug solution is pumped through a tube and drug 
coming out on the other side of the intestine is measured. The drug is also measured by 
sampling from the jugular vein in the animal. This is useful to measure the dynamic drug 
absorption into systemic circulation. 
(b) 
(a) 
Passive 
transport 
Drug solution 
Carriermediated 
transport 
pH 7.4 buffer 
Passive 
transport 
Drug solution 
pH 7.4 buffer 
Rat intestine 
Drug out 
Drug in

SOLUBILITY CHARACTERISTICS 963 
The dissolution equivalence is tested using the statistical dissolution model, termed 
f 2 or similarity factor, and is described by the equation 
f 
N 
R T t t 
t n 
2 
2 
1 
0 5 
50 1 
1 
100 = +( ) + ... 
... 
. ... 
...
= . 
. 
. log ( ) 
. 
(20) 
where N is the number of dosage form units (12 units are tested), t is dissolution 
time points from 1 to n, R t is the percent drug dissolved for the reference drug 
product, and T t is the percent drug dissolved for the test product. Two dissolution 
profi les are considered similar, if f 2 . 50 and if the coeffi cient of variation does not 
exceed 20% at early dissolution time points (usually 10 min) and 10% at other time 
points in the pH range 1 – 7.5. However, if . 85% of drug is dissolved in . 15 min, then 
no comparison is required and the dissolution is based on a single time point determination 
[50] . 
The BCS paradigm can be used to develop strategies for enhancing drug solubility 
and/or permeability (Tables 8 and 9 ). Solubility enhancement may involve only 
FIGURE 16 Biopharmaceutics classifi cation system. The drugs are classifi ed based on drug 
solubility and drug permeability. A drug is considered to be highly soluble if the highest dose 
of the drug is soluble in 250 mL of water varying in pH from 1 to 7.8 (GIT pH range). A drug 
is considered highly permeable if more than 90% of the drug is bioavailable by oral route. 
Class I drugs are highly soluble and highly permeable, class II drugs are poorly soluble but 
highly permeable, class III drugs are highly soluble but poorly permeable, and class IV drugs 
have poor solubility and permeability. 
e.g., Metronidazole, propranolol 
Class I Class II 
Class III Class IV 
e.g., Amhotericin B, taxol e.g., Atenolol, cimetidine 
e.g., Ibuprofen, griseofulvin 
Permeability Low 
Low 
High 
High Solubility

964 ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS 
TABLE 8 Solubility Enhancement Methods 
Technique Principle 
Salt formation Increases drug solubility by keeping the pH at which the drug is ionized. 
Particle size 
reduction 
Increase in surface area increases drug solubility. Particle size is reduced 
to nanodimensions (nanosuspensions) for increasing drug solubility. 
Change of 
form 
Crystalline drugs are converted to amorphous forms which show higher 
solubility than crystalline forms of the drug. 
Cosolvents Various water miscible cosolvents are used to increase the water solubility 
of drug. The cosolvents are selected based on the polarity of the drug. 
Common cosolvents include glycerol, ethanol, and propylene glycol. 
Complexation Drug is entrapped or complexed with excipients that can mask the 
lipophilic groups of the drug and enhance drug solubility in water. 
Cyclodextrins are commonly used to entrap hydrophobic drugs in the 
core, while the hydrophilic groups on the periphery help to solubilize 
the drug. 
Surfactants Surfactants are characterized by the presence of lipophilic and hydrophilic 
groups and form spherical structures known as micelles in water at 
a certain concentration. The hydrophobic drug is entrapped in the 
hydrophobic core of the micelle while the hydrophilic groups on the 
periphery help to solubilize the drug. 
Disperse 
systems 
The hydrophobic drug is dissolved in an organic solvent and in addition 
may also contain an emulsifi er. On contact with the intestinal fl uids, 
the drug is emulsifi ed (microemulsions) by bile salts and is absorbed 
through the intestine. 
TABLE 9 Permeability Enhancement Methods 
Method Mechanism 
Transcellular 
transport 
Sorption promoters can be used to enhance the transcellular transport 
in the intestine, including bile salts and fatty acid esters. They tend to 
fl uidize the lipid bilayer and enhance drug permeation across the 
membrane. 
Paracellular 
transport 
Enhancement is achieved by modulating the tight junctions between the 
cells. Chelating agents such as ethylenediamine tetraacetic acid can 
chelate calcium ions and transiently open the tight junctions for drug 
transport. 
Carrier - 
mediated 
transport 
Nutrient transport carriers are utilized for drug transport. Prodrugs are 
designed to meet the structural requirements for carrier - mediated 
transport. 
Blocking 
effl ux 
pump 
P - glycoprotein is a major effl ux mechanism that pumps out drug from the 
intestinal cells back into the intestinal fl uid. Several drugs and food 
substances are known to inhibit p - glycoprotein and enhance drug 
permeation. 
a physical intervention, as opposed to molecular modifi cation for permeability 
enhancement. Generally [46] , the permeability range of drugs varies by only 50 - fold 
(0.001 – 0.5 min . 1 ) in comparison to drug solubility, which varies by six orders of 
magnitude (0.1 . g/mL – 100 mg/mL). Hence, the formulator has greater fl exibility in 
altering the drug ’ s solubility in comparison to altering the drug ’ s permeability. This 

is evident form the fact [51] that the majority of the marketed drugs are highly 
soluble ( > 55% in classes I and III). Sometimes, enhancing the permeability by altering 
the drug ’ s chemical structure may be counterproductive. This because of the 
associated increase in molecular weight that attenuates the permeability enhancement 
gained with structural modifi cation. On the other hand, optimization of drug 
solubility may be more fruitful if the poor permeability is overcome by increasing 
drug solubility to provide a high drug concentration at the absorption site. However, 
this may be a diffi cult strategy if the dose is very high. The preformulation team 
should use the “ appropriate tools ” at every stage of the drug discovery and development 
process to guide or alert other development teams about drug solubility and 
permeability issues (Figure 17 ). 
6.2.4 STABILITY CHARACTERISTICS 
Drug stability is an essential component of preformulation testing. Establishing 
the stability of the drug under a variety of conditions is an expensive and time - 
consuming process. It cannot be overemphasized that the availability of a stability - 
indicating assay is the key to stability studies. The preformulation scientist works 
closely with the analytical method development team in developing a stability - 
indicating assay. During the early stages, a foolproof stability - indicating assay may 
FIGURE 17 Flow chart for determining absorption potential of a drug during drug discovery 
and development. Solubility and permeability studies from preclinical phase are used to 
calculate the maximum absorbable dose and, when correlated to BCS, this can provide information 
on its biopharmaceutics class. This is useful to estimate if the drug absorption would 
be dissolution and/or permeability limited for developing appropriate drug delivery 
strategies. 
Early drug discovery 
Exploratory studies 
Solubility/permeability 
Maximum absorbable dose 
BCS classification 
Ora ldosage form 
development 
Clinical development 
Drug development 
STABILITY CHARACTERISTICS 965

966 ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS 
not be available. This is understandable considering the fact that the initial drug lots 
are not pure and the purity is improved as the molecule progresses to subsequent 
development stages. Therefore, the intention of the preformulation scientist is not 
to generate a thorough kinetic rate profi le of the drug but to broadly defi ne the 
conditions under which the drug would be stable. Only relevant stability data are 
generated during various phases for developing preclinical and clinical formulations 
[52] . The stability data are evaluated and conveyed to the formulation development 
team upfront if stabilization and additional packaging requirements are needed. 
Often the chemical structure of the drug can give clues on the drug ’ s degradation 
pathway and its stability characteristics [53] . Table 10 lists some of the functional 
groups that are susceptible to common degradation pathways. 
Stability test conditions are chosen keeping in mind the environment that the 
drug will encounter during drug development, processing, storage, and use (Table 
11 ). One parameter is studied at a time while keeping all other parameters constant. 
Apart from classical stability studies, such as hydrolysis, oxidation, and photolysis, 
the preformulation scientist has to determine the stability of drugs in unconventional 
matrices such as animal feed used for toxicological studies [34] . The stability 
of a drug in the animal feed is complicated by the feed composition, including 
enzymes and vitamins among others. The moisture content in the feed may also vary 
with storage temperature. In such cases, it is appropriate to study the stability of the 
drug under the storage conditions encountered in the toxicological laboratory. Sensitive 
techniques such as liquid chromatography/mass spectrometry (LC/MS) are 
used to evaluate drug stability in such complex mixtures. 
In general, most of the drugs undergo fi rst - order degradation, while some drugs 
may follow zero - order kinetics and only a few drugs undergo second - order degradation 
[34] . The fi rst - and zero - order reactions are readily differentiated by studying 
drug stability at two different initial drug concentrations. First - order kinetics 
will depend on initial drug concentration, while a zero - order reaction will be 
TABLE 10 Groups Susceptible to Common Degradation Pathways 
Degradation 
Pathway Functional Groups 
Hydrolysis Esters, lactones, amides, lactams, oximes, imides, and malonic ureas 
Oxidation Amines, sulfi des, disulfi des, sulfoxides, phenol anions, thiols, nitriles, and 
catechols 
Photolysis Aromatic hydrocarbons, aromatic heterocyclics, aldehydes, and ketones 
TABLE 11 Stability Testing Conditions 
Parameters Conditions 
Temperature 5, 25, 30, 37, 40, and 60 ° C 
Moisture 30, 45, 60, 75, and 90% RH 
pH 1, 3, 5, 7, and 9 at room and body temperature 
Oxygen Sparging with 40% oxygen or adding 100 – 200 ppm of hydrogen peroxide 
Light 1.2 million lux hours of exposure to visible light and 200 h/m 2 exposure to 
UV light (360 – 400 nm) 

independent of initial drug concentration. Accelerated stability studies are conducted 
to expedite the degradative reactions, where temperature is the commonly 
used accelerant. The infl uence of temperature on drug stability kinetics is described 
by the Arrhenius equation: 
k Ae E RT = . a/ (21) 
where k is a reaction rate constant, A is a frequency factor, E a is activation energy, 
R is the gas constant, and T is absolute temperature. According to the Arrhenius 
equation, every 10 ° C rise in temperature increases the reaction rate by two - to fi vefold 
[31] . The usual temperatures selected for early stability studies include 5, 25, 37, 
40, and 60 ° C to cover the temperatures encountered in processing, use, and storage 
of the drug product. Using the Arrhenius equation, the rate constant from higher 
temperatures can be extrapolated to determine the stability at room temperature 
[31] . The slope of the plot of the reciprocal of temperature and the rate constant 
gives the activation energy. The activation energy usually varies between 15 and 
60 kcal/mol with a mean value of 19.8 kcal/mol [33] . A break in the line is usually 
indicative of change in the activation energy due either to change in the reaction 
order or the mechanism of degradation at higher temperature. In such cases, it 
becomes imperative to conduct detailed studies to understand the drug degradation 
mechanism. Some of the reactions seen at higher temperature may not be representative 
of the reactions at room temperature. Hence, short - term high - temperature 
studies should be supplemented with long - term real - time stability testing at room 
temperature. Additionally, the drug is also exposed to moisture, oxygen, and UV light 
(250 – 360 nm). The conditions used for stress studies may vary depending on the drug 
type and the drug development stage [54] . The stability studies in this chapter are 
discussed with respect to a solid dosage form which includes solid - state stability, 
limited liquid state stability, and drug – excipient compatibility. 
6.2.4.1 Solid - State Stability 
In general, solid - state reactions are slow, complex, and at times diffi cult to quantify. 
They may manifest as either physical and/or chemical instabilities. Physical instabilities 
include solid – solid transformations, desolvation of hydrates, and change in color 
[34] . On the other hand, chemical instabilities may involve a change in drug content 
as a result of hydrolysis, oxidation, or light - induced degradations [32] . The infl uence 
of temperature is studied by exposing the solid drug to increasing temperatures and 
also exposing the drug to various relative humidities at room temperature for two 
to eight weeks (Table 11 ). If substantial change is observed at higher temperatures, 
the drug samples stored at 5 ° C are analyzed. If no degradation is seen at higher 
temperature, then none can be expected at room temperature. The results from 
higher temperature should be carefully interpreted. For instance, a hydrate may lose 
moisture at higher temperatures and make a drug unstable which otherwise would 
be stable at room temperature. Similarly, chlortetracycline hydrochloride converts 
from the . form to the . form at above 65% relative humidity, in contrast to < 65% 
relative humidity, where no transformations are observed [32] . 
Oxidative degradation is studied by exposing the sample to an atmosphere of 
40%. The oxygen is combined with heat to accelerate the reaction. The results 
STABILITY CHARACTERISTICS 967

968 ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS 
should be compared with samples stored in an inert atmosphere. Photolytic degradation 
is studied by exposing the sample to UV light (254 and 360 nm) for two to 
four weeks and observed for color fading and/or darkening [33] . Color fading may 
not always mean drug decomposition. It may be just a physical change which can 
be overcome by including dyes in the formulation. As indicated earlier, the solid - 
state reactions may be slow, sometimes generating only 1 – 5% of degradation products, 
which may be below the detection limit of HPLC [34] . A combination of 
qualitative and quantitative analytical tools is helpful to detect drug degradation. 
Nevertheless, accelerated stability studies provide an early warning of potential drug 
degradation and the preformulation scientist should use discretion in interpreting 
and sharing the results with other development teams. 
6.2.4.2 Solution - State Stability 
Detailed solution - state stability is of limited value if the fi nal anticipated dosage 
form is a solid. However, solution - state stability studies can predict the stability of 
the drug in the granulating fl uid and the GI fl uids. Solution - state reactions are 
faster than the solid - state reactions. This is helpful in generating degradation products 
through so - called forced - degradation studies for toxicological screening and 
analytical method development. The studies are conducted by exposing the drug 
to extreme conditions such as 0.1 N HCl, 0.1 N NaOH, and water at 90 ° C [34] . 
Forced - degradation studies are useful to qualify the safety of the degradation 
products if it exceeds 0.1 or 0.05% of total daily dose for drugs with < 1 g or > 1 g 
dose/day, respectively [6] . 
The pH rate profi le is an important parameter that is studied in the solution state. 
In early preformulation studies, an approximate pH rate profi le is generated, usually 
including the pH encountered in salt selection and in vivo [55] . The studies are later 
followed with a detailed pH profi le in the whole pH range of 1 – 10. A typical pH 
rate profi le is shown in Figure 18 , which is useful to extract useful information on 
FIGURE 18 Representative pH kinetic rate profi le. The drug shows a minimal degradation 
at around 2 – 4 and the degradation rate is high in alkaline pH. 
pH 
2 4 6 8 10 
K (day–1)

drug stability. The profi le is used to predict if the drug degradation is catalyzed by 
hydronium or hydroxyl species (specifi c acid – base catalysis). The minimal point in 
the profi le is indicative of the pH at which the drug is relatively stable. However, 
the buffer used in the study can by itself accelerate the reaction, which is referred 
to as general acid – base catalysis [39] . The infl uence of buffer is nullifi ed by conducting 
the study at one to three buffer concentrations and then plotting the rate constant 
against the buffer concentration. The intercept on the y axis gives the rate 
constant at zero buffer concentration. 
Oxidation reactions are studied by passing oxygen in the head space of the drug 
solution and comparing the drug degradation with a drug solution (Table 11 ) fi lled 
with an inert gas in the head space. The reduced solubility of oxygen at higher temperatures 
may lead to an apparently reduced reaction rate in comparison to lower 
temperature [34] . Light sensitivity is studied by exposing the drug solution in a 
clear - fl int bottle to UV radiation and comparing the results with the drug solution 
in an amber - colored container [55] . 
The Solution - state studies should be extrapolated to the solid state with caution. 
The reaction in the solution state is usually done in a dilute drug solution, in contrast 
to reactions in the solid state, where the saturated drug solution at the surface 
undergoes a multiphase reaction [33] . Moreover, the reaction order may be different 
in the solution and solid states. Due to the excess solvent, reactions in the 
solution state are usually pseudo – fi rst order as opposed to fi rst - order or zero - order 
reactions in the solid state. In spite of these limitations, solution - state studies 
provide clues in selecting appropriate granulating solvent and in predicting in vivo 
drug stability. The pH – rate profi le data are also useful to predict the solid - state 
stability of salt forms or the stability of a drug in the presence of acidic and basic 
excipients [55] . Further, the pH – rate studies predict the stability of drug and its salt 
in the gastrointestinal pH. This is illustrated by the example of erythromycin and 
its salts [32] . Erythromycin is rapidly inactivated in the acidic environment of the 
stomach. This is overcome by using insoluble erythromycin estolate, which is stable 
in the gastric pH, unlike the other salts, which are easily displaced by hydrochloric 
acid in the stomach. 
6.2.4.3 Drug – Excipient Compatibility 
Excipients are the backbone of a solid dosage form, and they function as diluents, 
binders, disintegrants, and fi llers. The excipients are in intimate contact with the 
drug in a solid dosage form, therefore necessating the need for drug – excipient 
incompatibility testing. Since the formulator has a wide selection of excipients to 
choose from, it would be a daunting task for the preformulation scientist to screen 
all possible excipients. A general practice is to select those excipients that are 
routinely used in in - house manufacturing of dosage forms. At least two are selected 
from each class of functional excipients. Excipients that are known to cause potential 
incompatibilities (e.g., glucose and amines) with drugs are excluded from the 
study [34] . The excipients are intimately mixed with the drug in the ratio that is 
realistic for the desired solid dosage form. For example, diluents are mixed in the 
ratio 20 : 1, while other excipients are used in the ratio 1 : 5 with drug [32] . The 
drug – excipient mixtures are then stored in a tightly sealed container (ampoule or 
in a bottle, where the cap is sealed with wax) in the presence and absence of 5% 
STABILITY CHARACTERISTICS 969

970 ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS 
w/v moisture. The mixture is stored at 40 and 25 ° C and analyzed for three months. 
The mixture is physically observed for caking, liquefaction, and gas formation and 
chemically analyzed for drug degradation. A variety of techniques are used for 
studying drug – excipient compatibility. DSC is a useful primary screen which can 
rapidly detect potential drug – excipient incompatibilities [33] . For DSC studies, the 
drug and excipient are mixed together without moisture and are analyzed within 
a few hours of preparation. Moisture is avoided, as it complicates the interpretation 
of thermograms. If there is no interaction, then the thermogram should be the 
sum of thermogram of drug and excipient. The thermogram is observed for peak 
position, peak appearance, peak shape, appearance of new peaks, disappearance 
of drug peaks, peak shift, and change in enthalpy values. It is obvious that a mixture 
of two substances will result in depression of melting point, but if the change is 
signifi cant, then it may be indicative of eutectic formation. Some of the reactions 
seen at high temperatures in DSC may not be refl ective of room temperature. 
Therefore, excipients that are suspected to show incompatibilities are screened 
further through stability studies using TLC and HPLC (Figure 19 ). TLC is used to 
qualitatively detect any new spots with the drug – excipient mixture. The spots 
should be compared with the control drug sample. This is important since in the 
early discovery stage the drug is not pure and contains impurities. The fi ndings 
from TLC should be corroborated using HPLC. The studies may be further followed 
- up with stability studies using multicomponent mixtures. In addition, the 
drug and excipient may be tested by compressing in a hydraulic press or fi lling in 
a capsule to simulate the actual dosage form [32] . A well - designed drug – excipient 
preformulation study can thus help the formulator in judicious choice of excipients 
for the fi nal dosage form. 
FIGURE 19 Flow chart for studying drug – excipient compatibility. 
Use in formulation development 
Mix in suitable ratio
Excipient Drug 
Screen 
for interactions 
using DSC 
Interaction 
detected 
Short-term stability 
Long-term stability 
Characterize 
interaction 
by TLC and HPLC 
Yes 
No 
Excipient 
excluded 
Interaction 
confirmed

6.2.5 CONCLUSIONS 
Preformulation testing has a signifi cant role in a drug discovery and development 
program, as it provides valuable feedback to the various discovery and development 
teams in enabling druggability during lead identifi cation and optimization. 
The preformulation data may mean different things to different groups in the discovery/
development phase (Figure 20 ). For the chemistry team, the feedback from 
preformulation testing provides clues to optimize the chemical structure with 
respect to solubility, permeability, and stability. Preformulation studies give inputs 
to the biology group for ensuring optimal drug exposure based on solubility, permeability, 
and stability data, in addition to developing preclinical formulations for 
phar macokinetic and toxicological studies. The analytical team gets inputs from the 
preformulation group on developing stability - indicating assays and setting drug 
specifi cations. Once the lead is selected and as the molecule moves to the development 
phase, the preformulation group provides guidance to the bulk manufacturing, 
formulation, and clinical evaluation teams. The bulk manufacturing team uses 
the data generated from the preformulation studies on salts, polymorphic purity, 
and particle size specifi cations. It is the formulation team that utilizes the maximum 
data from the preformulation testing to design an appropriate dosage form. The 
physicochemical properties are utilized for improving the drug ’ s solubility, improving 
the drug ’ s permeability, developing stabilization strategies, selecting appropriate 
excipients, selecting processing conditions to design, and evaluating the fi nal dosage 
form. The clinical evaluation team utilizes the preformulation data along with the 
preclinical animal studies to understand the drug ’ s pharmacokinetics in humans 
through the MAD and BCS paradigms. Therefore, a strong preformulation team in 
a drug discovery setting is critical for optimizing the pharmaceutical properties of 
the drug. This can signifi cantly reduce the attrition rate, time, and cost of discovering 
a new drug. On the other hand, the preformulation team in a generic setting is 
valuable to optimize or further enhance the effi cacy of an existing drug by designing 
a new drug delivery system and thus giving a new life to an old drug. As opposed 
FIGURE 20 Role of preformulation in supporting other discovery and development 
teams. 
Manufacturing 
Drug discovery 
Biology 
Chemistry 
Preformulation 
Stability-indicating assay 
pH rate profile 
Impurity profile 
Solid-state stability 
Salt selectionPolymorphism, amorphous/crystallineHydrates/solvates, log P 
Preclinical formulation 
Solubility 
log P, permeability 
Analytical development 
Polymorphic purity 
Hydrate/anhydrate interconversions 
Particle size specifications 
Preformulation 
Clinical 
Formulation 
Drug development 
BCS 
MAD 
Solubility enhancement, permeability enhancementExcipient selectionProcessing conditions 
CONCLUSIONS 971

972 ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS 
to discovering a new drug, a new drug delivery system can be designed at one - third 
of the cost and money involved in a new drug discovery process [56] . The availability 
of high - throughput property screens and predictive models is expected to 
improve the effi ciency and maximize the output of preformulation data in the 
coming years. However, the main challenge lies in intelligent use of such tools to 
make sense from loads of data and communicate the appropriate information to 
the relevant discovery/development groups for getting the drug to the marketplace 
in time. 
REFERENCES 
1. Waterbeemd , H. V. , and Gifford , E. ( 2003 ), ADMET in silico modeling: Towards prediction 
paradise , Nat. Rev. Drug Discov. , 2 , 192 – 204 . 
2. Kerns , E. H. , and Di , L. ( 2003 ), Physicochemical profi ling in drug discovery , Drug Discov. 
Today , 8 , 316 – 323 . 
3. Lennarnas , H. , and Abrahamsson , B. ( 2005 ), The use of biopharmaceutical classifi cation 
of drugs in drug discovery and development: Current status and future extensions , 
J. Pharm. Pharmacol. , 57 , 273 – 285 . 
4. Lipinski , C. , Lombardo , F. , Dominy B. W. , and Feeney , P. J. ( 2001 ), Experimental and 
computational approaches to estimate solubility and permeability in drug discovery and 
development settings , Adv. Drug Deliv. Rev. , 46 , 3 – 26 . 
5. Giron , D. ( 1998 ), Contribution of thermal methods and related techniques to the rational 
development of pharmaceuticals. Part I , Pharm. Sci. Technol. Today , 1 , 191 – 199 . 
6. Ahuja , S. ( 2007 ), Assuring quality of drugs by monitoring impurities , Adv. Drug Deliv. 
Rev. , 59 , 3 – 11 . 
7. Datta , S. , and Grant , D. J. W. ( 2004 ), Crystal structures of drugs: Advances in determination, 
prediction and engineering , Nat. Rev. Drug Discov. , 42 , 42 – 57 . 
8. Tiwary , A. K. ( 2004 ), Modifi cation of crystal habit and its role in dosage form performance 
, Drug Dev. Ind. Pharm. , 27 , 699 – 707 . 
9. Garekani , H. A. , Ford , J. L. , Rubinstein , M. H. , and Rajabi - Sahboomi , A. R. ( 1999 ), Formation 
and compression characteristics of prismatic, polyhedral and thin plate - like crystals 
of paracetamol , Int. J. Pharm. , 187 , 77 – 89 . 
10. Gu , C. , Li , H. , Gandhi , R. B. , and Raghavan , K. ( 2004 ), Grouping solvents by statistical 
analysis of solvent property parameters: Implication to polymorph screening , Int. J. 
Pharm. , 283 , 117 – 125 . 
11. Giron , D. ( 2001 ), Investigations of polymorphism and pseudopolymorphism in pharmaceuticals 
by combined thermoanalytical techniques , J. Thermal Anal. Calor. , 64 , 37 – 
60 . 
12. Threafall , T. ( 2003 ), Structural and thermodynamic aspects of Ostwald ’ s rule , Org. Proc. 
Res. Dev. , 7 , 1017 – 1027 . 
13. Vippagunata , S. R. , Brittain , H. G. , and Grant , D. J. W. ( 2001 ), Crystalline solids , Adv. Drug 
Deliv. Rev. , 48 , 3 – 26 . 
14. Phadnis , N. V. , Cavatur , R. K. , and Suryanarayanan , R. ( 1997 ), Identifi cation of drugs in 
pharmaceutical dosage forms by x - ray powder diffractometry , J. Pharm. Biomed. Anal. , 
15 , 929 – 943 . 
15. Haleblain , J. , and McCrone , W. ( 1969 ), Pharmaceutical applications of polymorphism , 
J. Pharm. Sci. , 58 , 911 – 929 . 

16. Huang , L. , and Tong , W. ( 2004 ), Impact of solid state properties on developability assessment 
of drug candidates , Adv. Drug Deliv. Rev. , 56 , 321 – 334 . 
17. Morris , K. R. , Griesser , U. J. , Eckhardt , C. J. , and Stowell , J. G. ( 2001 ), Theoretical 
approaches to physical transformations of active pharmaceutical ingredients during 
manufacturing process , Adv. Drug Deliv. Rev. , 48 , 91 – 114 . 
18. Dunitz , J. A. , and Bernstein , J. ( 1995 ), Disappearing polymorphs , Acc. Chem. Res. , 28 , 
193 – 200 . 
19. Chemburkar , S. R. , Bauer , J. , Deming , K. , Spiwek , U. , Patel , K. , Morris , J. , Henry , R. , 
Spanton , S. , Dziki , W. , Porter , W. , Quick , J. , Bauer , P. , Donaubauer , J. , Narayanan , B. A. , 
Soldani , M. , Riley , D. , and McFarland , K. ( 2004 ), Dealing with the impact of ritonavir 
polymorphs on the late stages of bulk drug process development , Org. Proc. Res. Dev. , 4 , 
413 – 417 . 
20. Morissette , S. L. , Almarsson , O. , Peterson , M. L. , Remenar , J. F. , Read , M. J. , Lemmo , A. 
V. , Ellis , S. , Cima , M. J. , and Gardner , C. R. ( 2004 ), High - throughput crystallization: Polymorphs, 
salts, co - crystals and solvates of pharmaceutical solids , Adv. Drug Deliv. Rev. , 56 , 
275 – 300 . 
21. Davey , R. J. ( 2003 ), Pizzas, polymorphs and pills , Chem. Commun. , 13 , 1463 – 1467 . 
22. Khankari , R. K. , and Grant , D. J. W. ( 1995 ), Pharmaceutical hydrates , Thermochim. Acta , 
248 , 61 – 79 . 
23. Giron , D. , Goldbrown , Ch. , Mutz , M. , Pfeffer , S. , Piechon , Ph. , and Schwab , Ph. ( 2002 ), 
Solid - state characterization of pharmaceutical hydrates , J. Therm. Anal. Calor. , 68 , 
453 – 465 . 
24. Byrn , S. , Pfeffer , R. , Ganey , M. , Hoiberg , C. , and Poochikian , G. ( 1995 ), Pharmaceutical 
solids: A strategic approach to regulatory considerations , Pharm. Res. , 12 , 945 – 954 . 
25. Craig , D. Q. M. , Royall , P. G. , Kett , V. C. , and Hopton , M. C. ( 1999 ), The relevance of the 
amorphous state to pharmaceutical dosage forms , Int. J. Pharm. , 179 , 179 – 207 . 
26. Yu , L. ( 2001 ), Amorphous pharmaceutical solids: Preparation, characterization and 
stabilization , Adv. Drug Deliv. Rev. , 48 , 27 – 42 . 
27. Hancock , B. L. , and Parks , M. ( 2000 ), What is the true solubility advantage for amorphous 
pharmaceuticals? Pharm. Res. , 17 , 397 – 404 . 
28. Shah , B. , Kakamanu , V. K. , and Bansal , A. K. ( 2006 ), Analytical techniques for quantifi cation 
of amorphous/crystalline phases in pharmaceutical solids , J. Pharm. Sci. , 95 , 
1641 – 1665 . 
29. Giron , D. ( 2003 ), Characterization of salts of drug substances , J. Thermal Anal. Calor. , 73 , 
441 – 457 . 
30. Shekunov , B. Y. , Chattopadhyay , P. , Tong , H. H. , and Chow , H. H. ( 2007 ), Particle size 
analysis in pharmaceutics: Principles, methods and applications , Pharm. Res. , 24 , 
203 – 227 . 
31. Sinko , P. ( 2006 ), Martin ’ s Physical Pharmacy and Pharmaceutical Sciences , 5th ed. , 
Lippincott Williams and Wilkins , Philadelphia , pp 533 – 560 . 
32. Wadke , D. A. , and Jacboson , H. ( 1980 ), Preformulation testing , in Liberman , H. A. , and 
Lachman , L. , Eds., Pharmaceutical Dosage Forms: Tablets , Vol. 1, Marcel Dekker , New 
York , pp 1 – 59 . 
33. Wells , J. ( 2005 ), Pharmaceutical preformualtion , in Aulton , M. E. , Ed., Pharmaceutics, The 
Science of Dosage Form Design , Churchill Livingstone , Edinburgh , pp 113 – 138 . 
34. Fiese , E. F. , and Hagen , T. A. ( 1986 ), Preformulation , in Lachman , L. , Liberman , H. A. , 
and Kanig , J. A. , Eds., The Theory and Practice of Industrial Pharmacy , 3rd ed. , Lea and 
Febiger , Philadelphia , pp 171 – 196 . 
REFERENCES 973

974 ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS 
35. Krens , E. H. ( 2001 ), High - throughput physicochemical profi ling for drug discovery , 
J. Pharm. Sci. , 90 , 1838 – 1858 . 
36. Strickley , R. G. ( 2004 ), Solubilizing excipients in oral and injectable formulations , Pharm. 
Res. , 21 , 201 – 229 . 
37. Bastin , R. J. , Bowker , M. J. , and Slates , B. J. ( 2000 ), Salt selection and optimization 
procedures for pharmaceutical new chemical entities , Org. Proc. Res. Dev. , 4 , 427 – 
435 . 
38. Haynes , D. A. , Jones , W. , and Motherwell , W. D. S. ( 2005 ), Occurrence of pharmaceutically 
acceptable anions and cations in the Cambridge structural databases , J. Pharm. Sci. , 94 , 
2111 – 2120 . 
39. Florence , A. T. , and Atwood , D. ( 2005 ), Physicochemical Principles of Pharmacy , 4th ed. , 
Pharmaceutical Press , London , pp 55 – 92 . 
40. Ando , H. Y. , and Radebaugh , G. W. ( 2006 ), Property based drug design and preformulation 
, in Beringer , P. , DerMarclerosian , A. , Felton , L. , Gelone , S. , Gennaro , A. R. , Gupta , 
P. K. , Hoover , J. E. , Popovich , N. J. , Reilly Jr. , W. J. , and Hendrickson , R. , Eds., Remington ’ s: 
The Science and Practice of Pharmacy , Lippincott Williams and Wilkins , Philadelphia , pp 
720 – 744 . 
41. Leo , A. , Hansch , C. , and Elkins , D. ( 1971 ), Partition coeffi cient and their uses , Chem. Rev. , 
71 , 524 – 616 . 
42. Dokoumetzidis , A. , and Macheras , P. ( 2006 ), A century of dissolution research: From 
Noyes - Whitney to the biopharmaceutics classifi cation system , Int. J. Pharm. , 321 , 1 – 11 . 
43. Azaemi , A. , Roa , W. , and Lobenberg , R. ( 2007 ), Current perspectives in dissolution testing 
of conventional and novel dosage forms , Int. J. Pharm. , 328 , 12 – 21 . 
44. Costa , P. , and Lobo , J. M. S. ( 2001 ), Modeling and comparison of dissolution profi les , Eur. 
J. Pharm. Sci. , 13 , 123 – 133 . 
45. Yu , L. X. , Carlin , A. S. , Amidon , G. L. , and Hussain , A. S. ( 2004 ), Feasibility studies of 
utilizing disk intrinsic dissolution rate to classify drugs , Int. J. Pharm. , 270 , 221 – 227 . 
46. Curatolo , W. ( 1998 ), Physical chemical properties of oral drug candidates in the discovery 
and exploratory development settings , Pham. Sci. Technol. Today , 1 , 387 – 393 . 
47. Artursson , P. , Palm , K. , and Luthman , K. ( 2001 ), Caco - 2 monolayers in experimental and 
theoretical predictions of drug transport , Adv. Drug Deliv. Rev. , 46 , 27 – 43 . 
48. Amidon , G. L. , Lennernas , H. , Shah , V. P. , and Crison , J. R. ( 1995 ), A theoretical basis for 
a biopharmaceutic drug classifi cation: The correlation of in vitro drug product dissolution 
and in vivo bioavailability , Pharm. Res. , 12 , 413 – 419 . 
49. Stewart , B. H. , Chan , O. H. , Jezyk , N. , and Fleischer , D. ( 1997 ), Discrimination between 
candidates using models for evaluation of intestinal absorption , Adv. Drug Deliv. Rev. , 
23 , 27 – 45 . 
50. U.S. Department of Health and Human Services (DHHS) ( 2000 , Aug.), Guidance for 
Industry: Waiver of in vivo bioavailability and bioequivalence studies for immediate 
release solid oral dosage forms based on a biopharmaceutics classifi cation system, DHHS, 
Food and Drug Administration, Center for Drug Evaluation and Research, Washington, 
DC. 
51. Yamashita , S. , Yu , L. X. , and Amidon , G. L. ( 2006 ), A provisional biopharmaceutical classifi 
cation of the top 200 oral drug products in the United States, Great Britain, Spain and 
Japan , Mol. Pharm. , 3 , 631 – 643 . 
52. Xue - Qing , C. , Melissa , D. A. , Christoph , G. , and Gumundsson , S. O. ( 2006 ), Discovery 
pharmaceutics — Challenges and opportunities , AAPS J. , 8 , E402 – E408 . 
53. Guillory , J. K. , and Poust , R. I. ( 2002 ), Chemical kinetics , in Banker , G. S. , and Rhodes , 
C. T. , Eds., Modern pharmaceutics , Marcel Dekker , New York , pp 139 – 166 . 

54. IFAMA ( 2003 ), ICH stability testing of new drug substances and products, International 
Federation of Pharmaceutical Manufacturers Associations (IFPMA), Geneva. 
55. Carstensen , J. ( 2002 ), Preformulation , in Banker , G. S. , and Rhodes , C. T. , Eds., Modern 
Pharmaceutics , Marcel Dekker , New York , pp 167 – 185 . 
56. Pillai , O. , Dhanikula , A. B. , and Panchagnula , R. ( 2001 ), Drug delivery: An odyssey of 100 
years , Curr. Opin. Chem. Biol. , 5 , 439 – 446 . 
REFERENCES 975


977 
6.3 
TABLET DESIGN 
Eddy Castellanos Gil , 1,2,3 Isidoro Caraballo , 2 and 
Bernard Bataille 3 
1 Center of Pharmaceutical Chemistry and University of Havana, Havana, Cuba 
2 University of Sevilla, Seville, Spain 
3 University of Montpellier 1, Montpellier, France 
Contents 
6.3.1 Introduction 
6.3.2 Tableting Cycle 
6.3.3 Limitations for Direct Compression 
6.3.4 Previous Granulation: Biopharmaceutical Versus Technological Properties 
6.3.5 Tablet Design for Matrix System 
6.3.5.1 Controlled - Release Tablet by Direct Compression and Wet Granulation 
6.3.6 Tablet Design with Natural Products 
6.3.6.1 Tablet Design from Aqueous Plant Extract 
6.3.6.2 Natural Product as Vehicle for Manufactured Tablets 
6.3.6.3 Natural Product as Vehicle for Controlled - Release System 
6.3.6.4 Mechanism of Soluble Principle Active Propranolol Hydrochloride and 
Lobenzarit Disodium from Dextran Tablets 
6.3.7 Design Tools of Tablet Formulation 
6.3.7.1 MODDE 4.0 
6.3.7.2 iTAB 
6.3.7.3 Percolation Theory 
6.3.7.4 Artifi cial Neural Networks 
6.3.8 Coating Systems 
6.3.8.1 Subcoating of Tablet Cores as a Barrier to Water 
6.3.8.2 Kollidon VA 64 
6.3.8.3 SEPIFILM 
6.3.9 Development of Pharmaceutical Tablets Using Percolation Theory 
6.3.9.1 Case Study: Optimization of Inert Matrix Tablets for Controlled Release of 
Dextromethorphan Hydrobromide 
6.3.9.2 Critical Points of Hydrophilic Matrix Tablets 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

978 TABLET DESIGN 
6.3.9.3 Case Study: Estimation of Percolation Thresholds in Acyclovir Hydrophilic 
Matrix Tablets 
6.3.10 Ultrasound - Assisted Tableting (a New Perspective) 
References 
6.3.1 INTRODUCTION 
The compression of powders — the fourth state of matter in the words of Hans 
Leuenberger — is a complex task due to the variability in particle size and shape, 
even for particles of the same component, the unknown distribution of the particles 
in the die, their different ability to fl ow, and the forces needed to create bonds 
between them. 
Science - based formulation is nowadays a good strategy for the development of 
a pharmaceutical formulation. The new regulatory environment (process analytical 
technology) will transform science - based formulation in the next years. 
Therefore, there is a need for statistical tools able to predict the behavior of a 
powder mixture when it is subjected to compression forces. The purpose of this 
chapter is to provide a brief description of the main theoretical aspects of tablet 
design and formulation together with practical examples of tablet development and 
characterization using different techniques. 
6.3.2 TABLETING CYCLE 
Compressing powder or granule into a tablet is one of the simplest and oldest 
ways of forming a product known to humans. Nowadays, as technology has 
advanced, more complex machines are used with different procedures, but the basic 
principle, the tabletting cycle, remains the same. In tablet design many factors have 
to be taken into account, such as the physicolchemical properties of active compound 
and excipients. An important role also has to be attributed to tableting 
machines. 
Tablet press subclasses primarily are distinguished from one another by how the 
powder blend is delivered to the die cavity. Tablet presses can deliver powders 
without mechanical assistance (gravity) (e.g., Ronchii, Manesty, Stokes, and Colton 
machines), with mechanical assistance (power assisted) (e.g., Ronchii, Courtoy, 
Kilian, Manesty, Kikusui, Fette, and Hata machines), by rotational forces (centrifugal) 
(e.g., the Comprima machine), and in two different locations where a tablet 
core is formed and subsequently an outer layer of coating material is applied (compression 
coating) (e.g., Kilian Manesty, and Kikusui machines). 
The basic unit of any tablet press is tooling consisting of two punches and a die, 
called a station. The upper and lower punches come together in the die that contains 
the tablet formulation. Principally, two different types of machines are used, the 
eccentric and the rotary press. The eccentric press produces about 50 – 130 tablets 
per minute. The rotary press has a multiplicity of stations arranged on a rotating 
table with the dies. A few or many thousands of tablets can be produced per minute. 
There are numerous models of presses, manufactured by a number of companies, 
ranging in size, speed, and capacity. 

The eccentric press is widely used early in the development stage, because the 
tabletting machine and the tooling are inexpensive, it can be easily instrumented, 
little material is needed, and setting, servicing, cleaning, and tool changeover of the 
machine are easy . During the manufacturing process the tablet mixture is dosed by 
a hopper into the die. The position of the lower punch defi nes the volume of the 
subsequent tablet mass. The compression force is given by the position of the upper 
punch, which defi nes its immersion depth into the die, and the reagent force that is 
built up during the densifi cation of the material. The compressed tablet is ejected 
by the lower punch. 
During the compression process on an eccentric press, there are other pressure 
ratios at the upper and lower punches. The pressure at the upper punch is usually 
higher than the pressure at the lower punch. A part of the pressure is lost in the 
material and in the resulting radial friction force against the die wall during the 
compression [1, 2] . 
Figure 1 shows tablets (300 mg) of native dextran obtained from Leuconostoc 
mesenteroides B - 512F (Sigma) with a 10% water (w/w) punch in an eccentric 
machine (Manesty) at 15 kN with tablet side (fl at - fl ace diameter) 10 mm. Axial displacement 
of water was observed according to the change in color. 
On a rotary press the fi lling of the die and the following compression process is 
done at the same time at different stations. The compression is carried out in the 
simplest case with two rolls touching the upper and lower punches and compressing 
the powder mixture. In contrast to the eccentric press, the upper and lower punches 
exert pressure on the tablet mixture from both sides at the same time. 
Tablets compressed on a rotary press generally show a more consistent hardness 
when the upper and lower sides of the tablets are compared, where the upper 
side of tablets compressed on an eccentric press is usually harder than the lower 
side [3] . 
Figure 2 illustrates the difference in compression profi les of the upper and lower 
punches and the punch movement with fi ctitious rotary and eccentric presses. 
FIGURE 1 Native dextran tablet press in eccentric machine shows variability of color due 
to different pressure between upper and lower punches. 
TABLETING CYCLE 979

980 TABLET DESIGN 
Another reason, to use a rotary press rather than an eccentric press for tablet compression 
is the dwell time is usually shorter on a rotary press (see Figure 3 ). 
All compression mixes have an optimum compressing speed. This is why tablet 
press manufacturers install variable speed controls for the rotor or turret and the 
powder feeding mechanism. Many compression mixes are speed sensitive and will 
not produce satisfactory tablets at inappropriate speeds. The dwell time, where 
maximum pressure is applied to the mix, is relative to the peripheral speed of the 
turret and the diameter of the punch head fl at. Any air in the compression mix must 
be expelled to avoid laminating or splitting of the tablet. If air is compressed within 
the tablet, when the pressure applied by the punch is released, the compressed air 
expands and breaks the tablet. Precompression or dies with a taper in the bore will 
FIGURE 2 Punch movements and compression profi les of upper and lower punches of 
rotary and eccentric presses. 
Rotary press 
Eccentric press 
Lower compression force 
Upper compression force 
FIGURE 3 Dwell time as function of tableting machine. 
0.0 0.5 1.0 1.5 2.0 2.5 3.0 
0.0 
0.2 
0.4 
0.6 
0.8 
1.0 
Effective contact time 
Consolidation time 
Maximum UP force 
Dwell time 
Residence time

FIGURE 4 Stages of tableting process: ( a ) fi lling; ( b ) compression; ( c ) ejection. 
(a) 
(b) 
(c) 
often reduce this problem. Rotary presses sometimes have two pairs of compression 
rolls (for precompression and compression) . During precompression additional 
compression can take place and the absolute dwell time can be prolonged. In other 
words , there are numerous different tablet presses with various possibilities to carry 
out the compression process. 
A direct correlation between the results of an eccentric press and a rotary press 
cannot always be drawn. In addition there are many different tablet presses with 
different settings and possibilities. These problems can be overcome by using a 
compaction simulator early in the development stage. An advantage of such a simulator 
is its versatility, that is, all types of presses can be simulated with small amounts 
of solid. The problem, however, is that such a simulator is very expensive. 
The tableting cycle is well explained in the literature and is broken down into 
three stages (Figure 4 ): 
1. Filling The volume of the granule is measured (Figure 4 a ): 
A: The lower punch is allowed to descend to its lowest point. 
B: The bore of the die is fi lled completely with powder. 
C: The lower punch is raised to a predetermined point so that excess. 
D: The powder is leveled by passing under a blade. 
TABLETING CYCLE 981

982 TABLET DESIGN 
E: This ensures that the bore of the die is fi lled with as exact volume of the 
material to be used, and the next stage can begin. 
2. Compression Pressure is applied to form the granule into a solid (Figure 
4 b ): 
F: The upper punch is lowered into the bore of the die. 
G: Precompression gives the powder an initial “ punch ” to remove excess 
air. 
H: The powder is fully compressed. 
I: The correct pressure is reached. 
J: The upper punch is lifted out of the way ready for tablet ejection. 
3. Ejection The tablet is ejected and the next tablet will be formed (Figure 
4 c ): 
K: The lower punch begins to rise in the bore of the die lifting the tablet until 
step L is reached. 
L: Its base is level with the tap of the die. 
M: The tablet is pushed aside into the take - off chute by passing under a static 
blade. 
N: The lower punch moves to its lowest position ready for fi lling ( O ), similar 
to A and the entire cycle is repeated. 
Guidance on compression levels for each tablet type (series 1, 2, and 3) and maximum 
punch pressures are given in Table 1 : 
Series 1 : Flat fl ace, normal concave, shallow concave tablets 
Series 2 : Double radius, bevel and concave tablets 
Series 3 : Flat beveled edge, deep concave, ball or pill 
In Figure 4 the tablets progress from start to fi nish from left to right. On an actual 
machine this will be determined by the direction in which the entire turret rotates 
in relation to the fi xed item, such as the fi ll hopper precompressions and compression 
rollers. It is important to note that this direction may vary from machine to 
machine, but as a general rule, British, American, and some Asian machines 
rotate in a clockwise direction while European machines rotate anticlockwise 
(see Figure 5 ). 
6.3.3 LIMITATIONS FOR DIRECT COMPRESSION 
In tablet formulation, a range of excipient materials are normally required along 
with the active ingredient in order to give the tablet the desired properties. For 
example, the reproducibility and dose homogeneity of tablets are dependent on the 
properties of the powder mass. The tablet should also be suffi ciently strong to 
withstand handling but should disintegrate after intake to facilitate drug release. 
The choice of excipients will affect all these properties: 
1. Filler illers are used to make tablets of suffi cient size for easy handling by 
the patient and to facilitate production. Tablets containing a very potent active 

LIMITATIONS FOR DIRECT COMPRESSION 983 
FIGURE 5 Tableting machine (anticlockwise). 
TABLE 1 Guide Punch Pressures for Series 1, 2, and 3 
Tablet Size Pressure (kN) 
mm in. Series 1 Series 2 Series 3 
3 
1
8 
5 3 2 
4 
5 
32 
10 5.6 3.7 
5 
3 
16 
15 8.8 5.8 
6 
7 
32 
22 12 8.5 
7 
9 
32 
30 17 11 
8 
5 
16 
40 22 15 
9 
11 
32 
50 28 19 
10 
13 
32 
60 35 23 
11 
7 
16 
70 42 27 
12 
15 
32 
90 50 33 
13 1
2 100 59 39 
14 
9 
16 
120 67 46 
15 
19 
32 
130 78 53 
16 5
8 160 90 60 
17 
21 
32 
180 102 68 
18 
23 
32 
203 114 74 
19 3
4 226 127 85 
20 
25 
32 
251 141 94 
25 1 393 221 147 

984 TABLET DESIGN 
substance would be very small without excipients. A good fi ller will have good 
compactability and fl ow properties and acceptable taste and be non - hygroscopic 
and preferably chemically inert. It may also be advantageous to have a fi ller that 
fragments easily, since this counteracts the negative effects of lubricant additions 
to the formula [4] . 
2. Binder A material with a high bonding ability can be used as a binder to 
increase the mechanical strength of the tablet. A binder is usually a ductile material 
prone to undergo plastic (irreversible) deformation. Typically, binders are polymeric 
materials, often with disordered solid - state structures. Of special importance 
is the deformability of the peripheral parts (asperities and protrusions) of the binder 
particles [5] . 
This group of materials has the capacity of reducing interparticulate distances 
within the tablet, improving bond formation. If the entire bulk of the binder particles 
undergoes extensive plastic deformation during compression, the interparticular 
voids will, at least partly, be fi lled and tablet porosity will decrease. This increases 
contact area between the particles, which promotes the creation of interparticular 
bonds and subsequently increases tablet strength [6] . However, the effect of the 
binder depends on both its own properties and those of the other compounds within 
the tablet. A binder is often added to the granulation liquid during wet granulation 
to improve the cohesiveness and compactability of the powder particles, which 
assists the formation of agglomerates or granules. It is commonly accepted that 
binders added in dissolved form, during a granulation process, are more effective 
than those added in dry powder form during direct compression. 
3. Disintegrating Agent A disintegrant is normally added to facilitate the 
rupture of bonds and subsequent disintegration of the tablets. This increases the 
surface area of the drug exposed to the gastrointestinal fl uid; incomplete disintegration 
can result in incomplete absorption or a delay in the onset of action of the drug. 
There are several types of disintegrants, acting with different mechanisms: (a) promotion 
of the uptake of aqueous liquids by capillary forces, (b) swelling in contact 
with water, (c) release of gases when in contact with water, and (d) destruction of 
the binder by enzymatic action [7] . Starch is a traditional disintegrant; the concentration 
of starch in a conventional tablet formulation is normally up to 10% w/w. 
The starch particles swell moderately in contact with water, and the tablet disrupts. 
So - called superdisintegrants are now commonly used; since these act primarily by 
extensive swelling, they are effective only in small quantities [8] cross - linked sodium 
carboxymethyl cellulose (e.g., Ac - Di - Sol), which is effective in concentrations of 
2 – 4%, is a commonly used superdisintegrant. Larger particles of disintegrants have 
been found to swell to a greater extent and with a faster rate than fi ner particles, 
resulting in more effective disintegration [9] . 
4. Glidant, Antiadherent, and Lubricant Glidants are added to increase the 
fl owability of the powder mass, reduce interparticulate friction, and improve powder 
fl ow in the hopper shoe and die of the tableting machine. An antiadherent can be 
added to decrease sticking of the powder to the faces of the punches and the die 
walls during compaction, and a lubricant is added to decrease friction between 
powder and die, facilitating ejection of the tablet from the die. However, addition 
of lubricants (here used as a collective term and including glidants and antiadherents) 
can have negative effects on tablet strength, since the lubricant often reduces 

LIMITATIONS FOR DIRECT COMPRESSION 985 
the creation of interparticulate bonds [e.g., 4] . Further, lubricants can also slow the 
drug dissolution process by introducing hydrophobic fi lms around drug and excipient 
particles [e.g., 10] . These negative effects are especially signifi cant when long 
mixing times are required [11] . Therefore, the amount of lubricants should be kept 
relatively low and the mixing procedure kept short to avoid a homogenous distribution 
of lubricant throughout the powder mass. An alternative approach could then 
be to admix granulated qualities of lubricant [12] . 
5. Flavor, Sweetener, and Colorant Flavor and sweeteners are primarily used 
to improve or mask the taste of the drug, with subsequent substantial improvement 
in patient compliance. Coloring tablets also has aesthetic value and can improve 
tablet identifi cation, especially when patients are taking a number of different 
tablets. 
General instructions for the determination of tablet properties (e.g., hardness, 
disintegration, friability, dissolution profi le, and stability) are contained in pharmacopeia 
[e.g., European Pharmacopoeia (Eur. Ph.) and U.S. Pharmacopeia 
(USP)]. 
In the manufacture of tablets it is important to defi ne and appreciate the 
physical properties of the active substance, in particular particle size and 
fl owability. The technology involved in direct compression assumes great importance 
in tablet formulations because it is often the least expensive, particularly 
in the production of generics that the active substance permits. The limiting 
factors are the physical properties of the active substance and its concentration 
in the tablets. Even substances such as ascorbic acid, which are not generally 
suitable for direct compression owing to the friability of the crystals, can normally 
be directly pressed into tablets at concentrations of 30 – 40%. However, this technique 
is not as suitable if the content of ascorbic acid is higher. This limit may be 
shifted upward by special direct - compression auxiliaries, for example, Ludipress 
(BASF). 
Ludipress is derived from lactose, Kollidon 30, and Kollidon CL. It thus combines 
the properties of a fi ller, binder, disintegrant, and fl ow agent and also often 
acts as a release accelerator. By virtue of its versatility, formulations containing it 
are usually very simple. It can also be combined with almost all active substances 
with the exception of those that enter into a chemical interaction with lactose (Maillard 
reaction). Active substances (e.g., many analgetics) behave very differently 
with Ludipress when the dosage is extremely high. Acetylsalicylic acid (ASA) and 
metamizole can be pressed when little Ludipress has been added; ibuprofen requires 
a larger amount; and the fraction of Ludipress required in the tablets is too large 
for paracetamol (acetaminophen). 
An alternative to the Ludipress grades is the outstanding dry binder Kollidon 
VA 64 together with excipients (e.g., calcium phosphate, microcrystalline cellulose, 
lactose, or starch) and a disintegrant (e.g., Kollidon CL). This combination even 
allows 500 mg of paracetamol to be pressed into good tablets with a weight of 
700 mg. 
No other dry binder has binding power and plasticity comparable to Kollidon 
VA 64. Plasticity, in particular, is an important parameter in direct compression. 
As can be seen in Figure 6 (99.5% binder + 0.5% magnesium stearate), this property 
of Kollidon VA 64 is not adversely affected by increasing the pressure. The benefi cial 

986 TABLET DESIGN 
FIGURE 6 Plasticity of dry binders in tablets. 
0.4 
0.5 
0.6 
0.7 
0.8 
0.9 
1.0 
Plasticity 
HPMC 11,000 Microcrystaline 
cellulose 
Povidone K 30 Kollidon VA 64 
Compression force 25 kN 
Compression force 18 kN 
properties of Kollidon VA 64 can also be exploited for the production of concentrated 
active substance that is subsequently used for direct tableting. Kollidon VA 
64 and Ludipress can also be combined with one another. 
Acetylsalicylic Acid – Acetaminophen – Caffeine Tablet (250 mg + 250 mg + 
50 mg ) 
Formulation 
Acetaminophen powder 39.25% 
Caffeine powder 7.85% 
ASA powder 39.25% 
Kollidon VA 64 9.4% 
Kollidon CL 3.15% 
Aerosil 200 0.5% 
Magnesium stearate 0.6% 
Manufacture by Roller Compaction The active ingredients and Kollidon VA 64 
were granulated in a Gerteis Roller compactor. 
Process Parameters 
Force 5.0 kN/cm 
Crack 2.9 mm 
Looser 5.0 rpm 
Dosing screw 90.5 rpm 
Plug screw 108.6 rpm 
Granulator 50 rpm 
Tableting The granules were passed together with magnesium stearate, Aerosil 
200, and Kollidon CL through an 800 - . m sieve, blended for 10 min in a Turbula 

LIMITATIONS FOR DIRECT COMPRESSION 987 
FIGURE 7 Dissolution profi le for acetylsalicylic acid – acetaminophen – caffeine tablet 
(250 mg + 250 mg + 50 mg): paddle 50 rpm, 37 ° C deionized water. 
0 
20 
40 
60 
80 
100 
120 
0 
Time (min) 
Drug release (%) 
20 40 60 80 100 120 140 
mixer, and compressed into tablets with a force of about 12 kN (tablet press 
Korsch PH106, 30 rpm, compression force 11.5 kN) 
Tablet Properties 
Weight 629.7 mg 
S rel 1.4% 
Diameter 12 mm 
Form Biplanar 
Hardness 72 N 
Friability 2.75% 
Dissolution See Figure 7 . 
Enteric Film Coating of Tablets (Organic Solution) 
Formulation of Cores 
Component Percent/Tablet (w/w) 
ASA 33.4 
Ludipress 49.5 
Avicel PH 102 16.6 
Magnesium stearate 0.5 
Total 100.0 
Manufacture by Direct Compression All the components were mixed for 10 min, 
passed through an 0.8 - mm sieve, and compressed into tablets on a rotary press 
with a rate of 40,000 tablets/h at a compression force of 15 - kN. Core shape 
was convex with a diameter of 9 mm and the engraving BASF . Hardness of 
the tablets was about 60 N. 
Formulations of Coating Suspension The formulation is for 5 - kg cores (diameter 
9 mm; 300 mg): 

988 TABLET DESIGN 
Component 
Parts by Weight [g] 
No. 1 No. 2 
Kollicoat MAE 100 P 344.52 344.52 
Triethyl citrate 34.45 34.45 
Ethanol 4214.60 — 
Isopropanol/Wasser (70/30) — 4214.60 
Total 4593.57 4593.57 
Polymer applied 10.0 mg/cm 2 
Content of polymer 7.5% 
Preparation of Spray Suspension Kollicoat MAE 100P and triethyl citrate were 
stirred into the solvent until complete dissolution. 
Process Parameters 
Coating pan 24 Accela Cota 
Size of batch 5 kg 
Inlet air temperature 50 ° C 
Outlet air temperature 35 – 38 ° C 
Product temperature 30 – 35 ° C 
Inlet air rate 70 m 
Outlet air rate 140 m 
Spraying pressure 2.0 bars 
Nozzle diameter 1.0 mm 
Rate of spraying 30 g/min 
Time of spraying 2.5 h 
Preheating 3 min 
Final drying 5 min 
Dissolution: (See Figure 8 ). Dissolution was done according to USP monograph 
“ Aspirin Delayed - Release Tablets. ” 
FIGURE 8 Enteric fi lm coating of tablets, acetylsalicylic acid (organic solution): paddle 
100 rpm, 37 ° C; 0 – 2 h: 0.08 M HCl, 2 h+: phosphate buffer Ph 6.8. 
0 
20 
40 
60 
80 
100 
120 
0 50 100 150 200 
Time (min) 
Drug release (%) 
Ethanol 
Isopropanol/wasser 
(70/30)

LIMITATIONS FOR DIRECT COMPRESSION 989 
Beta Carotene Tablets (15 mg ) 
Formulation 
Formulation 1 Formulation 2 
Beta carotene dry powder 10% 160.0 g 150.0 g 
Ludipress 240.0 g — 
Dicalcium phosphate, granulated 
with 5% Kollidon 30 
— 175.0 g 
Avicel PH 101 — 100.0 g 
Kollidon CL 6.0 g 5.0 g 
Aerosil 200 — 2.5 g 
Talc — 20.0 g 
Calcium arachinate — 2.5 g 
Magnesium stearate 2.0 g — 
Manufacturing (Direct Compression) All components were mixed, passed 
through a 0.8 - mm sieve, and pressed with a medium compression force. 
Tablet Properties 
Formulation 1 Formulation 2 
Weight 400 mg 502 mg 
Diameter 12 mm 12 mm 
Form Biplanar Biplanar 
Hardness 59 N 57 N 
Disintegration 12 min 1 min 
Friability 0.1% 0% 
Chemical and Physical Stability (20 – 25 ° C) 
6 Months 12 Months 
Formulation 1 
Loss of beta carotene 3% 4% 
Hardness 60 N 59 N 
Disintegration 9 min 7 min 
Friability 0.15% 0.16% 
Formulation 2 
Loss of beta carotene 8% 9% 
Diclofenac Na – Dispersion – Tablet (50 mg ) 
Formulation 
Diclofenac Na 50.0 mg 
Avicel PH 102 143.8 mg 
Kollidon CL 50.0 mg 
Aerosil 200 5.0 mg 
Magnesium stearate 1.0 mg 

990 TABLET DESIGN 
Procedure The ingredients were mixed, passed through a 0.8 - mm sieve, and 
compressed into tablets with a force of about 10 kN. (The tablet press was 
Korsch PH106, 30 rpm, compression force was 11.8 kN. 
Tablet Properties 
Weight 248.0 mg 
S rel 1.7% 
Diameter 10 mm 
Form bipla Nar 
Hard Ness 93 N 
Friability < 0.1% 
Dissolution See Figure 9 . 
In direct – compression formulation, there is a wide particle size distribution. 
Usually, the active drug is at the fi ne end of the range. Such a wide particle size 
range can easily result in signifi cant segregation. Five primary mechanisms are 
responsible for most particle segregation problems [13] . Of these, only three typically 
occur with pharmaceutical powders: sifting, entrainment of air, and entrainment 
of particles in an air stream. 
Sifting is a process by which smaller particles move through a matrix of larger 
ones. It is by far the most common method of segregation. Sifting has been found 
to occur with particle size ratios as low as 1.3 : 1 or with a suffi ciently large mean 
particle size (the tendency to segregate by sifting decreases substantially with particle 
size < 500 . m. Free - fl owing material and interparticle motion also caused segregation 
by sifting. 
Two techniques can be used to decrease a material ’ s segregation tendencies: 
change the material or change the design of the equipment. 
Lisinopril (5 mg ) Reducing the ratio of excipient, lisinopril (2 : 1) tablets for direct 
compression can be obtained: 
FIGURE 9 Dissolution profi le for diclofenac – Na dispersion tablet (50 mg): paddle 50 rpm, 
37 ° C phosphate buffer, pH 7.2. 
0 
20 
40 
60 
80 
100 
120 
0 20 40 60 80 100 120 
Time (min) 
Drug release (%) 
Formulation 
Voltaren dispersion

FIGURE 10 Disolution profi le for lisinopril 5 mg: paddle 50 rpm, 37 ° C, HCl 0.1 N . 
0 
20 
40 
60 
80 
100 
120 
0 4 8 12 16 20 24 28 
Time (min) 
Lisinopril release (%) 
Formulation % 
Lisinopril 5.0 
Aerosil 0.5 
Calcium phosphate dibasic 19.8 
Starch 1500 2.0 
Magnesium stearate 0.7 
Cellulose microcrystalline (PH - 250) 72.0 
Lisinopril and Aerosil ( < 150 . m) were mixed for around 10 min. All other components 
were added and mixed for 15 min, passed through a 0.150 - mm sieve, and 
pressed with 10 kN compression force. 
Properties of 5 - mg lisinopril tablets are as follows: 
Weight 130 mg 
Diameter 8 mm 
Form Biplanar 
Hardness 98 N 
Disintegration 2 – 3 min 
Friability 0.05 
The content uniformity of the formulation was measured at the beginning, middle, 
and end of the batch (50 kg) (Table 2 ). 
Special attention should be given to the physical stability of the tablets manufactured 
by direct compression because some fi llers/binders are known to soften or 
harden on storage. 
6.3.4 PREVIOUS GRANULATION: BIOPHARMACEUTICAL VERSUS 
TECHNOLOGICAL PROPERTIES 
Granulation is the process by which primary powder particles are made to adhere 
to form larger, multiparticle entities called granules. Pharmaceutical granules 
BIOPHARMACEUTICAL VERSUS TECHNOLOGICAL PROPERTIES 991

992 TABLET DESIGN 
typically have a size range between 0.2 and 4.0 mm, depending on their subsequent 
use. In the majority of cases this will be in the production of tablets or capsules, 
when granules will be made as intermediate products and have a typical size range 
between 0.2 and 0.5 mm, but larger granules are used as a dosage form in their own 
right. 
Granulation normally commences after initial dry mixing of the necessary powdered 
ingredients so that a uniform distribution of each ingredient through the mix 
is achieved. After granulation the granules either will be packed or may be mixed 
with other excipients prior to tablet compaction. 
The principal reasons for granulation are as follows: 
1. To prevent segregation of the constituents of the powder mix. 
2. To improve the fl ow properties of the mix. 
3. To improve the compaction characteristics of the mix 
Methods of Granulation Granulation methods can be divided into two types: 
wet methods , which use a liquid in the process, and dry methods , in which no 
liquid is used. In a suitable formulation a number of different excipients will be 
needed in addition to the drug. The common types used are diluents, to produce a 
unit dose weight of suitable size, and disintegrating agents, which are added to 
aid the break - up of the granule when it reaches a liquid medium (e.g., on ingestion 
by the patient). Adhesives in the form of a dry powder may also be added, particularly 
if dry granulation is employed. These ingredients will be mixed before 
granulation. 
1. Dry Granulation In the dry methods of granulation the primary powder 
particles are aggregated under high pressure. There are two main processes: 
Either a large tablet ( slug ) is produced in a heavy - duty tableting press ( slugging ) 
or the powder is squeezed between two rollers to produce a sheet of material 
( roller compaction ). In both cases these intermediate products are broken down 
using a suitable milling technique to produce granular material, which is usually 
sieved to separate the desired size fraction. This dry method may be used for drugs 
TABLE 2 Study of Uniformity for Formulation Lisinopril 5 mg (Batch 50 kg) 
Number 0 – 5 kg 5 – 10 kg 10 – 15 kg 15 – 20 kg 20 – 25 kg 25 – 30 kg 35 – 40 kg 40 – 45 kg 45 – 50 kg 
1 4.99 5.00 5.00 4.95 5.02 5.08 5.00 4.96 5.00 
2 5.00 5.06 5.00 4.92 5.00 4.99 4.83 5.11 5.17 
3 5.00 4.81 4.90 5.19 4.92 5.03 4.92 5.00 5.03 
4 5.05 5.00 5.00 4.99 4.82 4.96 5.00 4.93 4.99 
5 4.89 4.99 4.90 5.05 5.13 5.04 4.98 4.93 4.87 
6 5.02 5.06 4.85 5.10 5.13 5.00 5.18 4.81 4.93 
7 5.00 5.08 5.11 5.00 5.00 4.83 4.89 5.00 5.00 
8 5.00 5.00 5.03 5.03 5.00 5.06 5.00 5.18 5.00 
9 4.97 4.99 5.01 5.04 5.05 4.85 5.00 5.00 4.99 
10 4.91 5.00 5.01 4.98 4.97 5.00 5.00 5.11 5.05 

that do not compress well after wet granulation or those which are sensitive to 
moisture. 
2. Wet Granulation Wet granulation involves the massing of a mix of dry 
primary powder particles using a granulating fl uid . The fl uid contains a solvent which 
must be volatile so that it can be removed by drying and be nontoxic. Typical liquids 
include water, ethanol, and isopropanol, either alone or in combination. The granulation 
liquid may be used alone or, more usually, as a solvent containing a dissolved 
adhesive (also referred to as a binder or binding agent ) which is used to ensure 
particle adhesion once the granule is dry. Water is commonly used for economical 
and ecological reasons. Its disadvantages as a solvent are that it may adversely affect 
drug stability, causing hydrolysis of susceptible products, and it needs a longer drying 
time than do organic solvents. This increases the length of the process and again 
may affect stability because of the extended exposure to heat. 
Captopril (25 mg ) + Hydrochlorothiazide (25 mg ) 
Formulation 
Formulation for 500 mg 
Captopril 5% 
Hydrochlorothiazide 5% 
Lactose 65% 
Carboxyethylcellulose sodium 10% 
Ac - Di - Sol 3% 
Starch 10% 
Stearic acid 2% 
Manufacturing (Wet Granulation) A mixture of all compounds (with 1.5% 
stearic acid) is granulated with solution 2 - propanol (around 8% v/w), passed 
through a 0.8 - mm sieve, and the rest (0.5% stearic acid) added and pressed 
with low compression force. 
Tablet Properties (Initial Time) 
Weight 500 mg 
Diameter 12 mm 
Form Normal concave 
Hardness 60 N 
Disintegration < 4 min 
Friability < 0.3% 
Dissolution (captopril + hydrochlorothiazide) 
30 min 90.00% 
60 min 100% 
Stability of Three Batches (5 kg each) at 25 ° C and 70% Relative Humidity (RH) 
during 12 Months 
BIOPHARMACEUTICAL VERSUS TECHNOLOGICAL PROPERTIES 993

994 TABLET DESIGN 
Formulation 
6 months 12 months 
Batch 1 Batch 2 Batch 3 Batch 1 Batch 2 Batch 3 
Hydrochlorothiazide (%) 98.91 99.47 100.65 99.06 100.40 102.88 
Captopril (assay) (%) 101.02 100.89 100.99 100.65 100.03 100.30 
Captopril disulfuric a 0.62 0.71 0.69 0.69 0.79 0.72 
Weight 500.12 501.23 499.65 499.00 500.33 500.14 
Hardness 63 N 67 N 61 N 60 N 63 N 61 N 
Disintegration (min) 3 3 4 4 4 4 
Friability 1.98 1.15 1.12 1.71 1.12 1.12 
Dissolution 
30 min 89.45 90.54 88.77 88.00 91.00 88.05 
60 min 100 100 100 100 100 100 
a Captopril degradation product (%). 
a - Methyldopa Tablet (250 mg ) 
Manufacturing (Wet Granulation) A mixture of . - methyldopa with lactose or 
calcium phosphate (for formulations F1 or F2 , respectively) is granulated with 
isopropanol solution of Kollidon 30 and passed through a sieve, the dry granules 
are mixed with Kollidon CL and magnesium stearate, and pressed with 
medium compression force. 
Tablet Properties 
F1 F2 
Weight 361 mg 362 mg 
Diameter 11 mm 11 mm 
Hardness 118 N 156 N 
Disintegration 5 min 4 min 
Friability < 0.1% < 0.1% 
Dissolution 
10 min 45% 55% 
20 min 82% 90% 
30 min 90% 98% 
TABLE 3 Comparative Study of Lactose Monohydrate and Calcium Phosphate, Dibasic 
Formulation F1 Formulation F2 
. - Methyldopa 275 g (78%) 275 g (78%) 
Lactose monohydrate 15.5% — 
Calcium phosphate, dibasic — 15.5% 
Kollidon 30 4% 4% 
Isopropanol 80 mL 80 mL 
Kollidon CL 2% 2% 
Magnesium stearate 0.5% 0.5% 
Formulation Table 3 presents a comparison of lactose monohydrate and calcium 
phosphate. 

Calcium phosphate, dibasic offers high hardness and faster dissolution profi le 
than lactose for . - methyldopa tablets in wet granulation. 
6.3.5 TABLET DESIGN FOR MATRIX SYSTEM 
The advantages of controlled - release systems include maintenance of drug levels 
within a desired range, the need for fewer administrations, optimal use of the drug 
in question, and increased patient compliance. Evaluation of matrix tablets is the 
same as for conventional formulations but the dissolution profi le and stability have 
to be carefully studied. Numerous methods for development of matrix tablets can 
be used, such as direct compression, wet granulation, pelletization, and spheronization 
exclusion. Nevertheless, the potential disadvantages cannot be ignored: the 
possible toxicity or no biocompatibility of the materials used, undesirable by - 
products of degradation, any surgery required to implant or remove the system, 
the chance of patient discomfort from the delivery device, and the higher cost of 
controlled - release systems compared to traditional pharmaceutical formulations. 
The importance of matrix systems that they release bioactive component over an 
extended period of time has long been recognized in the pharmaceutical fi eld. 
Matrix systems can be divided into three groups depending on the type of polymer 
formed: 
1. Inert Matrices Polymers that after compression form an indigestible and 
insoluble porous skeleton [14] constitute the inert matrices. The main challenge in 
the preparation of these systems is to achieve, by means of a suitable design, total 
drug release from the device as well as adequate and precise drug release, guaranteeing 
the integrity of the matrix. 
2. Hydrophilic Matrices Cellulose derivatives have been widely used in the 
formulation of hydrogel matrices for controlled drug delivery. Among them hydroxypropyl 
methylcellulose (HPMC) is the most extensively employed because of its 
ease of use, availability, and very low toxicity [15] . Drug release from these systems 
is controlled by the hydration of HPMC, which forms a gelatinous layer at the 
surface of the matrix through which the included drug diffuses. 
Drug release from swellable matrix tablets is based on the glassy – rubbery transition 
of the polymer which occurs as a result of water penetration into the matrix. 
Therefore, the gel layer is physically delimited by the erosion (swollen matrix – 
solvent boundary) and swelling (glassy – rubbery polymer boundary) fronts. 
Water - soluble drugs are released primarily by diffusion of dissolved drug molecules 
across the gel layer, while poorly water soluble drugs are released predominantly 
by erosion mechanisms. 
The factors infl uencing the release of drugs from hydrophilic matrices include 
viscosity of the polymer, ratio of the polymer to drug, mixtures of polymers, compression 
pressure, thickness of the tablet, particle size, pH of the matrix, entrapped 
air in the tablet, solubility of the drug, the presence of excipients or additives, and 
the mode of incorporation of these substances. 
3. Lipid Matrices These matrix tablets are formed with lipid polymers with low 
melting point. The drug is dissolved or solubilized in the melted lipid, such as cetyl 
TABLET DESIGN FOR MATRIX SYSTEM 995

996 TABLET DESIGN 
TABLE 4 Theophylline Formulation by Direct Compression and Wet Granulation 
(mg/tablet) 
Direct Compression Wet Granulation 
Granulated theophylline 264 264 
90SH - 4000SR HPMC (Metolose SR) 64.5 64.5 
Mg stearate 1.5 1.5 
Total 330 330 
alcohol, ceto - stearilic alcohol, and stearic acid. Solid lipid nanoparticles are an 
example of an innovative lipid matrix system. 
6.3.5.1 Controlled - Release Tablet by Direct Compression and 
Wet Granulation 
Theophylline is granulated in a fl uid bed with Pharmacoat 606 3% (Shin Etsu, 
Metolose SR) as shown in Figure 11 . Table 4 gives a comparison of direct compression 
(DC) and wet granulation (WG) using theophylline: 
Diret compression: using a twin - cell mixer, theophylline and HPMC are mixed 
for 10 min; then Mg stearate is added and mixed for 2 min. 
Tableting conditions for DC and WG: A rotary tableting machine (KIKUSUI) is 
used with 12 punches (punch size 10 mm diameter, 12 mm radius, compression 
force 98, 147, and 196 MPa; tableting speed 20, 40, and 60 min . 1 ). 
Condition for WG: Granulation machine, vertical granulator FM - VG - 05; charge 
300 g; binder solution ethanol – water 8 : 2; agitation 600 (blade)/1000 (chopper) 
min . 1 ; granulation time 5 min. 
Powder properties for compression: Bulk density 0.35 g/mL, tapped density 0.48, 
average particle size 122 . m. 
Theophylline tablets made by both direct compression and wet granulation have 
been assessed. There is almost no difference between direct compression and wet 
granulation methods (see Figures 12 – 14 ) under the following conditions: appropriate 
formulation (suffi cient level of HPMC in the tablet) and precise control of the 
wet granulation process. Direct compression using Metolose SR is recognized as a 
suitable process for matrix tablets. 
6.3.6 TABLET DESIGN WITH NATURAL PRODUCTS 
The development and production of tablets containing a high dose of active ingredients 
is a complex and extensive technological challenge. Dried plant extracts are 
often used as therapeutically active material in the manufacture of tablets. They are 

FIGURE 11 Granulation process: ( a ) before 90SH - 4000SR; ( b ) after 90SH - 4000SR. 
(a) (b) 
FIGURE 12 Dissolution profi le for theophylline tablets (DC and WG). 
0 
20 
40 
60 
80 
100 
0 3 6 9 12 15 18 
Time (h) 
Theophylline release 
(%) 
WG 
DC 
FIGURE 13 Hardness of theophylline tablets (Dc and WG). 
0
5 
10 
15 
20 
25 
75 100 125 150 175 200 225 
Compression force (MPa) 
Tablet hardness 
(kgf)
WG 
DC 
FIGURE 14 Weight deviation for theophylline tablets (DC and WG). 
0
1
2 
0 20 40 60 80 
Compression speed (min–1) 
Tablet weight 
deviation (CV %) 
WG 
DC 
TABLET DESIGN WITH NATURAL PRODUCTS 997

998 TABLET DESIGN 
often very fi ne, poorly compressible, and very hygroscopic powders. Tablets containing 
a high amount of spray - dried extract show prolonged disintegration times, 
affecting the release of active constituents [19] . Some alternatives have been proposed 
to minimize these problems. Granulation is the technique most often used to 
improve the technological properties of these products. However, because of the 
products ’ high hygroscopicity, extracts sometimes cannot be granulated using 
aqueous systems. Some reports have shown that the use of lubricants during direct 
compression of vegetable dried extracts increased the disintegration time. According 
to our experience and some previous work, tablets with high amounts of some 
lubricant such as aerosil (up to 25% w/w) and magnesium [16] stearate incorporated 
into the granules had shorter disintegration time than did tablets containing the 
powdered mixture. 
Natural products can be used as plant extracts with pharmacological activity 
(e.g., Mangifera indica L., vallerian, aloe, Cratoxylum prunifl orum , microporous 
zeolite), excipient for direct compression or granulation (chitin, chitosan, and 
dextran), and controlled - release systems (cellulose and native dextran). For natural 
products the most important factor is the standardization of the extract because 
properties such as the amount of active substance can be changed from batch to 
batch. Factors such as the origin of the extract, geographic zone, and age of the tree 
could affect the properties of natural extracts. 
6.3.6.1 Tablet Design from Aqueous Plant Extract 
A bioactive product of natural origin has been developed from folk knowledge 
of Asian, Latin - American, European, and U.S. ethnic medicine. We developed 
an extract of the M. i ndica L. (mango) stem bark, obtained by decoction of some 
varieties grown in tropical and subtropical climates, that is used at present as an 
antioxidant nutritional supplement (Vimang). The aqueous extract was dried by 
atomization in a spray dryer until a brown solid with 10.5% (RSD = 0.9%) water 
content (measured by Karl Fischer) was achieved. Tablets obtained by wet granulation 
(plant extract, 300 mg/unit) were used for the applications. The product is a 
fi ne brown powder that has provend to be useful in the treatment of a large population 
sample presenting physical stress due to age or deteriorated physiological 
conditions caused by chronic diseases such as cancer, diabetes, or cardiovascular 
disorders [17] . Recent studies have shown that treatment with the extract provided 
signifi cant protection against 12 - O - tetradecanoylphorbol - 13 - acetate (TPA) – 
induced oxidative damage and better protection when compared with other 
antioxidants (Vitamin C, E and beta – carotene) [18] . Furthermore, the results 
indicate that this extract is bioavailable for some vital target organs, including 
liver and brain tissues, peritoneal cell exudates, and serum. Therefore, it was 
concluded that it could be useful to prevent the production of reactive oxygen 
species (ROS) and oxidative tissue damage in vivo. All these effects are likely due 
to the synergic action of several compounds, such as polyphenols, terpenoids, steroids, 
fatty acids, and microelements, which have been reported to be present in 
the extract [17] . 

Mangiferin (1,3,6,7 - tetrahydroxyxanthone - 2 - C - . - d - glucopyranoside), a C - glucosylxanthone, 
which was fi rst isolated from the bark, branches, and leaves of M. indica 
L., has been found to be the major component of this extract. Mangiferin is a naturally 
occurring chemopreventive agent in rat colon carcinogenesis [19] ; exerts antidiabetic 
activity by increasing insulin sensitivity; shows signifi cant inhibitory effect 
on bone resorption; appears to act as a potential biological response modifi er with 
antitumor, immunomodulatory, and anti – human immunodefi ciency virus (HIV) 
effect; is capable of providing cellular protection as an antioxidant and a radical 
scavenger agent; is useful as an analgesic without adverse effects; and inhibits the 
late event in herpes simplex virus - 2 replication [20 – 24] . 
The quality control of 16 batches of Vimang active ingredient [by high - 
performance liquid chromatography (HPLC) and the ultraviolet (UV) method] 
obtained from different regions of the country (batches 1 – 8 from the west and 
batches 9 – 16 from the east) and its pharmaceuticals (optimum formula) were investigated 
and are demonstrated in Table 5 . Each sample was analyzed in triplicate and 
the average values are listed. All assay results fell between 100 and 300 . g of mangiferin 
per milligram of Vimang powder, except samples 12 – 16, which were rejected. 
The differences found are probably due to the fact that the mangiferin content in 
the plant varies with the season of the year and the zone where it was grown. The 
claimed contents of this natural product required by our producers are 85 – 115% 
for tablets. 
Different tablet formulations were tested, but even superdisintegrants such as 
Ac - Di - Sol and CMCNa up to concentrations of 5% were not suffi cient to disintegrate 
the tablets. With the use of pH - modifi ed product such as NaCO 2 or canalling 
as NaCl the release of extract from the tablets changed dramatically. Use of lactose 
TABLET DESIGN WITH NATURAL PRODUCTS 999 
TABLE 5 Content of Mangiferin ( m g/mg) in Natural Product Samples from Mangifera 
indica L. and assay Result of Pharmaceuticals 
Sample 
Content, . g/mg 
(RSD, %) Sample 
Content, . g/mg 
(RSD, %) 
No.1 (batch 901) 254 (0.7) No.9 (batch 0201) 125 (0.1) 
No.2 (batch 903) 195 (2.0) No.10 (batch 0202) 109 (1.5) 
No.3 (batch E - 1923) 187 (1.6) No.11 (batch 0203) 116 (1.2) 
No.4 (batch E - 1924) 180 (1.4) No.12 (batch 0204) 79 (2.4) * 
No.5 (batch E - 2032) 206 (1.6) No.13 (batch 0205) 56 (0.5) * 
No.6 (batch 0103) 149 (5.7) No.14 (batch 0206) 55 (2.3) * 
No.7 (batch 0104) 162 (0.4) No.15 (batch 0207) 66 (1.8) * 
No.8 (batch 0112) 159 (0.3) No.16 (batch 0208) 49 (0.2) * 
Pharmaceuticals from 
batch No.8 
Amount of Vimang ® 
(mg) 
Percentage of claimed content %, 
(RSD, %) 
Tablets (batch A) 299.91 99.97 (5.47) 
Tablets (batch B) 291.36 97.12 (3.14) 
Tablets (batch C) 310.51 103.51 (1.42) 
* RSD = relative standard deviation. 

TABLE 6 Formulations by Wet Granulation 
F15 F16 F17 F18 F19 F20 F21 F22 F23 F24 F25 F26 F27 F28 F29 F30 
Extract a 
50 50 50 50 50 50 50 50 50 50 50 50 50 50 50 50 
Lactose — — 20 a 
16 
a 
16 
a 
22 
a 
27 
a 
29 
a 
32 
a 
42 
a 
32 a 
+ 
10 35 a 
35 
a 
35 
a 
35 
a 
30 
a 
CMM 20 a 
20 
a 
NaCl 20 a 
15 a 
+ 
5 
10 a 
5 
a 
5 
a 
5 
5 
5 
5 
— 
— 
— 
5 
PEG6000 — — — — — — — — — — — — — 5 5 5 
PEG400 a 
— 
— 
— — 
— 
— 
— 
— 
— 
— 
— 
— 
— 
4 
Na 2 CO 3 
— 
— 
— 
5 a 
+ 
5 
10 a 
5 
5 
5 
5 
— 
— 
5 
— 
— 
— 
5 
CMCNa — — 5 
PVP a 
5 
5 
5 
4 4 
4 
4 
4 
4 
4 
4 
4 
4 
4 
4 
SDS 2 a 
1 a 
+ 
1 
1 
a 
+ 
1 
1 
1 
2 
2 
2 
2 
2 
2 
2 
2 
2 
Talc 1 1 — 1 1 2 2 2 2 4 4 4 4 4 4 4 
MgEs 1 1 1 1 1 
Aerosil 1 1 5 a 
+ 
2 
5 
a 
+ 
2 
5 
a 
+ 
2 5 a 
Acdisol — — — 5 5 5 a 
5 
3 
% 62% 65% 71.5% 89% 85% 88% 100% 100% 100% 46% nF 80% 70% 71% 72% 100% 
Release 
IN 31 IN 34 IN 23 
In 30 Min 
MIN MIN MIN 
Abbreviations: CMM, cellulose microcrystalline; PEG, polyethylene glycal; CMCNa, sodium carboxymethyl cellulose; PVP, Kollidon K30; SDS, sodium docecyl 
sulfate; MgEs, magnesium stearate; NF, no fl ow ability. 
a Internal phase. 
1000

TABLE 7 Two Formulations for mangifera indica L . (Mango) 
Formulation F Formulation F31 
Internal Phase 
Extract M. indica L. 50.0% a 50.0% a 
Lactose 31.0% a 
Povidone (PVP K30) 4.0% a 4.0% a 
Starch 10.0% a 
CMM 28.0% a 
Ac - Di - Sol 5.0% a 
External Phase 
SDS — 2.0% 
Na 2 CO 3 — 5.0% 
PEG6000 — 5.0% 
Talc 2.5% 2.5% 
Magnesium stearate 0.5% 0.5% 
a Product added to internal phase. 
FIGURE 15 Formulation F31. 
was better than CMM, especially for fl ow properties. In all cases tablets always 
eroded and never disintegrated. All formulations studied are shown in Table 6 . 
F23 and F30 are shown in Figure 15 . Two formulations, F31 (obtained from the 
result of F23 and F30 ) and F, were compared for water uptake and dissolution 
profi le, as shown in Table 7 . 
The properties of formulations F31 and F were measured in ENSLIN equipment 
in order to compare the kinetics of water uptake and to study the dissolution profi le, 
as shown in Figures 16 – 19 . As observed in Figure 19 , the dissolution of plant extract 
after 60 min is very slow when pH - modifi ed ( NA 2 CO 3 ) and sodium dodecyl sulfate 
(SDS) are not used (almost not change in color for formulation F ). 
The properties of formulation F31 are as follows: diameter 12 mm, normal 
concave; stability studies (see Table 8 ) under tropical condition, 30 ° C and 75% RH 
with desiccant (silice 1 g for plastic bottle of polyethylene no. 8 for 60 tablets). 
TABLET DESIGN WITH NATURAL PRODUCTS 1001

1002 TABLET DESIGN 
FIGURE 16 Formulation F31 in ENSLIN equipment at initial time. 
FIGURE 17 Formulation F31 in ENSLIN equipment after 60 min. 
6.3.6.2 Natural Product as Vehicle for Manufactured Tablets 
Chitin was evaluated as a direct - compression vehicle using powder fl ow properties 
and the physicomechanical properties of the manufactured tablets, and it was proven 
that this natural polymer has suitable characteristics for being used for this end. 
A comparative study of chitin obtained from lobsters, starch, and carboxymethyl 
chitosan as disintegrating agents was conducted. The infl uence of the method in the 
preparation of tablets on the disintegrating activity of both polymers was evaluated. 
Chitin proved to have good characteristics as a disintegrating agent independently 

FIGURE 18 Formulation F in ENSLIN equipment at initial time. 
FIGURE 19 Formulation F in ENSLIN equipment after 60 min. 
TABLE 8 Stability Studies for F31 under Tropical Condition 
Initial Time 3 months 6 months 9 months 12 months 
Weight 600.7 601.47 601.30 602.09 601.33 
Assay 99.85 100.98 100.64 99.10 101.07 
Disintegration, min 27 29 30 30 30 
Hardness, N 59 60 64 60 60 
Friability, % 0.89 0.74 0.80 0.77 0.81 
Humidity, % 5.71 5.11 5.45 5.21 5.64 
TABLET DESIGN WITH NATURAL PRODUCTS 1003

1004 TABLET DESIGN 
TABLE 9 Formulation with Chitin, Chitosan, and Croscarmelose Sodium 
I II III IV V VI VII 
Papaverin 19.25 18.25 17.50 18.25 17.50 18.25 17.50 
Binder 10.00 10.00 10.00 10.00 10.00 10.00 10.00 
Chitin — 1.00 1.50 
Chitosan — — — — — 1 1.50 
Croscarmelose sodium — — — 1.00 1.50 
Dibasic calcium 
phosphate 
70.00 70.00 70.00 70.00 70.00 70.00 70.00 
Magnesium stearate 0.75 0.75 0.75 0.75 0.75 0.75 0.75 
TABLE 10 Results for Direct Compression 
I II III IV V VI VII 
Weight, mg 348 347 345 347 348 346 347 
Release at 30 min, % < 40 92.6 92.4 94.2 96.6 92.1 94.8 
Disintegration, min > 60 2.30 1.40 0.28 0.22 1.30 1.10 
Hardness, kgf - Erw 3.92 3.99 3.97 3.97 4.03 3.99 3.92 
Friability 2.44 2.24 2.10 2.01 1.97 1.89 1.94 
TABLE 11 Results for Wet Granulation 
FI FII FIII 
Weight, mg 346.50 348.20 349.00 
Release at 30 min, % 81.00 86.00 < 70.00 
Disintegration, min 23.70 23.00 45.30 
Hardness, kgf - Erw 6.90 8.20 6.30 
Friability 0.14 0.08 0.13 
of the method used to make tablets (Tables 9 – 11 ) . The disintegrating activity of 
carboxymethyl chitosan was affected by the granulation process. 
Three formulas were prepared by wet granulation comparing starch, chitin, and 
chitosan as disintegrant (10%) formulations FI (10% starch), FII (10% chitin), and 
FIII (10% chitosan) (Table 11 ) . Chitin has good properties as disintegrant. This 
product can be used for direct compression and wet granulation. The method for 
development (direct compresion or wet granulation) of tablets infl uences chitosan 
disintegrant properties. 
6.3.6.3 Natural Product as Vehicle for Controlled - Release System 
Dextrans are composed of chains of d - glucan (1 – 20 . 10 6 ) with . - 1,6 as the main - 
chain linkage and variable numbers of . - 1,2, . - 1,3, or . - 1,4 branched - chain linkages. 
Dextran is synthesized from sucrose by dextransucrases, glucansucrases, and glucosyltransferases 
produced by Leuconostoc or Streptococcus . These bacteria growing 
in sugar juices produce dextran. High concentrations of dextran on solids ( > 1000 ppm) 
can result in severe fi nancial losses to the sugar industry [25] . 

Dextran fractions obtained from enzymatic hydrolysis of native dextrans are 
supplied in molecular weights from 1000 to 2 . 10 6 Da. The molecular weight of the 
fraction is in most cases a key property and is defi ned in terms of the average 
molecular weight ( M w ) and the number average molecular weight ( M n ). The functionality 
of this raw material for controlling drug release is studied as a function of 
molecular weight. Fractions 43,000 ( F3 ), 71,000 ( F2 ), and 170,000 ( F1 ) as native 
dextran 2 . 10 6 and 20 . 10 6 M w are used [26] . 
Wet - and Dry - Weight Studies The method used was based on that of Tahara [15] 
and Jamzad [27] . The swelling and erosion of dextran polymers of differing molecular 
weights were examined by measuring the wet and subsequent dry weights of 
matrices. The experiment consisted of allowing the tablet (dextran alone ) to dissolve 
in the medium (at the same condition described in drug release studies) for 
certain time periods (15, 30, 60 and 90 minutes) before being removed into a preweighed 
weighing boat. The excess dissolution medium was drained and blotted 
from around the tablet without touching it. The tablet and boat were then weighed 
to establish the wet weight of the tablet. The tablets were then dried to a constant 
weight in an oven at 105 ° C. Each determination at each time point was performed 
in triplicate and mean values were expressed. The dissolution medium uptake per 
weight of dextran remaining was calculated at each time point for a particular matrix 
to correct for the effect of erosion and dissolution in the measurement of degree of 
dissolution medium uptake [Equation (1) ]. Erode dextran was measured according 
to the equation described by Jamzad et al. [30] [ Equation (2) ]: 
Water uptake per unit 
polymer remaining 
wet weight dry weight 
d (%) = . 
ry weight 
. 100 (1) 
Mass polymer loss (%) 
original weight remaining (dry) weight 
orig 
= 
.
inal weight 
. 100 (2) 
The Davidson and Peppas model [Equation (3) ] was applied to these data to study 
the mechanism and the rate of water uptake. 
w Ktn = s (3) 
where w is the weight gain of the swelled matrix (water/dry polymer), K s the kinetic 
constant of water penetration, t the penetration time, and n the exponent which 
depends on the water penetration mechanism. 
Swelling and Erosion The change in wet weight, refl ecting swelling, over time for 
the fi ve polymer types is shown in Table 12 . The higher molecular weight polymers 
showed the highest maximum average relative swelling, which occurred since the 
initial time with little erosion. In contrast, the lower molecular weight polymers 
(fractions F1 , F2 , and F3 ) exhibited minimal swelling and the erosion mechanism 
predominated. Consequently, tablets were dissolved very fast (100% before 45 min). 
These polymers and fractions show a wide range of viscosities, which cause differences 
in their swelling and erosion behaviors [26] . These results agree with results 
obtained by Sakar [C] and Walker [D] for HPMC polymer. 
TABLET DESIGN WITH NATURAL PRODUCTS 1005

1006 TABLET DESIGN 
TABLE 12 Swelling and Erosion Properties of Dextran Tablets as Funtion of Molecular 
Weight 
Hardness (N) 
Mass Polymer Loss (%) 
15 min 30 min 60 min 90 min 
DT, MW 2 . 10 6 431 11.48 14.59 27.473 36.224 
DT, MW 20 . 10 6 482 5.29 7.33 11.835 16.153 
F1 431 52.89 87.38 
F2 420 62.79 89.21 
F3 460 66.3 95.85 
Percentage of Water Uptake 
15 min 30 min 60 min 90 min 
DT, MW 2 . 10 6 59.80 84.68 103.73 110.15 
DT, MW 20 . 10 6 110.03 142.94 178.50 210.02 
F1 31.61 33.14 
F2 17.61 26.89 
F3 16.82 17.65 
Note: All values are referred to applied force, 14 kN, particle size for dextran 150 – 200 . m, tablet weight 
300 ± 11 mg, dissolution media 1000 mL distilled water, temperature 37 ° C, 100 - rpm paddle. Abbreviations: 
DT, dextran; MW, molecular weight. 
For native dextran a linear relationship was seen between mass polymer loss and 
initial dissolution time. Native dextrans also showed the highest maximum dissolution 
medium uptake. Here, an increase in the molecular weight of dextran resulted 
in an increase in water uptake (native polymer with 10 times more than fraction F1 , 
13 times more than F2 , and 15 times more than F3 for the fi rst 30 min) and less 
erosion. Anywhere the rate of water uptake per unit weight of polymer started 
to decline with last initial time and in consequence for longer periods of time, nonlinear 
dependence could be expected. Applying the Davidson – Peppas model [Equation 
(3) ], a value of n = 0.356 ( r 2 = 0.998) for native dextran was obtained 
( r 2 = 0.984). An inverse relationship between erosion rate constant and molecular 
weight was reported by Reynolds et al. [28] . Tahara et al. [18] reported that the 
lower viscosity HPMC (50 - cps) polymer eroded faster than the 4000 - cps polymer, 
consistent with the current work. Thus, the higher molecular weight native dextran 
polymers have higher intrinsic water - holding capacity and the matrices formed 
from such polymers are less prone to erosion than the lower molecular weight 
fractions. 
6.3.6.4 Mechanism of Soluble Principle Active Propranolol Hydrochloride and 
Lobenzarit Disodium from Dextran Tablets 
Soluble drugs are considered to be released by diffusion through the matrix and 
poorly soluble drugs are released by erosion of the matrix. Moreover, it is considered 
that factors affecting swelling and erosion of these polymers may account for 
differences between in vitro dissolution results and subsequent in vivo performance 
when hydrophilic matrix tablets are compared [15] . 
Lobenzarit disodium (LBZ) is a drug conceived for the treatment of rheumatoid 
arthritis. This drug produces an improvement of immunological abnormalities and 

has a regulatory effect upon the antibody - producing system. Propranolol hydrochloride 
(PPL) is a . - adrenergic blocking agent, that is, a competitive inhibitor of the 
effects of catecholamines at . - adrenergic receptor sites. It is widely used in therapeutics 
for its antihypertensive, antiangorous, and antiarrhythmic properties. These 
two drugs are suitable candidates for the design of controlled - release delivery 
systems [25, 29] . According to their solubility in water they can be considered as 
high soluble (PPL) and soluble (LBZ) drugs. 
A comparative study of the dissolution profi le for PPL and LBZ was established 
as the analysis of a similarity factor defi ned as 
f 
n 
R T 
t 
n 
t t 2 
1 
2 
0 5 
50 1 
1 
100 = + . ( ) ... 
... 
. ... 
...
= 
. 
. log 
. 
(4) 
In the above equation f 2 is the similarity factor, n is the number of time points, R t 
is the mean percent drug dissolved of the reference formulation, and T t is the mean 
percent drug dissolved of the tested formulation. 
The evaluation of similarity is based on the following conditions: 
• A minimum of three time points 
• Twelve individual values for every time point 
• Not more than one mean value of > 85% dissolved 
• Standard deviation of the mean that is less than 10% from the second to last 
time point 
An f 2 value between 50 and 100 suggests that two dissolution profi les are similar 
[30] . In this study experimental data corresponding to 30, 60, 90, 120, 180, 240, 300, 
360, 420, and 480 min were considered. 
Figure 20 shows dissolution profi les for tablets of PPL or LBZ from the native 
dextran DTB110 - 1 - 2 matrix system, respectively 1 : 1 (w/w). The value for relative 
standard deviation (CV) was less than 5% for all points measured ( n = 12). 
The Higuchi and Hixson Crowell model as well as the nonlinear regression of 
Peppas and Peppas - Sahlin were employed to study the release data. Higuchi ’ s slope 
FIGURE 20 Disolution profi le for PPL and LBZ dextran tablets (direct compression): 
paddle 100 rpm, 37 ° C, deionized water. 
0 60 120 180 240 300 360 420 480 
0 
20 
40 
60 
80 
100 
LBZ 
PPL 
Amount released (%) 
Time (min) 
TABLET DESIGN WITH NATURAL PRODUCTS 1007

1008 TABLET DESIGN 
(3.179 and 4.500% min . 1/2 for LBZ and PPL, respectively), Korsmeyer ’ s rate constant 
(1.195% min . 0.697 and 4.125% min . 0.540 for LBZ and PPL, respectively), the low 
relaxational constant K r (0.101% min . 0.898 for LBL and . 0.040% min . 0.898 for PPL), 
compared with K d values (2.941 and 6.518% min . 0.449 , respectively) of Peppas - Sahlin 
indicated the diffusional mechanism as predominant for soluble drugs since native 
dextran tablets. The infl uence of solubility of the drug can be observed for the 
hydrophilic matrix (release of PPL is faster than LBZ in correspondence with its 
solubility in water). The value of the diffusional exponent, 0.697 (by the Korsmeyer 
equation), for the less soluble drug corresponds to the increment of the infl uence of 
the erosion mechanism, in agreement with other authors. This can be also observed 
in the Peppas y Salhin equation where the negative value obtained for K r for the 
dissolution profi le of PPL from dextran tablets should be interpreted in terms of a 
relaxation mechanism, which is insignifi cant compared to the diffusion process. 
The dissolution profi les for LBZ:B110 - 1 - 2 and PPL:B110 - 1 - 2 tablets were also 
compared using similarity factor f 2 . A value obtained for f 2 that is below 50 (37.48) 
indicates the infl uence of drug solubility in the dissolution profi le. Furthermore, 
other parameters, such as dextran – drug ratio, particle size of polymer and drug, 
and infl uence of pH, have to be studied to obtain an optimum and robust 
formulation. 
The mechanisms of drug release from dextran matrix occur in the early stage by 
polymer swelling, and the tablet thickness increases. Soon thereafter, polymer (and 
drug) dissolution starts occurring. The polymer dissolves because of chain disentanglement. 
Thus, there is a slow diminution of the thickness because of erosion 
until, fi nally, the tablet disappears (time > 480 min). 
6.3.7 DESIGN TOOLS OF TABLET FORMULATION 
Nowadays, most experimentation on tablet formulation development is still performed 
by changing the levels of each variable (factor) at a time, in an unsystematic 
way, keeping all other variables constant in order to study the effects of that specifi c 
variable on the selected response or to fi nd the optimal conditions of a complete 
system. This methodology (trial and error) is based on a large number of experiments 
and often relies merely on the analyst ’ s experience [31] . 
Statistical experimental design, also called design of experiments (DoE), is a 
well - established concept for planning and execution of informative experiments. 
DoE can be used in many applications. An important type of DoE application refers 
to the preparation and modifi cation of mixtures. It involves the use of “ mixture 
designs ” for changing mixture composition and exploring how such changes will 
affect the properties of the mixture [32] . 
In the DoE approach, fi rst process variables are “ screened ” to determine which 
are important to the outcome (excipient type, percentage, mixture time, etc.). The 
next step is optimization , when the best settings for the important variables are 
determined. In particular, response surface methodologies have been successfully 
applied in both drug discovery and development [33] . Advances in supporting software, 
automated synthesis instrumentation, and high - throughput analytical techniques 
have led to the broader adoption of this approach in pharmaceutical discovery 
and chemical development laboratories [34] . 

The benefi ts of using experimental design together with software to facilitate the 
formulation of a tablet for specifi c purposes, from screening to robustness testing, 
are well known. This technique has some advantages compared to the trial - and - 
error method. By applying a multivariate design for the screening experiments, 
many excipients are evaluated using comparatively few experiments. 
The formulation work is generally based on designed experiments. Most of the 
experiments are fractional or full - factorial designs and are generated and evaluated 
in some cases with the center point replicated. The robustness of the formulation 
and batch - to - batch variation of the excipients and the active pharmaceutical ingredient 
can be evaluated with experimental designs on different occasions. Experimental 
design and optimization of the formulation can be performed with the use 
of software. Some of them have been useful in tablet design. MODDE (version 4.0 
and 5.0, Umetri, Ume a , Sweden), iTAB [35] , and TabletCAD are some 
examples. 
6.3.7.1 MODDE 4.0 
Tablet design for controlled - release propranolol hydrochloride was performed with 
the use of MODDE software [25] . A central composite design (one of the most 
used designs in pharmaceuticals) was applied to the optimization. This experimental 
design required 17 experiments (2 k + 2 k + 3, where k is the number of variables) 
including three center points. Three variables and fi ve responses (according to USP 
25 tolerances for the dissolution profi le for propranolol hydrochloride extended - 
release capsule) were involved in the experimental design. The variables and their 
ranges studied are summarized in Table 13 . The high and low values of each variable 
were defi ned based on preliminary experiments. The critical responses were 
t 100% and t 30% corresponding to the time when 100 and 30% of drug contained in the 
tablets is delivered to the dissolution medium because this system was developed 
to release drug in 24 h ( t 100% . 24 h) and to prevent an overdose for the fi rst minutes 
( t 30% > 1.5 h). The other responses were in the amount of PPL dissolved at 4, 8, and 
14 h. 
Table 14 shows results obtained for every formula development according to 
MODDE 4.0 software. The collected experimental data were fi tted by a multilinear 
regression (MLR) model with which several responses can be dealt with simultaneously 
to provide an overview of how all the factors affect all the responses. The 
responses of the model, R 2 and Q 2 values, were over 0.99 and 0.93 for t 100% and 0.98 
and 0.89 for t 30% , respectively, implying that the data fi tted well with the model. 
Here, R 2 is the fraction of the variation of the response that can be modeled and 
Q 2 is the fraction of the variation of the response that can be predicted by the model. 
The relationship between a response y and the variables x i , x j , . . . can be described 
by the polynomial: 
TABLE 13 Levels of Formulation Variables (Central Composite Design) 
Parameter Low Value ( . 1) Central Value (0) High Value ( + 1) 
Ratio DT – HPMC (w/w) 1 : 1 4 : 1 7 : 1 
Cetyl alcohol (% w/w) 10 15 20 
Ratio excipients – PPL (% w/w) 30 50 70 
DESIGN TOOLS OF TABLET FORMULATION 1009

1010 TABLET DESIGN 
TABLE 14 Matrix of Central Composite Design and Results 
Run Order CEx – PPL DT – HPMC ce t 100% * t 30% * t 2 * t 3 * t 4 * 
10 30 1 : 1 10 10 0.5 a 71 a 90 a — a 
7 70 1 : 1 10 15 2.1 b 44 b 73 b 97 a 
1 30 7 : 1 10 11 0.5 a 63 a 82 a — a 
11 70 7 : 1 10 14 2.2 b 40 b 76 b 99 a 
9 30 1 : 1 20 14 1.3 a 49 b 70 b 99 a 
8 70 1 : 1 20 16 2.2 b 46 b 62 b 96 a 
5 30 7 : 1 20 14 1.6 b 49 b 70 b 99 a 
15 70 7 : 1 20 16 2.4 b 43 b 60 b 96 a 
3 30 4 : 1 15 20 1.2 a 54 b 79 b 94 b 
16 70 4 : 1 15 26 2.6 b 39 b 56 b 72 b 
17 50 1 : 1 15 15 1.6 b 45 b 63 b 98 a 
4 50 7 : 1 15 16 1.7 b 43 b 65 b 96 a 
2 50 4 : 1 10 21 1.4 a 52 b 71 b 93 b 
12 50 4 : 1 20 24 2.2 b 45 b 63 b 82 b 
6 50 4 : 1 15 24 2.1 b 48 b 65 b 85 b 
14 50 4 : 1 15 23.5 2.2 b 49 b 66 b 86 b 
13 50 4 : 1 15 24 2.1 b 48 b 66 b 86 b 
Note: cEx – PPL, ratio of excipients to propranolol; DT – HPMC, ratio of native dextran to hydroxypropylmethylcellulose; 
ce, percentage of cetyl alcohol (w/w) in the tablets. Time t 100% is time (hours) when 
100% of PPL is dissolved in dissolution medium and t 30% is time (hours) when 30% of PPL is dissolved 
in dissolution medium; t 2 is percent of PPL dissolved at 4 h, t 3 percent of PPL dissolved at 8 h, and t 4 
percent of PPL dissolved at 14 h. Values presented are the average of eighteen replicates for each batch. 
a Inside the USP range. b Outside USP range. 
y x x x x x x E i i j j ij i j ii i jj j = + + + + + + . . . + . . . . . . 0 
2 2 
where . j are coeffi cients to be determined and E is the overall experimental error. 
Figure 21 presents the dissolution profi les of all 17 trials generated from the central 
composite design. 
The response surface plots formed by plotting the values for t 30% and t 100% as a 
function of the most important variables are shown in Figure 22 , where the optimum 
condition obtained by the model can be seen. The optimum dextran (DT) – HPMC 
ratio of 4 : 1 (w/w) gave t 100% equal to 24 h. 
With tablet formulations composed of a matrix excipient and PPL at a ratio 
ranging from 40 : 60 to 70 : 30 (w/w), the values for t 100% were satisfactory (around 
24 h). However, the respective values for t 30% increased according to the ratio of 
matrix excipient and PPL ranging from 40 : 60 to 70 : 30 (w/w), showing that early 
drug release was demanded and the initial dose required for pharmacological effect 
could not be suffi cient. 
Sustained - release matrix tablets with good properties were obtained with a 
dextran – HPMC ratio of 4 : 1 (w/w), with a matrix excipient – PPL ratio of 60 : 40 (w/w), 
and with a cetyl alcohol amount of 15% (w/w). The hydrophilic polymers – PPL ratio 
of 60 : 40 (w/w) is more robust for any manufactured variability than 50 : 50 (w/w), 
because the central point of the design is near the lowest desired area (Figure 22 ). 
Under the optimal conditions, the mean value of hardness was 106 ± 3 N and the 
friability was less than 1% (0.2%). 

FIGURE 21 Dissolution profi le generated from central composite design. 
0 2 4 6 8 10 12 14 16 18 20 22 24 26 28 
0 
10 
20 
30 
40 
50 
60 
70 
80 
90 
100 
110 
Exp.10 
Exp.7 
Exp.1 
Exp.11 
Exp.9 
Exp.8 
Exp.5 
Exp.15 
Exp.3 
Exp.16 
Exp.17 
Exp.4 
Exp.2 
Exp.12 
Exp.6 
Exp.14 
Exp.13 
Amount released PPL (%) 
Time (h) 
FIGURE 22 Response surface plots formed by plotting values for t 30% and t 100% . 
30 
40 
50 
60 
70 
10 
12 
14 
1 
6 
182 
0 
1.0 
1.5 
2.0 
2.5 
Ratio DT:HPMC (4:1 w/w) 
t 
30% 
ce 
cE 
xc:P 
P 
L 
30 
40 
50 
60 
70 
1 
2 
3 
4 
5 
6 
7 
12 
14 
16 
18 
20 
22 
24 
Cetyl alcohol 15% w/w 
t 
100% 
DT:HPMC 
cEx 
c:P 
PL 
DESIGN TOOLS OF TABLET FORMULATION 1011

1012 TABLET DESIGN 
Cetyl alcohol (ce in Figure 23 ) has a signifi cant positive effect on both responses 
in the range studied. This may be because the hydrophobic polymer prevents the 
fast release of PPL for the fi rst few hours with an increase in the diffusional pathlength 
of the drug because the swelling of hydrophilic polymers (DT and HPMC) 
retards the rate of release. Interaction of cetyl alcohol and hydrophilic polymers – 
PPL was observed for t 30% . If a prolonged release rate is desired during this period, 
the ratio of hydrophilic polymers to cetyl alcohol can be increased, resulting in a 
decreased interspace volume after erosion of cetyl alcohol. In contrast to other 
products, such as lactose [15] , cetyl alcohol as hydrophobic polymer can be increased. 
The viscosity and texture of the gel layer and some modifi cations in polymer – 
polymer and polymer – solvent interaction are present. 
6.3.7.2 i TAB 
iTAB is I Holland ’ s new tablet design aid that calculates basic tablet parameters 
and stress analysis for “ rounds ” and “ shapes ” in three easy steps. iTAB is Windows 
FIGURE 23 Regression coeffi cients. 
cEx 
DT 
ce 
cEx*cEx 
DT*DT 
ce*ce 
cEx*DT 
cEx*ce 
DT*ce 
-0.4 
-0.2 
0.0 
0.2 
0.4 
0.6 
Variables and interactions, t30% 
Regression coefficients 
cEx 
DT 
ce 
cEx*cEx 
DT*DT 
ce*ce 
cEx*DT 
cEx*ce 
DT*ce 
-8 
-6 
-4 
-2
0
2 
Variables and interactions, t100% 
Regression coefficients

based, saving users from any extra set - up costs and making software upgrades 
easy. 
Users are not required to possess any technical expertise or have any formal 
training, as iTAB is very user friendly and intuitive throughout. Using the drag - and - 
click feature, iTAB results in 2D drawings ready for production and automatically 
generated design reports: 
Step 1 involves the selection of a specifi c tablet profi le and modifi cation of key 
parameters such as diameter, mass, volume, and surface area in real time. The 
iTAB safety zone ensures that unfeasible designs cannot be output from the 
system, meaning that only quality tablets can be produced. 
Step 2 runs a real - time fi nite - element analysis (FEA) simulation against the 
design to give an indicative punch tip maximum force calculation. 
Step 3 automatically produces a report summarizing key design data, for example, 
cup depth and surface area, that can be emailed directly to the I Holland team 
for further analysis, design work, and tooling production. 
iTAB allows non – computer - aided design (CAD) and non - FEA users to simply and 
quickly design nonembossed tablets and obtain detailed information with the click 
of a button. 
Core benefi ts include the avoidance of tablet manufacturing problems such as 
punch tip breakage from the outset. Immediate and accurate punch tip load calculations 
and 3D dynamic rotation of punch tip stress allow for instant decision making 
on design issues. iTAB is also unique in that it incorporates tooling and tablet 
design. 
6.3.7.3 Percolation Theory 
Leuenberger et al. introduced percolation theory in the pharmaceutical fi eld in 1987 
to explain the mechanical properties of compacts and the mechanisms of the formation 
of a tablet [36, 37] . Knowledge of the percolation thresholds of a system results 
in a clear improvement of the design of controlled - release dosage forms such as 
inert or hydrophilic matrices. 
Percolation theory is a statistical theory that studies disordered or chaotic systems 
where the components are randomly distributed in a lattice. A cluster is defi ned as 
a group of neighboring occupied sites in the lattice, being considered an infi nite or 
percolating cluster when it extends from one side to the rest of the sides of the 
lattice, that is, percolates the whole system [38] . 
Thus, a tablet is regarded simply as a heterogeneous binary system formed by 
the active principle and an excipient. As a function of their relative volume ratio, 
one or both components constitute a percolating cluster formed by particles of the 
same component that contact each other from one side to the other sides of the 
tablet. 
In a binary pharmaceutical tablet (cylindrical lattice), the sites can be occupied 
by the component drug or excipient. The percolation threshold of the drug indicates 
at which concentration this substance dominates the drug/excipient system. The 
concept is very similar to the point where a component passes from being the inner 
DESIGN TOOLS OF TABLET FORMULATION 1013

1014 TABLET DESIGN 
to being the outer phase of an emulsion. It is not surprising that the component 
becoming the “ outer phase ” or percolating phase will have more infl uence on the 
properties of the system. 
Furthermore, the concentration point at which a component is starting to percolate 
the system is usually related to a change in the properties of the system, which 
will now be more affected by this component. This is known as a critical point. Close 
to the critical point important changes can take place, for example, changes in the 
release mechanism of the active agent and modifi cation of the tablet structure 
(e.g., monolith versus a desegregating device). 
An important difference between particulate solids and emulsions is that in the 
solids two components can percolate the system at the same time, that is, two components 
can act as the outer phase simultaneously. In this case the system is known 
as a bicoherent system. 
Study of Ternary Tablets Percolation theory has been developed for binary 
systems, however, drug delivery systems usually contain more than two components. 
The existence and behavior of the percolation thresholds in ternary pharmaceutical 
dosage forms have been studied [39] employing mixtures of three substances with 
very different hydrophilicity and aqueous solubility (Polyvinylpyrrolidone (PVP) 
cross - linked, Eudragit RS - PM, and potassium chloride). 
After evaluation of the technological parameters and in vitro release behavior 
of the tablets, no sharp percolation thresholds were found in these ternary systems 
for the employed components separately. Nevertheless, a combined percolation 
threshold of the hydrophilic components was found, demonstrating that a multicomponent 
system can be reduced to a binary one using a discriminating property 
[39] . 
Matrix Systems with Different Particle Sizes Another disadvantage in the application 
of percolation theory to the rationalization of the pharmaceutical design was 
the prerequisite of an underlying regular lattice. Usually, drug delivery systems 
contain substances with different particle sizes. Therefore, the particles cannot be 
considered as each occupying one lattice site. 
This problem can be initially overcome using a volume ratio instead of a lattice 
site ratio, expressing the percolation thresholds as critical volume fractions [36, 
40 – 42] . Nevertheless, the infl uence of the particle size of the components on the 
percolation threshold cannot be explained using a volume fraction; that is, from this 
point of view, tablets with the same excipient volume are equivalent independent 
of their particle size. A fi rst qualitative study of the infl uence of particle size on the 
percolation threshold [43] demonstrated that this is in clear disagreement with 
experimental data. 
According to percolation theory, the effect of a reduction in the drug particle size 
should be similar to an increase in the excipient particle size in a binary system: It 
may be expected that the relative particle size of the component, but not its absolute 
particle size, will determine the properties of the system. 
A quantitative study of the infl uence of particle size on the percolation threshold 
employing inert matrix tablets prepared with KCl and Eudragit RS - PM as matrix - 
forming material [44 – 46] showed that experimental data are in agreement with this 
hypothesis. 

As Figure 24 shows, a linear relationship was found in this study [44] between 
the mean drug particle size and the corresponding drug percolation threshold 
(Figure 24 , line A ). Furthermore, the excipient particle size exerts a contrary effect 
than the drug particle size (Figure 24 , line B ); that is, the larger the excipient particle 
size, the lower the drug percolation threshold [46] . 
In addition, when the obtained drug percolation thresholds were plotted as a 
function of the drug – excipient particle size ratio of the matrices (see Figure 25 ), a 
linear relationship was found between the drug percolation threshold and the relative 
drug particle size [46] . These results are in agreement with the above exposed 
theoretical model based on percolation theory. 
One of the advantages of the proposed model versus classical theories is its ability 
to explain the changes in the release behavior of the matrices by means of a change 
in the critical points of the system (related to the drug and excipient percolation 
thresholds), which can be experimentally calculated, providing a scientifi c basis for 
the optimization of these dosage forms. 
FIGURE 24 Drug percolation threshold (mean ± SE) as function of mean particle size of 
drug (line A ) and excipient (line B ) employed. 
Percolation threshold (.c) 
A
B 
Mean size (.m) 
0.6 
0.4 
0.2
0
0 50 100 150 200 250 300 350 400 
FIGURE 25 Drug percolation threshold (mean ± SE) as function of drug – excipient particle 
size ratio employed. 
Percolation threshold (.c) 
Drug–excipient particle size ratio 
0.6 
0.4 
0.2
0
0 0.5 1 1.5 2 2.5 3 3.5 
DESIGN TOOLS OF TABLET FORMULATION 1015

1016 TABLET DESIGN 
Mechanical Properties The percolation approach was also employed to model the 
tensile strength of tablets [47, 48] . A critical tablet density was here understood as 
a minimal solid fraction needed to build a network of relevant contact points spanning 
the entire tablet. A rising tablet density led to a power law increase of the 
tensile strength showing an universal exponent T f = 2.7. 
It was shown that a power law based on percolation theory was suitable to fi t 
the obtained tensile strength data of the binary matrix tablets studied. The best 
fi tting was observed for a model where an initial tensile strength . 0 was supposed 
[49] : 
. . . . t c = . + k( ). 2 7 
0 
The observed critical relative densities are understood as threshold values for the 
tensile strengths of the tablets. One practical consequence of these works is to avoid 
the manufacture of matrix tablets close to these critical densities. The formulation 
may not be robust in this critical range from the viewpoint of mechanical tablet 
stability. 
6.3.7.4 Artifi cial Neural Networks 
Artifi cial neural networks (ANNs) are computer programs designed to model the 
relationships between independent and dependent variables. They are based on the 
attempt to model the neural networks of the brain [50] . Functions are performed 
collectively and in parallel by the units, rather than there being a clear delineation 
of subtasks to which various units are assigned. 
This methodology represents an alternative modeling technique that has been 
applied to pharmaceutical technology data sets, including tableting parameters [51] . 
The main advantage with respect to classical statistical techniques, such as response 
surface methodology, is that ANNs do not require the prior assumption of the nature 
of the relationships between input and output parameters, nor do they require the 
raw data to be transformed prior to model generation [51] . ANNs are capable of 
modeling complex, nonlinear relationships directly from the raw data. 
The functional unit of ANNs is the perceptron. This is a basic unit able to generate 
a response as a funtion of a number of inputs received from others perceptrons. 
For example, the response value can be obtained as follows: 
Y 
W I WI W 
W I WI W 
= 
+ + > 
+ + . {1 0 
0 0 
0 0 1 1 
0 0 1 1 
if 
if 
b
b 
where I x is the input received from perceptron x and W x the weight assigned to this 
input by the perceptron. The weights can be changed to adapt the answer to the 
desired one using a learning algorithm. 
Usually complex structures with more than 15 layers are employed, called the 
multilayer perceptron (MLP). Some of the commercial programs which have been 
used to fi t tableting parameters are INForm (Intelligensys, Billingham Teesside), 
CAD/Chem (AI Ware, Cleveland, OH), which is no longer commercially available, 
and the Neural Network Toolbox of MATLAB (MathWorks, Natick, MA). 

6.3.8 COATING SYSTEMS 
Coating processes have come to play an important role for the protection of substances 
prior to application or for their sustained release. Coatings on cores usually 
consist of a mixture of substances. The matrix formers are responsible for the stability 
of the coating structure, and they also determine the coating process. Depending 
on the type of matrix former on binder used, three coating categories can be 
distinguished: 
Coatings with Sucrose and Other Sugars Permit application of copious amounts 
of mass to the core and are widely used in the manufacture of pharmaceuticals 
and confectionery. 
Hot Melts Add a considerable amount of mass and are applied hot and solidify 
while cooling on the core. They are mainly used for confectionery. The most 
important raw materials are fats, mostly cocoa fat, polyethylene glycol (PEG) 
[52] , and the sugar – alcohol mixture xylitol – sorbitol. 
Film Coatings Require less material, forming thin membranes which largely 
follow the contours of the substrate, for example, scores and engravings. The 
partly pH - dependent solubility and selective permeability of coatings are 
affected by the fi lm formers. Such fi lms are sometimes also used as intermediate 
layers in sugar coatings. 
Whether or not a core is suitable for coating depends on its hardness, shape, 
surface, size, heat sensitivity, and tendency to interact with the coating material. 
Moreover, since sugar and fi lm coating processes involve very different techniques, 
they place different demands on the cores to be used [53] . Tablets used as cores 
must be biconvex in shape to prevent them from sticking together like coins in a 
roll. 
The ideal tablets for sugar coating will have a pronounced convex curvature and 
a narrow band. The consumption of coating material depends very much on the 
tablet shape and increases sharply if the tablets are not round. Film coating supplies 
coated products in which the core surface (e.g., with notches, engravings, and 
defects) is faithfully reproduced.The fi lms tend to chip at sharp edges or are particularly 
thin in these areas. For this reason slightly curved tablet cores are preferred 
for fi lm coating [54] . 
Film coating of pharmaceuticals is a common manufacturing stage for the following 
reasons: (i) to provide physical and chemical protection for the drug, (ii) to 
mask the taste or color of the drug, or (iii) to control the release rate or site of the 
drug from the tablet. When a coating composition is applied to a batch of tablets 
or granules (or to a batch of liquid drops or even gas bubbles), the core surfaces 
become covered with a polymeric fi lm that is formed as the surfaces dries. The major 
component in a coating formulation is a fi lm - forming agent which ideally is a high - 
molecular - weight polymer that is soluble in the proper solvent (today, most preferably 
in aqueous - based media). The polymer forms a gel and produces an elastic, 
cohesive, and adhesive fi lm coating. 
In the pharmaceutical industry, organic - solvent - based fi lm coatings have been 
used for over 40 years. In the 1990s, however, interest and demands in the use of 
aqueous - based fi lm coating systems rapidly increased owing to the well - documented 
COATING SYSTEMS 1017

1018 TABLET DESIGN 
drawbacks (unsafe, toxic, pollutive, and uneconomic) associated with organic - 
solvent - based coating systems. Consequently, and for the reasons mentioned above, 
much effort has been focused on the research and development of new aqueous - 
based fi lm coating formulations. Nowadays, aqueous - soluble/dispersable polymers 
available on the market consist primarily of either cellulose polymers, PEGs, or 
acrylate copolymers. There are, however, some material - related limitations in using 
these polymers in aqueous - based fi lm coatings. Consequently, application of new 
fi lm formers such as chitosan, native starches, and special types of proteins for 
pharmaceuticals and foodstuffs has been increasingly studied. 
The choice between sugars of fi lm coatings depends not only on the desired coatings 
quality but also on the technical requirements, that is, on the economy of the 
process. The fi nancial outlay for a selected technology often commits the manufacturer 
to this technology for a prolonged period of time. Film and sugar differ 
substantially in thickness and therefore also in the necessary mass of coating 
material. 
The most important coating raw material is the fi lm - forming polymer, which must 
be able to produce a coherent fi lm on the substrate under the given process conditions 
[55, 56] . Second in importance is the solvent or dispersing system in which the 
polymer is applied to the surface and introduced to form a fi lm. Other frequently 
used raw materials are plasticizers, glidants, fi llers, and colorants. All these substances 
act together and infl uence the properties of the fi lm. 
The fi rst decision to be made when developing a formulation concerns the desired 
function of the fi lm. Depending on the requisite dissolution performance in physiological 
media, the fi lm former to be tested — or several of them — is then selected 
from the available polymers. The economy of the coating process and the quality of 
the coated product depend on the correct calculation of the required amount of 
coating material. Empirical adjustment of new developments to existing products is 
therefore not recommended. The coating quantity needed for fi lm coatings is directly 
related to the surface area of the core. Table 15 shows a simplifi ed calculation of the 
surface area of tablets. It is based on the surface area of a cylinder circumscribing 
these shapes. All these data are taken from the literature. For sugar coating processes, 
the shape of the cores and end products have to be studied stereometrically. 
The sugar coating process balances irregularities, since the coating buildup does not 
follow the structure of the surface. Areas which require a high degree of rounding, 
such as high bands and very fl at curvatures, attract more coating mass. Where the 
band height varies owing to manufacturing technique, this results in weight differ- 
TABLE 15 Calculation of Surface Area ( mm 2 ) for Different Types of Cores 
Height (mm) 
Diameter (mm) 
3 4 5 6 7 8 9 10 12 14 
2 33 56 70 95 120 150 
3 42 62 85 115 145 175 210 250 340 
4 75 100 130 165 200 240 280 380 485 
5 185 225 270 315 415 530 
6 300 345 450 570 
7 505 615 
8 660 

ences between the individual sugar - coated tablets. This is one of the reasons why 
sugar - coated products always show a much wider weight distribution than fi lm - 
coated tablets. 
Several authors have made proposals for the most convenient size of tablet for 
sugar coating. A further rule has been established according to which the convex 
radius should be between 0.7 and 0.75 times the tablet diameter and the band height 
between 0.07 and 0.12 times that diameter. However, for calculation of the band 
height only a factor of 0.12 is advisable, since, otherwise, the minimum value of 
1 mm will be fallen short of by far [53, 57 – 60] . For fi lm coating the recommended 
convex radius is 1.5 times the tablet diameter (less curved tablets are preferred). 
Numerous formulation are available for all fi lm - forming polymers offered in the 
market. Thus, it is possible to dispense with many of the preliminary tests and 
concentrate development work on the special problems of the formulation in question. 
In this chapter, we will focus on SEPIFILM and Kollidon VA 64. Table 16 
presents proven basic formulations reported on in the literature [61] and polymers 
used for the most important commercially available fi lm formers, which also can be 
used as a basis for further tablet coating. 
The major operational coating process parameters related to fi lm coating are able 
to be measured and monitored continuously in some pan - type coaters. The inlet 
airfl ow rates infl uence the coating process and the subsequent quality of the coated 
tablets. Increasing the inlet airfl ow rate accelerated the drying of the tablet surface. 
At high inlet airfl ow rate, obvious fi lm coating defects, that is, unacceptable surface 
roughness of the coated tablets, are observed and the loss of coating material 
increased. 
Today advantages of aqueous fi lm coating are well recognized and fi lm coating 
technology is much developed to successfully perform these types of coatings. 
Process automation and monitoring of critical process parameters can be utilized 
to increase the overall process effi ciency and predictability and to improve the 
homogeneity and reproducibility of the tablet batches. This will ensure high quality 
and safety of the fi nal coated products, which are mandatory requirements of tablet 
manufacturing. 
6.3.8.1 Subcoating of Tablet Cores as a Barrier to Water 
As tablets are nowadays coated mostly with aqueous solutions or dispersions, it has 
become increasingly necessary to provide the tablet cores with a barrier layer prior 
to sugar or fi lm coating. This is mainly to protect water - sensitive drugs against 
hydrolysis and chemical interactions, for example, between different vitamins, and 
to prevent the swelling of high - performance tablet desintegrants that are very sensitive 
even to small quantities of water. It can be especially useful when controlled - 
release systems with hydrophilic polymer are studied and the water contained can 
change the dissolution profi le. Kollidon VA 64 also can be used to improve the 
adhesion of subsequent coatings by hydrophilization of the surface. 
6.3.8.2 Kollidon VA 64 
We are studying the ability of Kollidon VA 64 as a subcoating in a combined hydrophilic 
(dextran – HPMC) – hydrophobic (cetyl alcohol) matrix core prior to sugar 
COATING SYSTEMS 1019

1020 TABLET DESIGN 
TABLE 16 Basic Formulations for Film - Coated Tablets 
Formulation with Hydroxypropyl Methylcellulose (HPMC) 
A B 
Oprady 73.0% — 
Pharmacoat — 80.0% 
PEG 6000 — 8.0% 
Talc 20.0% 5.0% 
Pigments included, TiO 2 7.0% 7.0% 
Solid content 20.0% 12.0% 
Coating quantity, mg/cm 2 1 – 5 1 – 5 
Formulation with Methacrylic Acid Copolymers 
A B C 
Eudragit L100 5.0% — — 
Eudragit S100 — 7.5% — 
Eudragit L30D - 55 — — 16.5% 
PEG 6000 0.7% 1.0% 1.6% 
Talc 6.0% 2.0% 4.0% 
Pigments included, TiO 2 3.3% — — 
Isopropyl alcohol 41.0% 86.5% — 
Acetone 41.0% — — 
Water 3.0% 3.0% 77.9% 
Solid content 14.0% 10.5% 22.1% 
Coating quantity, mg/cm 2 2 – 4 2 – 4 3 – 5 
Formulation with Hydroxypropyl Cellulose Acetate Succinate (HPMCAS) 
Aqoat AS - MF 10.0% 
Triethyl citrate 2.8% 
Talc 3.0% 
Sorbitan sesquioleate 0.0025% 
Water 84.2% 
Formulation with Ethylcellulose (EC) 
A B 
Ethylcellulose 5.0% — 
Aquacoat (30% solids) — 30.0% 
PEG 6000 — 2.0% 
Glycerol triacetate 1.0% — 
Ethanol 94.0% — 
Water — 68.0% 
Formulation with Carboxymethyl Ethylcellulose (CMEC) 
Duodcell 8.0% 
Trisodium citrate 0.70% 
Tween 80 0.04% 
Glycerol monocaprylate 2.40% 
Water 88.86% 
Coating quantity 7 mg/cm 2 
Formulation with Polyvinyl Acetate Phthalate (PVAP) 
PVAP 11.0% 
PEG 400 1.0% 
Ethanol 66.0% 
Water 22.0% 

coating. The copovidone (i.e., Kollidon VA 64) not only increases the mechanical 
properties of the tablet (less friability) but also prevents the amount of water 
absorbed from the air in tropical and subtropical stability conditions (25 and 75% 
relative humidity). 
Figure 26 shows a comparative study of uncoated tablet and sugar - coated tablet 
after a barrier to water with copovidone 0.5 mg/cm 2 of the warm tablet cores using 
a 10% solution in ethanol. During two years the coating tablets (with initial humidity 
2.5%) remained stable and dissolution profi les were similar to the initial time 
with similarity f 2 = 82 observed and friability decreased from 0.25% (uncoated 
cores) to 0.02% (coated tablets). 
Similar results are reported in the literature when Kollidon VA 64 is compared 
to povidone (Kollidon K 25, K 30, and 90 F). Copovidone absorbs about three times 
less water than the other soluble Kollidon K 25, 30, and 90F after seven days at 
25 ° C up to 80% relative humidity [62] . Kollidon VA 64 is manufactured by free - 
radical polymetization of 6 parts of vinylpyrrolidone and 4 parts of vinyl acetate in 
2 - propanol. A water - soluble copolymer with a chain structure is obtained. In contrast 
to the soluble grades of Kollidon, the number 64 is not a K value but the mass 
ratio of the two monomers, vinylpyrrolidone and vinyl acetate. The K value of 
Kollidon VA 64 is of the same order of magnitude as that of Kollidon 30. Synonyms 
for Kollidon VA 64 are copovidone, copovidonum, copolyvidone, copovidon, and 
PVP - VAc - copolymer [Eur. Ph., Japanese Pharmaceutical Excipients, and USP 
National Formulary (NF)] [63] . 
Copovidone forms soluble fi lms independently of the pH value, regardless of 
whether it is processed as a solution in water or in organic solvents. He offers better 
plasticity and elasticity than other povidones. On the other hand, fi lms are also less 
tacky. Kollidon VA 64 usually absorbs water, and it is seldom used as the sole fi lm - 
forming agent in a formulation. Normally it is better to combine it with less hygroscopic 
substances such as cellulose derivates [54] , shellac, polyvinyl alcohol (PVA), 
FIGURE 26 Water absorption of precoating and uncoated cores of combinated dextran – 
HPMC matrix tables. 
0 100 200 300 400 500 600 700 800 
0
1
2
3
4
5
6
7
8 
Water absorption, % 
Days 
Precoating cores with Kollidon VA 64 
Uncoated cores 
COATING SYSTEMS 1021

1022 TABLET DESIGN 
PEG (e.g., Macrogol 6000), or sucrose. Others plasticizers such as triethyl citrate, 
triacetin, or phtalates are not required. The properties of coatings can be improved 
with combination copovidone – cellulose derivates [64, 65] . Cellulose polymers of 
high viscosity, such as HPMC 2910, are used in fi lm coating. Spray suspension at 
12% HPMC K4M offers values of viscosity above 700 mPa · s and can not be normally 
used because 250 mPa · s is considered the limit for spraying of a coating suspension. 
This value of viscosity can be reduced signifi cantly up to 250 mPa · s if 60% 
HPMC is substituted by Kollidon VA 64 [66] and this leads us to apply this polymer 
concentration and therefore to economize the spraying procedure. 
6.3.8.3 SEPIFILM 
SEPIFILM and SEPISPERSE Dry are ready - to - use, immediate - release, fi lm coating 
compositions designed for pharmaceutical and nutritional supplement applications. 
Based on a unique technology, SEPIFILM/SEPISPERSE Dry are granular forms 
offering the following benefi ts: 
Easy handling: no dust 
Easy mixing: no lumps, no foam 
Homogeneous composition: no segregation 
Most coating compositions are based on hypromellose (HPMC) as fi lm - forming 
polymer and contains microcrystalline cellulose (MCC) (see Table 17 ). 
SEPPIC was the fi rst company to introduce MCC in coating formulations some 
20 years ago. The use of MCC allows higher solid content and enhances the adhesion 
of the fi lm to the tablet core, consequently improving logo defi nition. Different 
formulas are provided by Sepifi lm, such as SEPIFILM LP, SEPIFILM 003 and 752, 
and SEPISPERSE Dry. The last one can be provided with SEPIFILM and Kollicoat 
IR. 
Moisture Protection of SEPIFILM LP Water vapor transmission rates were 
measured on free fi lms, including titanium dioxide (Figure 27 ) . SEPIFILM LP 
shows signifi cantly lower moisture permeability compared to regular or PVA - based 
coating formulations. Removal of titanium dioxide (SEPIFILM LP clear) improves 
moisture resistance. 
Like many other herbal extracts, valerian extract is very hygroscopic. Inclusion 
into tablets raises stability issues. Coatings reduce moisture absorption but often 
lead to tablet explosion or visual deterioration (black specs). Tablets formulated 
with 250 mg valerian extract, spray - dried lactose, and compressible starch were 
TABLE 17 General Formulation for SEPIFILM Coating 
Film 
Film - forming agent Hypromellose (former HPMC) 
Binder Microcrystalline cellulose 
Hydrophobic plasticizer Stearic acid (vegetable origin) 
Colors Pigments, lakes 

FIGURE 27 Water vapor transmission rates of free fi lms. 
0.9 
0.8 
0.7 
0.6 
0.5 
0.4 
0.3 
0.2 
0.1
0 
mg/h·m 
Sepifilm 
LP clear 
Sepifilm 
LP 
HPMC + 
Macrogol 
stearate 
HPMC + 
PEG 400 
PVA 
FIGURE 28 Tablets coated with ( a ) conventional hypromellose and ( b ) Sepifi lm LP 770 
white. 
(a) (b) 
stored at 40 ° C and 90% RH (relative humidity). Pictures were taken after one 
month storage (Figure 28 ). 
Tetrazepam Tetrazepam, a well - known muscular relaxant, undergoes chemical 
degradation when exposed to moisture and oxygen. 3 - Ketotetrazepam is one of the 
main degradation substances. The amount of 3 - ketotetrazepam, measured by HPLC, 
has been monitored under 25 ° C/60 RH and 40 ° C/75% RH aging conditions. 
SEPIFILM LP 770 signifi cantly improves the stability of tetrazepam. Uncoated 
tablets and tablets coated with a PVA - based formulation show a strong increase in 
the amount of degradation substance (Figure 29 ). 
Dissolution Profi le SEPIFILM LP can effi ciently improve the moisture barrier 
on moisture - sensitive active pharmaceutical ingredients (API) or hygroscopic cores. 
The breakthrough in this technology is that SEPIFILM LP does not modify the 
dissolution profi le when compared to conventional coating (Figure 30 ). 
COATING SYSTEMS 1023

1024 TABLET DESIGN 
FIGURE 29 Stability of tetrazepam: uncoated tablets and different tablets coated. Dosage 
of 3 - ketotetrazepam in 50 mg tetrazepam tablets stored at 25 ° C and 60% RH leads to partial 
degradation of tetrazepam at T 0 . Initial amount of 3 - ketotetrazepam is lower in uncoated 
tablets: cores are likely to absorb water during the aqueous coating process. 
0.45% 
0.40% 
0.35% 
0.30% 
0.25% 
0.20% 
0.15% 
0.10% 
0.05% 
0.00% 
0 1 
month 
2 
months 
3 
months 
4 
months 
Uncoated 
PVA based
Sepifilm LP 770 
FIGURE 30 Dissolution profi le of tetrazepam, 3 months storage at 40 ° C and 75% RH. 
100% 
90% 
80% 
70% 
60% 
50% 
40% 
30% 
20% 
10% 
0% 
% Released 
0 5 10 15 20
min 
25 30 35 40 60 
PVA based 
Uncoated 
Sepifilm 
LP 770 
HPMC/ 
PEG based 
SEPIFILM 003 and 752 The association of cellulose with a fi lm coating agent 
was originally patented by SEPPIC. Microcrystalline cellulose is probably one of 
the most extensively used excipients in pharmaceutical and nutritional products. 
Unlike other fi llers, such as lactose, cellulose is inert, vegetable derived, and accepted 
worldwide and its shelf life is unlimited. Figure 31 shows the advantage of cellulose 
microcrystalline in fi lm coating. 
Faster Film Coating Operations Cellulose is insoluble and does not increase viscosity. 
Dispersions with higher solid content can therefore be used and total spraying 
time is signifi cantly decreased. 
The maximum acceptable viscosity of a typical coating dispersion is 500 mPa · s, 
which corresponds to an 11% hypromellose 6 mPa · s dispersion. SEPIFILM can be 
dispersed up to 15% in order to reach the same viscosity (Figure 32 ). 

FIGURE 31 ( a ) Tablet coated with HPMC smooth fi lm, medium discontinuity between fi lm 
and core. ( b ) Tablet coated with SEPIFILM 752 white: clear edge perfectly coated, good 
adhesion of fi lm to core and continuity between fi lm and core. 
(a) (b) 
FIGURE 32 Viscosity for deferments HPMC coating dispersion. 
1400 
1200 
1000 
800 
600 
400 
200
0 
0 5% 10% 15% 20% 
Sepifilm 003 HPMC 6 cPs HPMC 15 cPs 
Viscosity, cPs 
Enhanced Film Adhesion Comparative adhesion values were measured on 3% 
coated placebos using a modifi ed crushing strength tester: Regular fi lm formulation 
exhibited an adhesion value of 4 N. Addition of cellulose signifi cantly improved fi lm 
adhesion as it became impossible to remove fi lm without disrupting tablets or fi lm. 
Breakage occurred for an applied force of 6 N, which can be considered a minimum 
adhesion value (Figure 33 ). 
Ingredient Segregation Avoided Powders such as hypromellose and titanium 
dioxide exhibit dramatically different particle size distribution and density. Such 
particle heterogeneity may result in constituent segregation inducing fi lm imperfections 
or color deviations on tablets. 
FIGURE 33 Tablet surface and edges are smoothly covered whereas no logo bridging is 
observed. 
COATING SYSTEMS 1025

1026 TABLET DESIGN 
As seen in (Figure 34 ), SEPIFILM rules out this weak feature of powdered 
coating agents as all constituents are closely bound together. Ingredient segregation 
is avoided and batch - to - batch consistency is therefore guaranteed. 
The PVA – PEG graft copolymer Kollicoat IR is the new instant - release, aqueous 
coating polymer from BASF. Due to its low viscosity and excellent mechanical 
properties, it permits solid content of up to 30% and leads to fi ne and smooth tablet 
fi lm coatings. 
In 2005, SEPPIC and BASF Pharma Solutions began collaboration in marketing 
tablet fi lm coating systems. From this collaboration, a new range of colored 
SEPIFILM coating systems based on Kollicoat (a registered trademark of BASF 
Aktiengesellschaft) polymers will be developed to meet individual customer needs. 
Kollicoat IR has already been approved in Europe as a fi nished drug in Germany 
(a reference member state in a mutual recognition procedure). Common uses in 
pharmaceuticals depend on regulations of each country and defi nition of uses. 
Actual regulations of a specifi c country and/or application should be checked before 
use. 
For pharmaceuticals, this combination is based on 88% of Kollicoat ® IR White 
and 12% of coloring system SEPISPERSE ™ Dry. 
Recommended Equipment 
A propeller stirrer is standard equipment even though a defl occulating blade is 
very effi cient. 
Deionized or distilled water at room temperature. 
The blade diameter should be 3 times shorter than the tank ’ s width. 
The tank should be 1.5 times higher than wide. The blade should be slightly off 
center. 
The blade should be positioned close to the bottom of the vessel. 
FIGURE 34 Granules of SEPIFILM 1306 green ( . 350). Pigments, lakes, and cellulose fi bers 
are thoroughly coprocessed into homogeneous granules. 

TABLE 18 SEPIFILM LP (with or without SEPISPERSE Dry) 
Manesty XL 
Lab01 (A * ) 
Manesty 
Accelacota 
(B) 
IMA - GS HT/M 
(C) 
Driacoater 
500 (C) 
Solid content LP 770 at 
12% 
LP 014/Dry 
at 12% 
LP 014/Dry at 
11% 
LP 014/Dry 
at 12% 
Batch size, kg 5 100 88.2 3 
Spraying rate 15 – 27 mL/min 200 – 300 g/min 180 – 200 mL/min 7 – 15 g/min 
Inlet air temperature, 
° C 
60 70 – 75 62 55 – 60 
Outlet air 
temperature, ° C 
47 — — 42 
Bed temperature, ° C 43 40 – 45 36 – 40 — 
Atomizing air/pattern 
pressure, bars 
2 3.5 – 5 2.5 3 
Airfl ow, m 3 /h 440 1800 1000 270 
Spraying time, min 
(% weight gain) 
76 (1% w.g.) 60 (2% w.g.) 90 (2.2% w.g.) 70 (3% w.g.) 
Mixing Procedure 
SEPIFILM Product Range 
1. Adjust rotation speed in order to create a vortex. 
2. Quickly pour SEPIFILM into the vortex. 
3. Reduce mixer speed to avoid drawing air into the liquid (risk of foam 
formation). 
4. Increase speed again as viscosity builds up. 
It is not necessary, though recommended, to keep stirring during the coating 
process. 
Kollicoat® White + SEPISPERSE Dry 
1. Adjust rotation speed to create a vortex. 
2. Quickly pour SEPISPERSE Dry into the vortex. 
3. Reduce mixer speed to avoid drawing air into the liquid. Stir for 15 min. 
4. Pour Kollicoat IR White into the colored dispersion. 
5. Stir an additional 5 min. 
Some practical conditions for SEPIFILM and equipment machines are shown in 
Tables 18 – 20 . 
We used SEPIFILM LP in ranitidine core because this is a moisture - sensitive 
drug and can be a challenge to formulators because of its tendency to hydrolyze 
when exposed to humidity and/or high temperatures. 
Cores of 300 mg that contained 50% ranitidine HCl, cellulose microcrystalline 
(PH - 102) 27.25%, pregelatinized starch 22%, Aerosil 0.5%, and magnesium stearate 
COATING SYSTEMS 1027

1028 TABLET DESIGN 
TABLE 19 SEPIFILM Formulations 050 and 752 
Driacoater 500 Glatt Coater 1000 
Solid content of dispersion SEPIFILM 050 at 15% SEPIFILM 752 at 20% 
Batch size, kg 3 80 
Spraying rate, g/min/kg 7 (1 nozzle) 3.1 (3 nozzles) 
Rotation speed, rpm 10 8 
Inlet air temperature, ° C 60 65 
Outlet air temperature, ° C 44 — 
Bed temperature, ° C 39 – 40 38 – 40 
Atomizing air/pattern pressure, bars 3 3.5 
Airfl ow, m 3 /h 330 1800 
Spraying time, min (3% weight gain) 85 55 
TABLE 20 Kollicoat IR White + SEPISPERSE Dry 
Manesty Accelacota 24 in., 
300 - mg Propranolol HCl Tablets 
Manesty Premier 200, 
Placebo Tablets 
Solid content of dispersion IR + Dry at 20% IR + Dry at 20% 
Batch size, kg 7 180 
Spraying rate 5.3 g/min/kg 300 – 375 mL/min 
Inlet air temperature, ° C 60 60 
Outlet air temperature, ° C 38 – 43 45 
Bed temperature, ° C 39 – 41 — 
Atomizing air/pattern 
pressure, bars 
2/1 2.5/2.5 
Airfl ow, m 3 /h 220 2200 
Spraying time, min 
(%weight gain) 
37 (3.4% w.g.) 80 (3% w.g.) 
0.5% were coated to a 3% weight gain similar to case A * (see Table 18 ). Tablet 
weight, diameter, thickness, hardness, and disintegration times were measured after 
coating. The fi lm - coated tablets were packaged in bottles with a desiccant. Stability 
testing was conducted at 40 ° C/75% RH for 12 months. Application of fi lm coating 
(3% wg) resulted in a slight increase in tablet hardness (tablet breaking force 13 – 
14 kp). Tablet disintegration time was not signifi cantly affected by the fi lm coating 
application (around 13 min) and 100% of the drug was released within 25 min 
compared to the USP limit of not less than 80% ( Q ) in 45 min. Figure 35 shows the 
dissolution profi les for the uncoated and coated tablets. 
No signifi cant changes were recorded for coated tablets after 12 months of 
storage for any property measured (Table 21 ). The stability of this formulation is 
partly due to the inclusion of pregelatinized starch in the formulation. Starch 1500 
acts as a moisture scavenger and retains moisture in its complex structure of glucose 
polymer chains. A slight increase in tablet hardness was seen after storage (see 
Figure 36 ). No signifi cant changes were seen in the disintegration time after 
storage. 

FIGURE 35 Dissolution profi le of coated and uncoated ranitidine HCl tablets. 
0 
20 
40 
60 
80 
100 
120
0 10 20 30 40 50 
Time (min) 
Percent released 
Uncoated tablet 
Coated tablet (3% 
WG) 
TABLE 21 Stability Data Summary: Test 
Test USP Limit Initial 1 month 3 months 6 months 12 months 
Breaking force (kp) NMT 1.00% 13.7 14.4 14.2 14.5 14.4 
Friability, % 0.0 0.0 0.0 0.0 0.0 
Dissolution, T85%, 
min 
NLT 85% in 
45 min 
16 16 16 16 16 
Assay, % 90 – 110 102 100 100 99 99 
Impurities, % NMT 2% 0.5 0.8 1.0 1.2 1.7 
Notes: For 40 ° C/75% RH storage conditions. Abbreviations: NMT, no more than; NLT, no less than. 
FIGURE 36 Coated tablet hardness on storage, 40 ° C, 75% RH storage conditions. 
0
2
4
6
8 
10 
12 
14 
16 
1 2 3 4 5 
Tablet breaking force (kp) 
Selected Coating Problems and Practical Solutions 
(a) Defective Coatings Caused by poor quality of the core or inadequate coating 
formulations. 
(b) Chipping Solid content is too high or it is too brittle for want of plasticizer 
and the fi lm does not adhere properly to the substrate surface (too lipophilic 
surface or surface lacking in porosity). 
Solution Revise the formulation for core and coating. 
COATING SYSTEMS 1029

1030 TABLET DESIGN 
(c) Blistering Drying or spraying is performed at high speed, and solvent may 
be retained in the fi lm. They evaporate on postdrying and may then form 
blisters in the fi lm. 
Solutions Lower the inlet air temperature and reduce the spray rate. 
Check if adhesion of the fi lm and core is adequate. 
(d) Cracks in Film or Along Edges Caused by too much internal stress, owing 
to differences in the thermal expansion of fi lm and core or also caused by 
the swelling of the core during the coating operation. 
(e) Embedded Particles Particles broken off from the core are embedded in 
the fi lm during spraying. 
Solutions The core lacks mechanical stability. Check the formulation of 
the core. 
(f) Picking The fi lm surface contains substances that are not molecularly dispersed 
and start to melt at the core bed temperature of the fi lm coating 
process. These substances (e.g., PEG, stearic acid) may interfere with the 
fi lm - forming polymers and produce holes in the fi lm surface [67] . 
Solutions Replace these substances or lower the core bed temperature. 
Decrease the speed of praying. 
(g) Dull Surfaces The fi lm will be dull and totally devoid of gloss if a coating 
process does not produce the requisite smooth surface. This happens if the 
spray droplets start to dry before reaching the cores and are too viscous to 
form a smooth fi lm. 
Solutions (1) Lower the inlet air temperature and reduce the atomizing 
air quantity or pressure. (2) Add substances that enhance fi lm formation, 
for example, plasticizer or extra solvents. 
(h) Twinning The cores permanently stick together. 
Solutions Decrease the excessive spraying. Check the tablet shape and 
bands. If the tablets are plane or almost plane, this continues until many 
of them stick together. 
(i) Bridging the fi lm fails to follow the contours of the tablet over break lines 
or engravings and settles in these without adhering to the substrate. The 
bridging forces in the fi lm exceed the interfacial forces between fi lm and 
core. 
Solutions change the tablet surface or add plasticizer. 
6.3.9 DEVELOPMENT OF PHARMACEUTICAL TABLETS USING 
PERCOLATION THEORY 
In 1991, Bonny and Leuenberger [40] explained the changes in dissolution kinetics 
of a matrix controlled - release system over the whole range of drug loadings on 
the basis of percolation theory. For this purpose, the tablet was considered a disordered 
system whose particles are distributed at random. These authors derived a 
model for the estimation of the drug percolation thresholds from the diffusion 
behavior. 

Knowledge of the percolation thresholds and the related critical points of the 
system allows a rational optimization of the matrix formulation, avoiding the trial - 
and - error method usually employed in the pharmaceutical industry. The ideal formulation 
of an inert matrix, following percolation theory, must be above the drug 
percolation threshold (i.e., the drug plus the initial pores percolate the system). This 
fact guarantees the release of the total drug dose. On the other hand, the matrix 
must also contain an infi nite cluster of excipient (i.e., the excipient must also be 
above its percolation threshold). This percolating cluster of excipient avoids the 
disintegration of the matrix during the release process and controls the drug release 
[43] . 
This kind of system, containing percolating clusters of both drug and excipient, 
is called a bicoherent system. Furthermore, in order to decrease the variability in 
the biopharmaceutical and mechanical behavior of the matrices, due to little change 
in the tablet composition, it is not convenient to formulate the matrices just at the 
percolation threshold. In this way, knowledge of the percolation thresholds of drug 
and excipient supposes an important decrease in the cost of the optimization process 
as well as in the time to market. The percolation thresholds of different pharmaceutical 
powders have already been estimated, including drugs such as morphine 
hydrochloride [68] , naltrexone hydrochloride [69] , dextromethorphan hydrobromide 
[70] , and lobenzarit disodium [71] as well as matrix - forming excipients such 
as hydrogenated castor oil, ethylcellulose, and acrylic polymers. 
6.3.9.1 Case Study: Optimization of Inert Matrix Tablets for Controlled Release 
of Dextromethorphan Hydrobromide 
The objective of this work was to estimate the percolation thresholds of dextromethorphan 
hydrobromide and Eudragit RS - PM which characterize the release 
behavior of these inert matrices in order to rationalize the design of these 
controlled - release systems. 
Dextromethorphan hydrobromide is an antitussive drug with no analgesic or 
addictive action. Its antitussive effect is similar to codeine. The recommended oral 
dose for adults is 10 – 30 mg three to six times a day, not to exceed 120 mg daily. It is 
absorbed rapidly and completely when taken orally with a lag time of 15 – 30 min 
[72] . 
In order to estimate the percolation threshold of dextromethorphan hydrobromide, 
the matrices were studied from different points of view: 
1. Release Profi les and Release Kinetics Figure 37 shows the percentage of drug 
released from the studied matrices. As can be appreciated, a very similar behavior 
was observed for matrices containing up to 50% w/w of drug. This can be attributed 
to the swelling process (approximately 11% v/v) undergone by the matrices during 
the release assay. This process makes the infl uence of the percolation threshold on 
the release profi les less evident. 
Higuchi ’ s kinetic model and Peppas ’ nonlinear regression ( Q = a . + b . t k ) were 
employed to study the release data. The results obtained are shown in Table 22 . As 
can be seen, the exponent k underwent a change (0.4534 – 0.5472) between matrices 
containing 20 and 30% w/w of drug. Even if this is not an important change, it may 
be related to some changes in the matrix structure due to the drug percolation 
PHARMACEUTICAL TABLETS USING PERCOLATION THEORY 1031

1032 TABLET DESIGN 
FIGURE 37 Percentage of drug released vs. time for tablets prepared with different loadings 
of dextromethorphan hydrobromide. 
100 
90 
80 
70 
60 
50 
40 
30 
20 
10
0 
% Dextromethorphan hydrobromide 
released 
0 120 240 360 480 600 720 840 96010801200 
Time (min) 
% w/w of 
drug 
20 
30 
40 
50 
60 
70 
80 
90 
+ + + + + + + + + + + + + + + + + + + + + + + + + 
+ 
++++++++++++++ 
+ 
threshold. The masking effect of the swelling process on the drug percolation threshold 
has to be taken into account. 
2. Estimation of Drug Percolation Threshold The drug percolation threshold 
was calculated using the property . described by Bonny and Leuenberger [40] . This 
property is defi ned by the equation 
. 
. 
= 
. 
b 
A C 2 s 
(5) 
where . is proportional to the square root of the effective diffusion coeffi cient D eff , 
which is expected to obey, in the nearby of the percolation threshold, the scaling 
law 
D kD eff c = . 0( ) . . . 
where D 0 is the diffusion coeffi cient of the drug in pure solvent, k a constant, . the 
total porosity of the matrix (sum of initial porosity and porosity due to the dissolution 
of the drug), . c the critical porosity or percolation threshold, and . the critical 
TABLE 22 Dissolution Data from Dextromethorphan – HBr/Eudragit RS - PM Matrices 
Drug Load 
(% w/w) n 
Q a b t . + Q = a r + b r t k 
b ± S. E. r k r 
20 248 1.43 . 10 . 4 ± 2.9 . 10 . 7 0.999 0.45335 0.999 
30 248 2.47 . 10 . 4 ± 4.7 . 10 . 7 0.999 0.54724 0.999 
40 248 3.38 . 10 . 4 ± 1.0 . 1.0 . 6 0.998 0.58981 0.999 
50 248 4.43 . 10 . 4 ± 2.0 . 1.0 . 6 0.998 0.59792 0.999 
65 241 8.60 . 10 . 4 ± 2.0 . 10 . 6 0.999 0.54536 0.999 
70 248 1.15 . 10 . 3 ± 2.0 . 10 . 6 0.999 0.54697 0.999 
80 241 1.45 . 10 . 3 ± 5.0 . 10 . 6 0.999 0.57677 0.999 
90 85 2.09 . 10 . 3 ± 2.9 . 10 . 5 0.992 0.71910 0.999 

TABLE 23 Calculation of Tablet Property b and Related Parameters in Matrices of 
Dextromethorphan Hydrobromide 
Drug (% w/w) . 0 . n b F b Probability b A . . 10 3 
20 0.145 0.300 248 248,833 9.9 . 10 . 16 0.216 0.218 
30 0.142 0.378 248 277,630 9.9 . 10 . 16 0.329 0.305 
40 0.129 0.453 248 88,245.6 9.9 . 10 . 16 0.450 0.357 
50 0.115 0.531 248 79,555.4 9.9 . 10 . 16 0.579 0.413 
65 0.091 0.657 241 126,817 9.9 . 10 . 16 0.787 0.687 
70 0.093 0.705 248 278,000 9.9 . 10 . 16 0.851 0.887 
80 0.083 0.799 241 97,483.4 9.9 . 10 . 16 0.995 1.033 
90 0.083 0.898 85 5,324.51 9.9 . 10 . 16 1.133 1.395 
exponent for conductivity. This exponent has a value of 2.0 in 3D systems.The values 
of . as well as the parameters involved in its calculation are shown in Table 23 . 
The percolation threshold of dextromethorphan hydrobromide was estimated as 
the intersection with the Y axis from a linear regression of the total porosity, . , 
versus the property . (Figure 38 ). Following the method of Bonny and Leuenberger, 
only the . values above p c1 showing a linear dependence on the total porosity 
(circles in Figure 38 ) are considered in the regression. The selected . values corresponded 
to matrices with 50 – 80% w/w of drug. 
The estimated critical porosity is 0.3691 ± 0.0541, considering a 95% confi dence 
interval ( P = 0.05). This range corresponds to a dextromethorphan hydrobromide 
content of between 23 and 36% w/w. 
Estimation of the percolation threshold by visual methods is not very accurate, 
mainly due to extrapolation from 2D to 3D systems. Nevertheless, scanning electron 
microscopy was employed as an auxiliary technique in order to investigate the distribution 
of the particles of dextromethorphan hydrobromide in the matrices. 
Figure 39 shows two scanning electron microscopy (SEM) micrographs corresponding 
to the tablet side facing the lower punch for matrices containing 20 and 
FIGURE 38 Determination of drug percolation threshold. The circles represent the values 
selected for the regression, according to its linear behavior. These values correspond to drug 
loads between 50 and 80% w/w (three tablets per lot). Each point represents one experimental 
datum. 
0.8 
1 
0.6 
0.4 
0.2
0 
0 0.5 1 1.5 
., .103 
Total porosity 
PHARMACEUTICAL TABLETS USING PERCOLATION THEORY 1033

1034 TABLET DESIGN 
FIGURE 39 SEM micrographs corresponding to bottom side of matrices using BSE detector. 
The light gray particles correspond to dextromethorphan – HBr and the dark gray particles 
to the excipient Eudragit RS - PM. ( a ) Matrices containing 20% w/w of drug. ( b ) Matrices 
containing 30% w/w of drug. 
(a) 
(b) 
500 .m 
500 .m 
30% w/w of drug using backscattering electron (BSE) detector at the same magni- 
fi cation. In the tablet containing 30% of drug (Figure 39 b ), an infi nite drug cluster 
can be observed. The drug particles (light - gray particles) begin to form a connective 
network from the left to the right and from the top to the bottom of the micrograph. 
In the tablet containing 20% w/w of drug (Figure 39 a ), the particles of the drug 
(light - gray particles) seem to form isolated groups in the matrix. 
Therefore, considering both micrographs in Figure 39 , a 2D geometric phase 
transition can be observed. Figure 39 a shows the drug as gray particles on a black 
background, whereas in Figure 39 b there is a black - on - gray array, with black particles 
corresponding to Eudragit RS - PM surrounded by a gray background formed 
by dextromethorphan hydrobromide particles. 

When the cross section of these matrices (20 and 30% w/w drug loading) was 
observed (Figure 40 ), the same pattern was found, changing from gray on black 
(Figure 40 a , 20% w/w of drug) to black on gray (Figure 40 b , 30% w/w of drug). 
Therefore, according to the different methods employed, the drug percolation 
threshold in the studied matrices is expected to be between 20 and 30% w/w of 
dextromethorphan hydrobromide (total porosity between 30.0 and 37.8% v/v of 
drug). 
3. Estimation of Excipient Percolation Threshold . In principle, for binary pharmaceutical 
systems, two percolation thresholds are expected: the drug percolation 
FIGURE 40 SEM micrographs corresponding to cross section of tablets using BSE detector: 
( a ) matrices containing 20% w/w dextromethorphan – HBr (light gray particles); ( b ) matrices 
containing 30% w/w of drug. 
(a) 
(b) 
500 .m 
500 .m 
PHARMACEUTICAL TABLETS USING PERCOLATION THEORY 1035

1036 TABLET DESIGN 
threshold p c1 and the excipient percolation threshold p c2 . The second is the point 
where the excipient ceases to percolate the system. 
Nevertheless, in a previous study dealing with inert matrices of naltrexone – HCl 
[74] , two different excipient percolation thresholds p c2 were found for the matrix - 
forming excipient Eudragit RS - PM: the site percolation threshold related to a 
change in the release kinetics and the site - bond percolation threshold derived from 
the mechanical properties of the tablet, where the excipient failed to maintain tablet 
integrity after the release assay. 
An evident change in the release kinetics between tablets containing 80 and 90% 
w/w of drug can be observed in Table 23 (from k . 0.57 to k . 0.7 in the Peppas 
equation). Therefore, the site percolation threshold of the excipient can be estimated 
between the matrices containing 80 and 90% w/w of dextromethorphan 
hydrobromide (10 – 20% v/v of excipient). Above this threshold, a percolating cluster 
of excipient particles exists. These particles are able to control the drug release 
kinetics, but their cohesion forces can be insuffi cient to maintain tablet integrity 
after the release assay. 
Formulations containing more than 65% w/w of drug were unable to maintain 
tablet integrity after the 20 - h release assay. According to this result, the site - bond 
percolation threshold of the excipient ranges between 65 and 70% w/w of drug, 
corresponding to 29.5 and 34% v/v of excipient. Above this percolation threshold, 
that is, for concentrations of excipient > 34% v/v, there is a percolating cluster of 
excipient particles bound by suffi cient forces to maintain tablet integrity after drug 
release. 
In conclusion, according to percolation theory, the studied matrices should be 
formulated with drug content between 30 and 65% w/w (37.8 – 66% v/v of total 
porosity). These concentrations are optimal to ensure release of the total drug dose, 
to have controlled release of the drug, and to avoid disintegration of the matrix. In 
order to increase the robustness of the formulation, the limits of this range should 
be avoided. 
6.3.9.2 Critical Points of Hydrophilic Matrix Tablets 
Recently percolation theory is starting to be applied to the study of hydrophilic 
matrix systems. Figure 41 shows an example of the changes observed in several 
release parameters employed to estimate the critical point and the related percolation 
threshold in hydrophilic matrices prepared using KCl as the model drug [73] . 
Application of the percolation theory allows explanation of the changes in the 
release and hydration kinetics of swellable matrix - type controlled delivery systems. 
According to this theory, the critical points observed in dissolution and water uptake 
studies can be attributed to the excipient percolation threshold. Knowledge of these 
thresholds is important in order to optimize the design of swellable matrix tablets. 
Above the excipient percolation threshold an infi nite cluster of this component is 
formed which is able to control the hydration and release rate. Below this threshold 
the excipient does not percolate the system and drug release is not controlled. 
Miranda et al. demonstrated experimentally the infl uence of the particle size of 
the components on the percolation threshold in hydrophilic matrices as well as the 
importance of the initial porosity in the formation of the gel layer (sample - spanning 
cluster of excipient) [74] . 

FIGURE 41 ( a ) Higuchi slope; ( b ) normalized Higuchi slope; ( c ) relaxational constant of 
Peppas and Sahlin versus percentage of excipient volumetric fraction for batch A (50 – 100 . m 
KCl and 150 – 200 . m HPMC K4M). 
60 
50 
40 
30 
20 
10
0
0 
5
4
3
2
1
0 
0 
25 
20 
15 
10
5
0 
0 10 20 30 40 50 60 70 80 90 100 
10 20 30 40 50 60 70 80 90 10 20 30 40 50 60 70 80 90 100 
(a) (b) 
(c) 
% v/v HPMC % v/v HPMC 
% v/v HPMC 
y = –2.8783x + 93.63 
R2 = 0.8731 
y = –0.1124x + 15.01 
R2 = 0.9903 
y = –0.0172x + 1.7095 
R2 = 0.9414 
y = –1.4671x + 39.337 
R2 = 0.8063 
y = –0.2704x + 7.4919 
R2 = 0.8973 
y = –0.0047x + 0.4418 
R2 = 0.9283 
6.3.9.3 Case Study: Estimation of Percolation Thresholds in Acyclovir 
Hydrophilic Matrix Tablets 
The principles of the percolation theory were applied to design controlled - release 
matrix tablets containing acyclovir in order to estimate the percolation threshold of 
the excipient in acyclovir matrix tablets and to characterize the release behavior of 
these hydrophilic matrices in order to rationalize the design of these controlled - 
release systems. 
Acyclovir is a potent inhibitory of viruses of the herpes group, particularly herpes 
simplex virus (HSV I and II) and herpes zoster varicella virus. Unfortunately, acyclovir 
has a short half - life (2 – 3 h), and the oral dosage form must be taken fi ve times 
daily, which is very inconvenient for patients [75, 76] . Consequently, the aim of this 
study was to develop a controlled - release formulation of acyclovir that could be 
taken twice daily. The materials used to prepare the tablets were acyclovir (Kern 
Pharma, Tarrasa, Barcelona) and hydroxypropyl methylcellulose (Methocel K4M) 
(Colorcon) a hydrophilic cellulose derivative as the matrix - forming material. 
Binary mixtures were prepared with varying drug contents (60, 70, 80, 90, and 
95%) keeping constant the drug and excipient particle size. Table 24 gives the composition 
of the studied batches as well as the tablet thicknesses. The mixtures were 
compressed on an eccentric machine (Bonals A - 300) without any further excipient. 
Cylindrical tablets with a mean dosage of 500 mg and a diameter of 12 mm were 
prepared at the maximum compression force accepted by the formulations. 
PHARMACEUTICAL TABLETS USING PERCOLATION THEORY 1037

1038 TABLET DESIGN 
TABLE 24 Composition of Hydrophilic Matrices Prepared with Acyclovir/ HPMC K4M 
(150 – 200 m m) and Percent HPMC plus Initial Porosity 
Batch 
Percent w/w Percent (v/v) HPMC 
+ Initial Porosity 
Tablet Thickness 
(mm) Acyclovir HPMC K4M 
A 95 5 20.76 3.01 ± 0.056 
B 90 10 26.41 3.25 ± 0.052 
C 80 20 37.60 3.84 ± 0.051 
D 70 30 45.11 4.68 ± 0.063 
E 60 40 55.82 5.81 ± 0.056 
The release profi les were measured with the USP 25 dissolution apparatus 2 
(Turu Grau, model D - 6) at 100 rpm in distilled water (900 mL) at 37 ± 0.5 ° C for 
12 h. Filtered samples taken at different times were determined for acyclovir content 
through ultraviolet absorption at . max (242 nm). 
Figure 42 shows the release profi les obtained from hydrophilic matrices formulated 
with acyclovir and HPMC K4M 150 – 200 . m. 
In order to study the release mechanism of acyclovir from the tablets, the fi tting 
of the drug release data to the following kinetic equations has been studied: zero - 
order equation, Q = k 0 t ; Higuchi equation [77] , Q = k H t 1/2 ; Korsmeyer – Peppas equation 
[78] , Q = kt n ; and Peppas - Sahlin equation [79] , Q k k m m = + d r
2 , where Q is the 
amount of drug remaining at time t , k 0 is the zero - order release constant, k H is the 
Higuchi rate constant, k is the Korsmeyer – Peppas kinetic constant, n is the exponent 
indicative of the release mechanism (for matrix tablets an n value of 0.5 indicates 
diffusion control and an n value of 1.0 indicates erosion or relaxation control [80] , 
intermediate values suggest that at least two processes contribute to the overall 
release mechanism), k d is the diffusion rate constant, k r is the relaxation rate constant, 
and m is the purely Fickian diffusion exponent for a device of any geometric 
shape which exhibits controlled release. In our case, the aspect ratios and exponent 
values ( m ) are shown in Table 25 [79] . The results obtained are shown in Table 26 . 
FIGURE 42 Acyclovir release from matrix tablet with total drug content of 95, 90, 80, 70, 
and 60 prepared with acyclovir – HPMC K4M (150 – 200 . m) (mean ± SD, n = 3). 
120 
100 
80 
60 
40 
20
0
0 100 200 300 400 
Time (min) 
% Acyclovir release 
95% acyclovir 90% acyclovir 80% acyclovir 
70% acyclovir 60% acyclovir 
 .

TABLE 25 Aspect Ratios and Exponent Values ( m ) for 
Hydrophilic Matrices Studied 
Batch Aspect Ratio Exponent (m) 
A 3.80 0.45 
B 3.59 0.44 
C 3.03 0.43 
D 2.65 0.42 
E 2.16 0.43 
TABLE 26 Values of Kinetic constants Derived with Selected Equations in Range 
5 – 70% Acyclovir Release for All Batches Studied 
Batch A B C D E 
Acyclovir, 
% w/w 95 90 80 70 60 
Zero - order 
equation 
k 0 1.222 0.122 0.096 0.057 0.042 
r 2 0.984 0.974 0.994 0.995 0.987 
Sum of squares 
total 
3545.9 10,352.6 6,531.4 5,149.2 1,268.0 
Sum of squares 
residual 
53.7 278.3 51.8 24.8 16.4 
Higuchi 
equation 
k H 12.440 3.4167 2.8518 1.7683 1.4694 
r 2 0.998 0.993 0.956 0.932 0.959 
Sum of squares 
total 
3,545.9 10,352.6 12,367.8 5,149.2 1,268.0 
Sum of squares 
residual 
7.0 66.9 483.6 362.4 54.6 
Korsmeyer – 
Peppas 
equation 
k H 3.167 0.290 0.254 0.027 0.041 
n 0.782 0.843 0.856 1.114 1.008 
r 2 0.999 0.994 0.998 0.998 0.997 
Sum of squares 
total 
13,220.9 26,297.6 33,309.1 13,879.4 5,673.5 
Sum of squares 
residual 
16.7 159.5 65.5 10.1 15.2 
Peppas and 
Sahlin 
equation 
k d 2.239 2.056 0.357 . 0.81 . 0.38 
k r 1.615 0.161 0.205 0.202 0.127 
r 2 0.998 0.999 0.994 0.997 0.997 
Sum of squares 
total 
49,567.0 13,220.9 28,716.1 13,879.4 5,673.5 
Sum of squares 
residual 
42.9 23.3 165.0 43.7 15.2 
Notes: k 0 (%/min), zero - order constant; k H (%/min 1/2 ), Higuchi ’ s slope; k (%/min n ), kinetic constant of 
Korsmeyer model; n , diffusional exponent; k d (%/min m ), diffusional constant of Peppas and Sahlin model; 
k r (%/min 2 m ), relaxational constant of Peppas and Sahlin model; m , diffusional exponent that depends 
on geometric shape of releasing device through its aspect ratio (see Table 25 ). 
The analysis of the release profi les and the kinetic data indicate two different 
behaviors and a sudden change between them. In the fi rst behavior, which corresponds 
to the matrices that release the drug at slow rates, the release was controlled 
by the fully hydrated gel layer. For these matrices, erosion of the hydrophilic gel 
structure has shown an important infl uence on drug release. This is indicated by the 
better fi t of the drug release kinetics to the zero - order equation, the n value of 
PHARMACEUTICAL TABLETS USING PERCOLATION THEORY 1039

1040 TABLET DESIGN 
Korsmeyer – Peppas equation near 1, and the higher value of the relaxation constant 
k r in comparison with the diffusion constant k d in the Peppas – Sahlin equation. 
Taking into account the drug solubility (2.5 mg/ML), prevalence of the erosion 
versus swelling mechanism can be expected. After the transition point, the tablets 
allow the free dissolution of the drug when they are exposed to the dissolution 
medium due to the fact that the gel layer is not established since the fi rst moment 
and, in these conditions, this structure cannot control the drug release. The 
Korsmeyer release rate increases from 0.290 to 3.167% min . 1/2 . For these matrices, 
according to the Higuchi ( r 2 = 0.998), Korsmeyer ( n = 0.782), and Peppas – Sahlin 
(k r < k d ) equations, drug release is governed by the diffusion process. 
In hydrophilic matrices the drug threshold is less evident than the excipient 
threshold, which is responsible for the release control [73] . In order to estimate the 
percolation threshold of HPMC K4M, different kinetic parameters were studied: 
Higuchi rate constant, normalized Higuchi rate constant, and relaxation rate constant. 
The evolution of these release parameters has been studied as a function of 
the sum of the excipient volumetric percentage plus initial porosity. Recent studies 
of our research group have found the existence of a sample - spanning cluster of 
excipient plus pores in the hydrophilic matrix before the matrix is placed in contact 
with the liquid, clearly infl uences the release kinetics of the drug [73] . 
Figures 42 – 45 show changes in the different kinetic parameters: the Higuchi rate 
constant, normalized Higuchi rate constant, and relaxation rate constant. To estimate 
the excipient percolation threshold, these parameters were plotted versus the 
excipient volumetric fraction plus initial porosity. 
The kinetic parameters studied show a nonlinear behavior as a function of the 
volumetric fraction of the excipient plus initial porosity. As an approximation for 
estimating the trend of the parameter, one regression line has been performed 
below and the other above the percolation threshold. The point of intersection 
between both regression lines has been taken as an estimation of the percolation 
threshold [73, 74] . 
As percolation theory predicts, the studied properties show a critical behavior 
as a function of the volumetric fraction of the components. A critical point has been 
found between 21 and 26% v/v of excipient plus initial porosity (see Table 24 ). This 
critical point can be attributed to the excipient percolation threshold. 
FIGURE 43 Higuchi slope (mean ± SD, n = 3) versus percentage of excipient volumetric 
fraction plus initial porosity for all batches studied. 
% v/v excipient plus initial porosity 
KH (%min - 1/2) 
16 
14 
12 
10
8
6
4
2
0 
0 10 20 30 40 50 60

FIGURE 44 Normalized Higuchi slope (mean ± SD, n = 3) versus percentage of excipient 
volumetric fraction plus initial porosity for all batches studied. 
% v/v excipient plus initial porosity 
KH (%min - 1/2)/% v/v excipient plus initial porosity
0.7 
0.6 
0.5 
0.4 
0.3 
0.2 
0.1 
–0.1
0 
0 10 20 30 40 50 60 
FIGURE 45 Relaxational constant of Peppas – Sahlin (mean ± SD, n = 3) versus percentage 
of excipient volumetric fraction plus initial porosity for all batches studied. 
0 
0 
0.5
1 
1.5
2 
10 20 30 40 50 60 
% v/v of excipiente plus initial porosity 
Kr (%/min - 2 m) 
The Effective Medium Approximation (EMA), based in some assumptions, 
allows us to employ linear regressions as an approximation of the behavior of a 
disordered system outside the critical range. Based on EMA theory, two linear 
regressions have been performed as an approximation for estimating the percolation 
threshold as the point of intersection between both regression lines (see Figures 
43 – 45 ). The values of the excipient percolation thresholds estimated for all the 
batches studied, based on the behavior of the kinetic parameters, ranged from 25.99 
to 26.77%. 
Therefore, the results obtained from the kinetics analysis are in agreement with 
the release profi les, indicating a clear change in the release rate and mechanism 
between matrices containing 90 and 95% w/w of drug (5 – 10% w/w of excipient). The 
existence of a critical point can be attributed to the excipient percolation threshold. 
From the point of view of percolation theory, this means that above 10% w/w of 
HPMC K4M, the existence of a network of HPMC (able to form a hydrated layer 
from the fi rst moment) controls the drug release. 
PHARMACEUTICAL TABLETS USING PERCOLATION THEORY 1041

1042 TABLET DESIGN 
The process of water penetration into hydrophilic matrix tablets was also studied 
using a modifi ed Enslin apparatus. This apparatus contains a fritter and a system to 
regulate the water level. When the tablet is placed on the fritter, the water is 
absorbed from a reservoir which is placed on a precision balance. The amount of 
water uptake at each time point was read from the balance as weight loss in the 
reservoir. Figure 46 shows the obtained release profi les. 
An increase in the rate of water uptake can be observed when the HPMC concentration 
decreases. A critical point was found between 90 and 95% w/w of acyclovir. 
This range corresponds with the critical point observed in release profi le studies. 
The water uptake data were subjected to the Davidson and Peppas model to calculate 
the rate of water penetration [81] . The results show a change in the water 
uptake constant between the matrices containing 90 – 95% w/w of acyclovir, which 
refl ects the presence of the critical point previously observed. 
Knowledge of the percolation threshold of the components of the matrix formulations 
contributes to improve their design. First, in order to develop robust formulations, 
that is, to reduce variability problems when they are prepared at industrial 
scale, it is important to know the concentrations corresponding to the percolation 
thresholds. The percolation thresholds correspond to formulations showing a high 
variability in their properties as a function of the volume fraction of their components. 
Therefore, in order to design robust dosage forms, the nearby of the percolation 
thresholds should be avoided. 
Second, the excipient percolation threshold in hydrophilic matrices represents 
the border between a fast release of the drug (below the threshold) and a drug 
release controlled by the formation of a coherent gel layer (above the excipient 
percolation threshold). Therefore, knowledge of this threshold will allow us to avoid 
the preparation of a number of unnecessary lots during the development of a pharmaceutical 
formulation, resulting in a reduction of the time to market. 
FIGURE 46 Weight gain of systems as function of swelling time for matrix tablet with total 
drug content of 95, 90, 80, 70, and 60% prepared with acyclovir – HPMC K4M (150 – 200 . m) 
(mean ± SE, n = 3). 
95% acyclovir 90% acyclovir 80% acyclovir 70% acyclovir 60% acyclovir 
+ 
+ 
+ 
+ 
+ 
+ 
+ 
+ 
+ 
+ 
+ 
+ 
+ 
+ 
+ 
+ 
+ 
+ 
+ 
+ 
+ 
+ 
+ 
800 
600 
400 
200
0 
% Water uptake/dry polymer 
Time (min) 
0 50 100 150 200 250 300 350 400

6.3.10 ULTRASOUND - ASSISTED TABLETING (A NEW PERSPECTIVE) 
The compression of a powder is a complex process that is usually affected by different 
kinds of problems. These problems have been widely investigated and mainly 
concern the volume reduction and the development of a strength between the particles 
of the powder suffi cient to ensure tablet integrity [82] . The application of 
ultrasonic energy shows a great ability to reduce and even avoid these problems 
[83] . Ultrasound refers to mechanical waves with a frequency above 18 kHz (the 
approximate limit of the human ear). In an ultrasound compression machine, this 
vibration is obtained by means of a piezoelectric material (typically ceramics) that 
acts as a transducer of alternate electric energy of different frequencies in mechanical 
energy. An acoustic coupler, or “ booster, ” in contact with the transducer increases 
the amplitude of the vibration before it is transmitted (usually in combination with 
mechanical pressure) to the material to be compressed. 
Ultrasound - assisted powder compression has been widely employed in metallurgy 
as well as in the plastic and ceramic industries [84] . The fi rst references in the 
pharmaceutical industry are two patents in 1993 [85] and 1994 [86] . Since then, some 
papers have presented experimental data in this fi eld [45, 87 – 92] . 
Two main objectives are pursued nowadays by means of the application of 
ultrasound - assisted compression: 
1. Increase in the drug dissolution rate due to amorphization of the drug 
2. Preparation of controlled - release dosage forms with thermoplastic 
excipients 
As a consequence of the application of ultrasonic energy, the drug can lose its 
crystalline structure. This will result in an increase of the dissolution rate of the 
active substance, which can be very adequate for slowly dissolving drugs. Nevertheless, 
depending on the storage conditions, the drug can recover, at least partially, 
its crystallinity [89, 90] . 
To overcome this problem, it has been proposed to use an adequate excipient, 
preventing the recovery of the crystallinity, leading in some cases to the preparation 
of solid solutions into the die of the tableting machine. 
Several analytical techniques, such as infrared (IR) spectroscopy, differential 
scanning calorimetry, HPLC, and thin - layer chromatography (TLC), have been used 
to investigate possible drug degradation due to ultrasonic energy. No important 
permanent modifi cation of the drug has been found, with the exception of the loss 
of crystallinity [89, 90] . 
Concerning the design of controlled - release dosage forms, using a thermoplastic 
excipient (e.g., copolymers of acrylic and metacrylic acid), an important decrease in 
the release rate has been found for tablets compressed with the assistance of ultrasonic 
energy in comparison with traditional tablets. 
Although the effects of the ultrasonic energy on the material are not completely 
clarifi ed, this slow release rate has been attributed to different phenomena: 
Mechanical Pressure This pressure is exerted by the punches of the ultrasound - 
assisted tableting machine. This is the main compression mechanism when 
low ultrasonic energies are employed (below 25 J in the mixtures studied by 
ULTRASOUND-ASSISTED TABLETING (A NEW PERSPECTIVE) 1043

1044 TABLET DESIGN 
Rodriguez et al. [87, 88] ) or when the materials used are not thermoplastic. 
In these cases the machine acts as a multiple - impact mechanical press. 
Thermal Effects Due to the poor conductivity for ultrasounds (low module of 
elasticity and high quantity of air trapped inside) usually exhibited by the 
materials included in pharmaceutical formulations, a fast decay of ultrasonic 
energy to thermal energy is obtained. This process has been studied, monitoring 
the temperature inside the compression chamber by means of a thermistor. 
In the studied mixtures [87, 88] , a fast rise in temperature was obtained in 
tenths of a second followed by a relatively fast decrease (see Figure 47 ). 
The peak temperature obtained for low ultrasonic energy (25 J) is below 80 ° C, 
whereas for high energies (125 – 150 J) it is above 140 ° C. In mixtures of ketoprofen 
with acrylic polymers [90] , the increase in temperature was slightly 
lower. In this respect it must be mentioned that a recent modifi cation of the 
ultrasound - assisted tableting machine that involves the suppression of Tefl on 
isolators in contact with the powder must result in a faster decrease in temperature 
inside the compression chamber. Thermal effects can cause the total 
or partial fusion of some components of the formulation. Nevertheless, in the 
assayed controlled - release formulations, the components are usually below its 
melting points. 
Plastic Deformation Plastic deformation results from the combination of 
thermal and mechanical effects. The thermoplastic excipient was subjected to 
a temperature above its glass transition temperature ( T g ) and to a high - frequency 
mechanical pressure that can avoid the elastic recovery of the 
material. 
Sintering The combination of temperature, pressure, and friction effects can 
result in the sintering of particles, so that the limits between them are no 
longer evident [46, 87] . 
Recent studies [91 – 93] have shown that, for one component of the system undergoing 
thermoplastic deformation, the continuum percolation model can be used to 
predict the changes in the system with respect to a traditional pharmaceutical 
FIGURE 47 Temperature profi le inside compression chamber. (Courtesy of Tecnea Srl.) 
0 2 4 6 8 16 18 20 14 12 10 
0 
20 
40 
60 
80 
100 
120 
Time (sec) 
Temperature (°C)

dosage form. The continuum percolation model dispenses with the existence of a 
regular lattice underlying the system; therefore, the substance is not distributed into 
discrete lattice sites. This model deals with the volume ratio of each component 
and a continuum distribution function. The volume ratio is expressed as a space – 
occupation probability to describe the behavior of the substance [94, 95] . 
The continuum percolation model predicts an excipient percolation threshold 
around 16% v/v. This can explain the important decrease in the critical point corresponding 
to the excipient percolation threshold, a critical point that governs the 
mechanical and release properties of the matrix. 
Ultrasound compaction lowers the percolation threshold of the thermoplastic 
excipient, resulting in a drastic reduction (about 50%) in the amount of matrix - 
forming excipient [98] needed to obtain the controlled-release system as well as in 
a better control of the drug release. The structure of the excipient inside the US - 
tablets does not correspond to a particulate system but to an almost continuous 
medium; therefore, there is no an excipient particle size inside these matrices (see 
Figure 48 ). Consequently, the percolation threshold of the active agent is higher 
than in traditional tablets. The insoluble excipient almost surrounds the active agent 
particles, slowing down the contact with the dissolution medium. 
These facts can involve important advantages for the pharmaceutical industry, 
such as the preparation of controlled - release inert matrices containing high drug 
doses, with very little increase in the weight of the system. This fact is especially 
interesting when a high drug dose has to be included in the dosage form, as frequently 
occurs in controlled - release systems. 
On the other hand, application of ultrasonic energy results in an increase in the 
temperature of the die during the compaction process. The consequences of this 
fact should be taken into account and cannot be neglected in the case of thermolabile 
drugs and/or excipients [87, 88] . 
Further research is needed in the area of ultrasound - assisted compression of 
pharmaceutical powders, including a higher number of drugs and excipients. 
FIGURE 48 SEM micrograph of matrix tablet containing potassium chloride as drug model 
and commercial acrylic – metacrylic copolymer. The white KCI particles are surrounded by an 
almost continuous dark gray mass of excipient. (Courtesy of M. Mill a n.) 
ULTRASOUND-ASSISTED TABLETING (A NEW PERSPECTIVE) 1045

1046 TABLET DESIGN 
REFERENCES 
1. Unckel , H. ( 1945 ), Processes during compression of metal powders , Archiv fuer das 
Eisenhuettenwesen , 18 , 161 – 167 . 
2. Toor , H. L. , and Eagleton , S. D. ( 1956 ), Plug fl ow and lubrication of polymer particles , 
Ind. Eng. Chem. , 48 , 1825 – 1830 . 
3. Ritschel , W. A. , and Bauer - Brandl , A. ( 2002 ), Die Tablette, Handbuch der Entwicklung, 
Herstellung und Qualit a tssicherung , Editio Cantor Verlag Aulendorf, (W u rtt), Germany 
505. 
4. De Boer , A. H. , Bolhuis , G. K. , and Lerk , C. F. , ( 1978 ), Bonding characteristics by 
scanning electron microscopy of powders mixed with magnesium stearate , Powder 
Technol. , 20 , 75 – 82 . 
5. Nystr o m , C. , Alderborn , G. , Duberg , M. , and Karehill , P - G. ( 1993 ), Bonding surface 
area and bonding mechanism — Two important factors for the understanding of powder 
compactability , Drug Dev. Ind. Pharm. , 19 , 2143 – 2196 . 
6. Mattsson , S. , and Nystr o m , C. ( 2000 ), Evaluation of strength - enhancing factors of a 
ductile binder in direct compression of sodium bicarbonate and calcium carbonate 
powders , Eur. J. Pharm. Sci. , 10 , 53 – 66 . 
7. Rudnic , E. M. , and Kottke , M. K. ( 1999 ), Tablet dosage forms , in Banker , G. S. , and 
Rhodes , C. T. , Eds., Modern Pharmaceutics , 3rd rev. and expanded eds., Marcel Dekker , 
New York , pp. 333 – 394 . 
8. Pesonen , T. , Paronen , P. , and Ketolainen , J. ( 1989 ), Disintegrant properties of an 
agglomerated cellulose powder , Int. J. Pharm. , 57 , 139 – 147 . 
9. Rudnic , E. M. , and Kottke , M. K. , Tablet dosage forms . In Banker GS , Rhodes CT Eds., 
Modern Pharmaceutics 3rd eds. New York : Marcel Dekker ; 1999 , pp 333 – 394 . 
10. Westerberg , M. , and Nystrom , C. ( 1991 ), Physicochemical aspects of drug release. XII. 
The effect of some carrier particle properties and lubricant admixture on drug dissolution 
from tableted ordered mixtures , Int. J. Pharm. , 69 , 129 – 141 . 
11. Bolhuis , G. K. , and H o lzer , A. W. , Lubricant sensitivity. VIII. Effect of third components 
on the fi lm formation of lubricants . In Alberborn G , Nystr o m C Eds. Pharmaceutical 
Powder Compactation Technology , New York : Marcel Dekker ; 1996 , pp 550 – 555 . 
12. Johansson , M. E. ( 1984 ), Granular magnesium stearate as a lubricant in tablet formulations 
, Int. J. Pharm. , 21 , 307 – 315 . 
13. Carson , J. W. ( 1998 July), Overcoming particle segregation in the pharmaceutical and 
cosmetic industries, paper presented at Interphex USA, New York. 
14. Vila - Jato , J. L. , Remu n a n , M. C. , Seijo , B. , and Torres , D. ( 1997 ), Nuevas formas de 
administraci o n de medicamentos , in Vila - Jato , J. L. , Ed., Tecnolog i a Farmac e utica II: 
Formas Farmac e uticas , Editorial S i ntesis , Madrid . 
15. Tahara , K. , Yamamoto , K. , and Nishihata , T. ( 1995 ), Overall mechanism behind matrix 
sustained release (SR) tablets prepared with hydroxypropyl methylcellulose 2910 , 
J. Controlled Release , 35 , 59 – 66 . 
16. Soares , L. A. L. , et al . ( 2005 ), Dry granulation and compression of spray dried plant 
extracts , AAPS PharmSciTech , 6 , E359 – E366 . 
17. N u n ez Sell e s , A. J. , Capote , H. R. , Ag u ero , J. , Garrido , G. , Delgado , R. , Martinez , G. , 
Leon , O. S. , and Morales , M. ( 2000 ), New antioxidant product derived from Mangifera 
indica L, Abstracts of Papers , paper presented at the 220th National Meeting of 
the American Chemical Society, New Orleans, LA, American Chemical Society, 
Washington, DC. 

18. S a nchez , G. M. , Re , L. , Giuliani , A. , N u nez - Sell e s , A. J. , Davison , G. P. , and Le o n - 
Fern a ndez , O. S. ( 2000 ), Protective effects of Magnifera indica L. extract, mangiferin 
and selected antioxidents against TPA - induced biomolecules oxidation and peritoneal 
macrophage activation in mice , Pharmacol. Res. , 42 , 565 – 573 . 
19. Yoshimi , N. , Matsunaga , K. , Katayama , M. , Yamada , Y. , Kuno , T. , Qiao , Z. , Hara , A. , 
Yamahara , J. , and Mori , H. ( 2001 ), The inhibitory effects of mangiferin, a naturally 
occurring glucosylxanthone, in bowel carcinogenesis of male F344 rats , Cancer Lett , 163 , 
163 – 170 . 
20. Ichiki , H. , Miura , T. , Masayoshi , I. , Ishihara , E. , Komatsu , Y. , Tanigawa , K. , Ichiki , H. , 
and Okada , M. ( 1998 ). New antidiabetic compounds, mangiferin and its glucoside , Biol. 
Pharm. Bull. , 21 , 1389 . 
21. Li , H. , Miyahara , T. , Tezuka , Y. , Namba , T. , Nemoto , N. , Tonami , S. , Seto , H. , Tada , 
T. , and Kadota , S. ( 1998 ), The effect of Kampo formulae of bone resorption in vitro 
and in vivo. I. Active constituents of Tsu-Kan-gan. Biol. Pharm. Bull. , 21 , 1322 . 
22. Guha , S. , Ghosal , S. , and Chattopadhyay , U. ( 1996 ), Antitumor, immunomodulatory 
and anti - HIV effect of mangiferin, a naturally occurring glucosylxan thone , Chemotherapy 
, 42 , 443 – 451 . 
23. Ghosal , S. , Rao , G. , Saravanan , V. , Misra , N. , and Rana , D. ( 1996 ), A plausible chemical 
mechanism of the bioactivities of mangiferin , Indian J. Chem. , 35B ( 6 ), 561 – 566 . 
24. Born , M. , Carrupt , P. A. , Zini , R. , Bree , T. , Tillement , J. P. , Hostettmann , K. , and Testa , 
B. ( 1996 ), Electrochemical behaviour and antioxidant activity of some natural polyphenols 
, Helv. Chim. Acta , 79 , 1147 . 
25. Castellanos Gil , E. , Iraizoz Colarte , A. , Bataille , B. , Pedr a z , J. L. , and Hein a m a ki , J. 
( 2006 ), Development and optimization of a novel sustained - release dextran tablet formulation 
for propranolol hydrochloride , Int. J. Pharm. , 317 , 32 – 39 . 
26. Castellanos Gil , E. , Bataille , B. , Iraizoz Colarte , A. , Delarbre , J. L. , El Ghzaoui , A. , 
and Durand , D. ( 2008 ), A sugar cane native dextran as an innovative functional excipient 
for the development of pharmaceutical tablets . Eur. J. Pharm. Biopharm 68 ( 2 ), 
319 – 329 . 
27. Jamzad , S. , Tutunji , L. , and Fassihi , R. ( 2005 ), Analysis of macromolecular changes and 
drug release from hydrophilic matrix systems , Int. J. Pharm. , 292 , 75 – 85 . 
28. Reynolds , T. D. , Gehrke , S. H. , Hussain , A. S. , and Shenouda , L. S. ( 1998 ), Polymer 
erosion and drug release characterization of hydroxypropyl methylcellulose matrices . J. 
Pharm. Sci. , 87 , 1115 – 1123 . 
29. Miranda , A. , Mill a n , M. , and Caraballo , I. ( 2006 ), Study of the critical points in Lobenzarit 
disodium hydrophilic matrices for controlled drug delivery , Chem. Pharm. Bull. 
54 , 598 – 602 . 
30. Anonymous ( 1999 , July), Note for guidance on quality of modifi ed release products. 
Oral dosage forms and transdermal dosage forms, section 1 (quality), The European 
Agency for the Evaluation of Medicinal Products Human Medicines Evaluation 
(EMEA), London. 
31. Kincl , M. , Turk , S. , and Vrecer , F. ( 2005 ), Application of experimental design methodology 
(DOE) in development and optimization of drug release method , Int. J. Pharm. , 
291 , 39 – 49 . 
32. Eriksson , L. , Johansson , E. , and Wikstrom , C. ( 1998 ), Mixture design — Design generation, 
PLS analysis, and model usage , Chemometr. Intell. Lab. Syst. , 43 , 1 – 24 . 
33. Gooding , O. W. ( 2004 ), Process optimization using combinatorial design principles: 
Parallel synthesis and design of experiment methods , Curr. Opin. Chem. Biol. , 8 , 
297 – 304 . 
REFERENCES 1047

1048 TABLET DESIGN 
34. Congreve , M. S. , and Jamieson , C. ( 2002 ), High - throughput analytical techniques for 
reaction optimization , Drug Discovery Today , 2 , 139 – 142 . 
35. I HOLLAND ( 2006 ), ITab software, available: http://www.in-pharmatechnologist. 
com. 
36. Leuenberger , H. , Rohera , B. D. , and Haas , C. ( 1987 ), Percolation theory — A novel 
approach to solid dosage form design , Int. J. Pharm. , 38 , 109 – 115 . 
37. Holman , L. E. , and Leuenberger , H. ( 1988 ), The relationship between solid fraction and 
mechanical properties of compacts — The percolation theory model approach , Int. J. 
Pharm. , 46 , 35 – 44 . 
38. Stauffer , D. , and Aharony , A. ( 1992 ), Introduction to Percolation Theory , 2nd ed., 
Burgess Science , London . 
39. Caraballo , I. , Fernandez Ar e valo , M. , Mill a n , M. , Rabasco , A. M. , and Leuenberger , H. 
( 1996 ), Study of percolation thresholds in ternary tablets , Int. J. Pharm. 139 , 177 – 186 . 
40. Bonny , J. D. , and Leuenberger , H. ( 1991 ), Matrix type controlled release systems: I. 
Effect of percolation on drug dissolution kinetics , Pharm. Acta Helv. , 66 , 160 – 164 . 
41. Bonny , J. D. , and Leuenberger , H. ( 1993 ), Matrix type controlled release systems. II. 
Percolation effects in non - swellable matrices , Pharm. Acta Helv. , 68 , 25 – 33 . 
42. Blattner , D. , Kolb , M. , and Leuenberger , H. ( 1990 ), Percolation theory and compactibility 
of binary powder systems , Pharm. Res. , 7 , 113 – 117 . 
43. Caraballo , I. , Fernandez - Ar e valo , M. , Holgado , M. A. , and Rabasco , A. M. ( 1993 ), 
Percolation theory: Application to the study of the release behaviour from inert matrix 
system , Int. J. Pharm. , 96 , 175 – 181 . 
44. Caraballo , I. , Millan , M. , and Rabasco , A. M. ( 1996 ), Relationship between drug percolation 
threshold and particle size in matrix tablets , Pharm. Res. , 13 ( 3 ), 387 – 390 . 
45. Mill a n , M. ( 1998 Apr.), Estudio del Umbral de Percolaci o n para la optimizaci o n de 
matrices inertes de liberaci o n controlada, Ph.D. thesis, University of Seville, Seville, 
Spain. 
46. Mill a n , M. , Caraballo , I. , and Rabasco , A. M. ( 1998 ), The role of the drug/excipient 
particle size ratio in the percolation model for tablets , Pharm. Res. , 15 ( 2 ), 216 – 220 . 
47. Kuentz , M. T. , Leuenberger , H. , and Kolb , M. ( 1999 ), Fracture in disordered media and 
tensile strength of microcrystalline cellulose tablets at low relative densities , Int. J. 
Pharm. , 182 , 243 – 255 . 
48. Kuentz , M. T. , and Leuenberger , H. ( 2000 ), A new theoretical approach to tablet 
strength of a binary mixture consisting of a well and a poorly compactable substance , 
Eur. J. Pharm. Biopharm. , 49 , 151 – 159 . 
49. Ram i rez , N. , Melgoza , L. M. , Kuentz , M. , Sandoval , H. , and Caraballo , I. ( 2004 ), Comparison 
of different mathematical models for the tensile strength – relative density pro- 
fi les of binary tablets , Eur. J. Pharm. Sci. , 22 ( 1 ), 19 – 23 . 
50. Bourquin , J. , Schmidli , H. , van Hoogevest , P. , and Leuenberger , H. ( 1997 ), Basic concepts 
of artifi cial neural networks (ANN) modeling in the application to pharmaceutical 
development , Pharm. Dev. Technol. , 2 , 95 – 109 . 
51. Plumb , A. P. , Rowe , R. C. , York , P. , and Brown , M. ( 2005 ), Optimisation of the predictive 
ability of artifi cial neural network (ANN) models: A comparison of three ANN 
programs and four classes of training algorithm , Eur. J. Pharm. Sci. , 25 ( 4 – 5 ), 395 – 405 . 
52. Gans , E. H. , and Chavkin , L. ( 1954 ), The use of polyethylene glycol in tablet coating , 
Pharm. Ass. Sc. (Baltin) Ed., 43 , 483 – 485 . 
53. Porter , S. C. , and Bruno , C. H. ( 1990 ), Coating of pharmaceutical solid - dosage forms , 
in Liebermann , H. A. , Lachmann , L. , and Schwarz , J. B. , Eds., Pharmaceutical Dosage 
Forms: Tablets , Vol. 3, 2nd eds. , Marcel Dekker , New York , Chapter 2. 

54. Hess , H. , and Janssen , H. J. ( 1969 ), Lacquered tablets and fi lm coated tablets, Pharm . 
Acta Helv. , 44 , 581 . 
55. Bindsch a dler , C. , Gurny , R. , and Dolker , B. ( 1983 ), Theoretical concepts regarding the 
formation of fi lms from aqueous microdispersions and application to coating, Labo - 
Pharma , Probl. Tech. , 31 , 389 – 394 . 
56. Radebaugh , G. W. ( 1990 – 1996 ), Film coating and fi lm - forming materials: Evaluation , 
in Swarbrick , J. , and Boylan , J. C. , Eds., Encyclopedia of Pharmaceutical Technology , 
Vol. 6, Marcel Dekker , New York . 
57. Moe , E. ( 1945 ), Overtraeking og Dragering , Dansk Farmaceutfoeningsverl , 
Copenhagen . 
58. Hasegava , K. , Ida , T. , Saika , K. , and Utsumi , I. ( 1965 ), Suitable dimensions of tablets 
for coating , Yakugaku Zasshi , 85 , 796 . 
59. Selmeczi , B. , and Smogyi , J. ( 1996 ), Suitable dimensions of tablets for coating , Pharmacie 
, 21 , 604 . 
60. Bauer , K. H. , Fr o mming , K. H. , and F u hrer , C. ( 1993 ), Pharmazeutische Technologie 
[Pharmaceutical Technology ], 4th ed. , Thieme , Stuttgart . 
61. Bauer , K. H. , Lehmann , K. , Osterwald , H. P. , and Rothgang , G. ( 1998 ), Coated Pharmaceutical 
Dosage Forms , Medpharm Scientifi c Publisher , Stuttgart . 
62. B u hler , V. ( 2003 , Sept.), Kollidon. Polyvinylpyrrolidone for the Pharmaceutical Industry 
, 7th ed. , BASF Pharma Ingredients, Ludwigshafen , Germany . 
63. U.S. Pharmacopeial Forum (2002, May/June), Vol. 28. No. 3, 948–951. 
64. Heng , P. W. S. , Wan , L. S. C. , and Tan , Y. T. F. ( 1996 ), Relationship between aggregation 
of HPMC coated spheroids and tackiness/viscosity/additives of the coating formulations 
, Int. J. Pharm. , 138 , 57 – 66 . 
65. Tan , Y. T. F. , Wan , L. S. C. , and Heng , P. W. S. ( 1998 ), Evaluation of adhesion strength 
measurement for predicting aggregation propensity during fl uidized bed coating , S.T.P. 
Pharm. Sci. , 8 ( 3 ) 149 – 153 . 
66. B u hler , V. ( 2001 ), Generic Drug Formulation , 4th ed. , BASF Fine Chemicals, Ludwigshafen 
, Germany . 
67. Rowe , R. C. , and Forse , S. F. ( 1983 ), Pitting — A defect on fi lm - coated tablets , Int. J. 
Pharm. , 17 ( 2 – 3 ), 347 – 349 . 
68. Melgoza , L. M. , Caraballo , I. , Alvarez - Fuentes , J. , Millan , M. , and Rabasco , A. M. 
( 1998 ), Study of morphine hydrochloride percolation threshold in Eudragit® RS - PM 
matrices , Int. J. Pharm. , 170 , 169 – 177 . 
69. Caraballo , I. , Melgoza , L. M. , Alvarez - Fuentes , J. , Soriano , M. C. , and Rabasco , A. M. 
( 1999 ), Design of controlled release inert matrices of naltrexone hydrochloride based 
on percolation concepts , Int. J. Pharm. , 181 , 23 – 30 . 
70. Melgoza , L. M. , Rabasco , A. M. , Sandoval , H. , and Caraballo , I. ( 2001 ), Estimation of 
the percolation thresholds in dextromethorphan hydrobromide matrices , Eur. J. Pharm. 
Sci. , 12 ( 4 ), 453 – 459 . 
71. Boza , A. , Blanquero , R. , Mill a n , M. , and Caraballo , I. ( 2004 ), Application of a new 
mathematical method for the estimation of the mean surface area to calculate the percolation 
threshold of Lobenzarit disodium salt in controlled release matrices , Chem. 
Pharm. Bull. , 52 ( 7 ), 797 – 801 . 
72. Reisine , T. , and Pasternak , G. ( 1996 ), Analg e sicos opioides y sus antagonistas , in 
Goodman , A. , Hardman , J. G. , Limbird , L. E. , Molinoff , P. B. , and Ruddon , R. W. Eds., 
Las Bases Farmacol o gicas de la Terap e utica , Vol. I, McGraw - Hill Interamericana , M e xico , 
pp. 557 – 593 . 
REFERENCES 1049

1050 TABLET DESIGN 
73. Miranda , A. , Mill a n , M. , and Caraballo , I. ( 2006 ), Study of the critical points of HPMC 
hydrophilic matrices for controlled drug delivery , Int. J. Pharm. , 311 , 75 – 81 . 
74. Miranda , A. , Mill a n , M. , and Caraballo , I. ( 2006 ), Study of the critical points in 
Lobenzarit disodium hydrophilic matrices for controlled drug delivery , Chem. Pharm. 
Bull. , 54 , 598 – 602 . 
75. Tu , J. , Wang , L. , Yang , J. , and Li , X. ( 2001 ), Formulation and pharmacokinetic studies 
of a cyclovir controlled - release capsules , Drug Dev. Ind. Pharm. , 27 , 687 – 692 . 
76. Wangstaff , J. A. , Faulds , D. , and Goa , L. K. ( 1994 ), Acyclovir: A reappraisal of its 
antiviral activity pharmacokinetic properties and therapeutic effi cacy , Drugs , 47 ( 1 ), 
153 – 205 . 
77. Higuchi , T. ( 1963 ), Mechanism of sustained - action medication. Theoretical analysis of 
rate of release of solid drugs dispersed in solid matrices , J. Pharm. Sci. , 52 , 1145 – 1149 . 
78. Korsmeyer , R. W. , Gurny , R. , Doelker , E. , Buri , P. , and Peppas , N. A. ( 1983 ), Mechanisms 
of solute release from porous hydrophilic polymers , Int. J. Pharm. , 15 , 25 – 35 . 
79. Peppas , N. A. , and Sahlin , J. J. ( 1989 ), A simple equation for the description of solute 
release. 3. Coupling of diffusion and relaxation , Int. J. Pharm. , 57 , 169 – 172 . 
80. Ford , J. L. , Mitchell , K. , Rowe , P. , Armstrong , D. J. , Elliot , P. N. C. , Rostron , C. , and 
Hogan , J. E. ( 1991 ), Mathematical modelling of drug release from hydroxypropylmethylcelluose 
matrices: Effect of temperature , Int. J. Pharm. , 71 , 95 – 104 . 
81. Fuertes , I. , Miranda , A. , Mill a n , M. , and Caraballo , I. ( 2006 ), Estimation of the percolation 
thresholds in acyclovir hydrophilic matrix tablets , Eur. J. Pharm. Biopharm. , 64 ( 3 ), 
336 – 342 . 
82. Leuenberger , H. , and Rohera , B. D. ( 1986 ), Fundamentals of powder compression. II. 
The compression of binary powder mixtures , Pharm. Res. , 3 , 65 – 74 . 
83. Levina , M. , Rubinstein , M. H. , and Rajabi - Siahboomi , A. R. ( 2000 ), Principles and 
application of ultrasound in pharmaceutical powder compression , Pharm. Res. , 17 ( 3 ), 
257 – 265 . 
84. Kromp , W. , Trimmel , P. , Prinz , F. B. , and Williams , J. C. ( 1985 ), Vibratory compaction 
of metal powders, Mod . Dev. Powder Metall. , 15 , 131 – 141 . 
85. Gueret , J. - L. H. ( 1993 ), Process for the compaction of a powder mixture providing an 
absorbent or partially friable compact product and the product obtained by this process, 
U.S. Patent 5,211,892. 
86. Motta , G. ( 1994 ), Process for preparing controlled release pharmaceutical forms and 
the forms thus obtained, International Patent WO 94/14421. 
87. Rodriguez , L. , Cini , M. , Cavallari , C. , Passerini , N. , Saettone , M. F. , Fini , A. , and 
Caputo , O. ( 1997 ), Physico - chemical properties of some materials compacted using an 
ultrasound - assisted tableting machine , in Rubinstein, M., Ed., Proceedings 16th Pharmaceutical 
Technology Conference, Athens, Greece , Vol. I , pp. 267 – 278 . 
88. Rodriguez , L. , Cini , M. , Cavallari , C. , Passerini , N. , Fabrizio Saettone , M. , Fini , A. , and 
Caputo , O. ( 1998 ), Evaluation of theophylline tablets compacted by means of novel 
ultrasound - assisted apparatus , Int. J. Pharm. , 170 , 201 – 208 . 
89. Fini , A. , Fern a ndez - Herv a s , M. J. , Holgado , M. A. , Rodriguez , L. , Cavallari , C. , Passerini 
, N. , and Caputo , O. ( 1997 ), Fractal analysis of beta - cyclodextrin - indomethacin 
particles compacted by ultrasound , J. Pharm. Sci. , 86 , 1303 – 1309 . 
90. Sancin , P. , Caputo , O. , Cavallari , C. , Passerini , N. , Rodriguez , L. , Cini , M. , and Fini , A. 
( 1999 ), Effects of ultrasound - assisted compaction on ketoprogen/Eudragit S100 mixtures 
, Eur. J. Pharm. Sci. , 7 , 207 – 213 . 
91. Caraballo , I. , Millan , M. , Fini , A. , Rodriguez , L. , and Cavallari , C. ( 2000 ), Percolation 
thresholds in ultrasound compacted tablets , J. Controlled Release , 69 , 345 – 355 . 

92. Mill a n , M. , and Caraballo , I. ( 2006 ), Effect of drug particle size in ultrasound compacted 
tablets: Continuum percolation model approach , Int. J. Pharm. , 310 , 168 – 174 . 
93. Caraballo , I. ( 2001 ), Improvement on the release control using ultrasound assisted 
compression, Eurand Award 2001 “ for Outstanding Novel Research in Oral Drug 
Delivery, ” organized by Eurand and the Controlled Release Society, 28th Meeting of 
the Controlled Release Society, San Diego, CA, June 26. 
94. Efros , A. L. ( 1994 ), F i sica y Geometr i a del desorden , Hayka , Moscow , pp. 144 – 147 . 
95. Kuentz, M. T. , and Leuenberger , H. (1998), Modifi ed Young ’ s modulus of microcrystalline 
cellulose tablets and the directed continuum percolation model , Pharm. Dev. 
Technol. , 3(1 ), 1 – 7 . 
FURTHER READING 
A. BASF Pharma ingredients generic drug formulations, 2004, 2005. 
B. Baveja , S. K. , Ranga Roa , K. V. , and Padmalatha Devi , K. ( 1987 ), Zero - order release 
hydrophilic matrix tablets of . - adrenergic blockers , Int. J. Pharm. , 40 , 223 – 234 . 
C. Bettini , R. , Colombo , P. , Massimo , G. , Catellani , P. L. , and Vitali , T. ( 1994 ), Swelling and 
drug release hydrogel matrices: Polymer viscosity and matrix porosity effect , Eur. J. 
Pharm. Sci. , 2 , 213 – 219 . 
D. Campos - Aldrete , M. E. , and Villafuerte - Robles , L. ( 1997 ), Infl uence of the viscosity grade 
and the particle size of HPMC on metronidazole release from matrix tablet , Eur. J. Pharm. 
Biopharm. , 43 , 173 – 178 . 
E. Ferrero , C. , Mu n oz - Ruiz , A. , and Jim e nez - Castellanos , M. R. ( 2000 ), Fronts movements 
as a useful tool for hydrophilic matrix release mechanism elucidation , Int. J. Pharm. , 202 , 
21 – 28 . 
F. U.S. Pharmacopoeia 25 ( 2002 ), National Formulary 20, U.S. Pharmacopeial Convention, 
Rockville, MD. 
G. V a zquez , M. J. , Peres - Marcos , B. , G o mez - Amoza , J. L. , Mart i nez - Pacheco , R. , Souto , C. , 
and Concheiro , A. ( 1992 ), Infl uence of technological variables on release of drugs from 
hydrophilic matrices , Drug Dev. Ind. Pharm. , 18 , 1355 – 1375 . 
H. Velasco , M. V. , Ford , J. L. , Rowe , P. , and Rajabi - Siahboomi , A. R. ( 1999 ), Infl uence of drug: 
hydroxypropylmethylcellulose ratio, drug and polymer particle size and compression 
force on the release of diclofenac sodium from HPMC tablets , J. Controlled Release , 57 , 
75 – 85 . 
I. Tu , J. , Wang , L. , Yang , J. , Fei , H. , and Li , X. ( 2001 ), Formulation and pharmacokinetic 
studies of acyclovir controlled - release capsules , Drug Dev. Ind. Pharm. , 27 ( 7 ), 687 – 692 . 
FURTHER READING 1051


1053 
6.4 
TABLET PRODUCTION SYSTEMS 
Katharina M. Picker - Freyer 
Martin - Luther - University Halle - Wittenberg, Institute of Pharmacy, Division of 
Pharmaceutics and Biopharmaceutics, Halle/Saale, Germany 
Contents 
6.4.1 Introduction 
6.4.2 Physics of Tablet Formation 
6.4.2.1 Tableting Process 
6.4.2.2 Final Formation of Tablet 
6.4.3 Requirements for Tablet Production Systems 
6.4.4 Tablet Manufacturing Process 
6.4.4.1 Filling 
6.4.4.2 Compression 
6.4.4.3 Ejection 
6.4.5 Tableting Machines 
6.4.5.1 Single - Punch Tableting Machines 
6.4.5.2 Rotary Tableting Machines 
6.4.5.3 Application of Tableting Machines 
6.4.6 Tableting Machine Simulators (Compaction Simulators) 
6.4.6.1 Hydraulic Compaction Simulators 
6.4.6.2 Mechanic Compaction Simulators 
6.4.6.3 Application of Tableting Machine Simulators 
6.4.7 Instrumentation of Tableting Machines 
6.4.7.1 Force Measurement 
6.4.7.2 Displacement Measurement 
6.4.7.3 Temperature Measurement 
6.4.7.4 Measurement of Time 
6.4.8 Analysis of Tableting Process 
6.4.8.1 Force – Time Analysis 
6.4.8.2 Displacement – Time Analysis 
6.4.8.3 Force – Displacement Analysis 
6.4.8.4 Force – Displacement – Time Analysis 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

1054 TABLET PRODUCTION SYSTEMS 
6.4.9 Analysis of Final Tablet Formation 
6.4.10 Complete Description of Process of Tablet Formation 
6.4.11 Special Accessories of Tableting Machines 
6.4.11.1 Optimization of Die Filling 
6.4.11.2 Tablet Weight Control 
6.4.11.3 Control of Mixing Homogeneity 
6.4.11.4 Cleaning 
6.4.12 Important Factors during Manufacturing Process 
6.4.12.1 Climatization 
6.4.12.2 Lubrication 
6.4.12.3 Occurring Problems during Manufacturing 
6.4.13 Future of Tablet Production Systems 
References 
6.4.1 INTRODUCTION 
Tablet production systems can be defi ned as all machines which are able to produce 
tablets. They include tableting machines for production and research as well as 
tableting machine simulators, which are able to mimic the production processes of 
tableting machines of different size and velocity in order to facilitate scale - up. 
Tablets have been produced for more than 150 years. The fi rst tableting machine, 
developed by Brockedon in 1972 [1] , was a manually operated single - punch machine. 
Currently high - speed tableting machines can produce more than a million tablets 
per hour. 
However, the amount of drug in the early steps of development still makes the 
use of small tableting machines necessary. Thus, before fi nal production a scale - up 
from small machines useful for the production of single tablets to high - speed 
machines is necessary. Since this scale - up is still based on “ trial and error ” [2] , 
tableting machine simulators can be used to simulate different steps in the 
scale - up. 
In research tableting machine simulators are often used since they allow precise 
and well - adjustable measurements during tableting with only a small amount of 
material. 
The aim of this chapter is to give an overview of the different techniques used 
for production and research and further to show the possibilities, that instrumentation 
of tablet production systems gives in order to analyze the tableting process. The 
knowledge derived can be used for formulation development as well as to facilitate 
tablet production and scale - up. Applied techniques which are necessary for production 
in a good manufacturing practice (GMP) environment will also be discussed. 
6.4.2 PHYSICS OF TABLET FORMATION 
Tablets can be defi ned as two - phase systems which consist of a solid phase (the 
compressed powder) and the gaseous phase (the air). The solid phase forms a coher

PHYSICS OF TABLET FORMATION 1055 
ent network inside the tablet and a defi ned form with precise outer dimensions 
results. Here we defi ne the process of tablet formation as the transformation of the 
powder, a noncoherent solid phase, into the compact tablet, and this process lasts 
until no further changes are induced by tableting. “ Tableting ” is only part of the 
process of tablet formation. It is defi ned as the process by which the powder is 
transformed into a coherent form, the tablet. In conclusion tablet formation is the 
result of tableting and all the changes induced by tableting [3, 4] . 
6.4.2.1 Tableting Process 
During the tableting, the powder in the die of the tableting machine is transformed 
by the infl uence of lower and upper punches into a coherent form — “ the tablet ” 
(Figure 1 ). During tableting different processes occur which are responsible for the 
cohesion of the tablet [5, 6] . First the particles are pushed together and reoriented 
in the die until they have arranged in the closest packing. This process is followed 
by elastic and/or plastic deformation of the particles, as can be observed by confocal 
laser microscopy [7] . Some materials show brittle fracture. During brittle fracture 
new particles are produced which can again deform or fracture. Under the infl uence 
of the applied forces the particles approach each other up to bonding [8] . The 
bonded particle collectives continue to deform. The mechanism of deformation and 
bonding is dependent on material properties and process conditions. Important 
bonding mechanisms are van der Waals forces, surface fi lms, liquid or solid bridges, 
and mechanical interlocking [9] . 
The nature of the resulting bonds [9, 10] and the extent of particle approach [11] 
determine the cohesion of the fi nal tablet and are responsible for the compactibility 
of the materials [8] . Further, the formation of bonds occurs not only during the 
FIGURE 1 Densifi cation of powder bed: ( a ) particles in die; ( b ) reorientation of particles; 
( c , d ) particle deformation and fracture; ( e ) tablet in die. 
(a) (b) (c) 
(d) (e)

1056 TABLET PRODUCTION SYSTEMS 
compression phase but also during decompression of the tablet [12, 13] . Hence 
Leuenberger and Ineichen [14] describe tableting as a percolation phenomenon. 
Looking closer at the tableting process it becomes clear that the applied forces 
during tableting are not only the result of machine movement. When punches are 
moving in a machine without material, no force can be measured. The force only 
develops when the punches come into contact with the powder bed. The materials 
resist pressure deformation, and while the punches are moving, a counterforce 
builds up which is the measured force at the punches. In conclusion punch forces 
are determined by the material and as a result materials can be characterized by 
the measured punch forces. 
6.4.2.2 Final Formation of Tablet 
After ejection of the tablet from the die of the tableting machine, the tableting 
process is fi nished. However, decompression continues and thus the tablet formation 
process is not yet fi nished. The remaining stress inside the tablet can resolve and 
produce changes in the structure of the tablet. The tablet partially releases the 
energy gained during tableting. The tablet often shows the occurrence of relaxation, 
which is called elastic recovery or relaxation of the tablet [15, 16] . This relaxation 
is primarily axial relaxation and to a small part radial relaxation, as Newton and 
Rowley [17] fi gured out. Hiestand [15] called this event “ compression born 
repulsion. ” 
Van der Voort Maarshalk [18] showed that the axial relaxation of the tablet, that 
is, the change in tablet height, is dependent on the stored energy and hinders the 
formation of bonds. The elastic recovery [19 – 21] can be measured by measuring 
tablet height at different times after tableting. Simultaneously changes in the inner 
structure of the tablet can occur, for example, shifts of the crystal planes [22, 23] . 
The fusion of amorphous regions and the recrystallization of amorphous parts are 
assumed [24 – 26] . These structural changes are induced by the tableting process and 
also become visible when transformation of drugs into other polymorphic forms 
occurs [27 – 29] . However, even during fi nal formation of the tablets, bonds are 
formed [15, 30, 31] , and thus fi nal tablet formation can be defi ned as a valuable part 
of the tablet formation process. 
6.4.3 REQUIREMENTS FOR TABLET PRODUCTION SYSTEMS 
Tablet production systems should be able to produce tablets in a reasonable time 
and without loss of material. Usually they consist of two punches and a die, as 
schematically shown in Figure 1 . The material is placed into the die cavity, which 
is closed on the lower side by the lower punch. A tablet is formed when the powder 
is compressed by the punches as described above. The forces evolving to produce 
a tablet can be up to 80 kN for pharmaceutical purposes [32] ; for the production of 
bigger tablets they are even higher. Usually they range between 10 and 30 kN. Thus 
punches and dies are produced from hardened steel. 
To withstand the evolving forces, the die has to be fi xed tightly in the machine, 
and for that purpose it is accurately fi t in a die table consisting of steel. The die 
table is further friction locked to the machine frame. 

Tablet production systems can be operated manually; however most of the 
systems nowadays operate automatically in order to produce a suffi cient number of 
tablets. Further requirements depend on the type of machine, the production 
process, the operation mode of the machine, and the production rate of the 
machine. 
6.4.4 TABLET MANUFACTURING PROCESS 
The manufacturing process of tablets principally consists of three stages: fi lling the 
die with the powder, compressing the powder, and ejecting the tablet from the 
die. 
6.4.4.1 Filling 
Die fi lling a tableting machine is a volumetric process. In any case the die is fi lled 
to the upper edge of the die and the powder surface fl ushes with the surface of the 
die and the die table. The fi lling depth is determined by the height of the die cavity, 
the fi lling volume is determined by the diameter of the die hole and the fi lling depth. 
Both the diameter of the die hole and the fi lling depth are used to calculate the fi nal 
fi lling volume. 
Three different techniques exist for fi lling the die. The simplest is to use a fi lling 
shoe which moves back and forth over the die and fi lls the die to its upper edge. 
The surface is leveled off by the fi lling shoe. 
Another possibility is a fi lling shoe which does not move. In this case the dies 
and the accessory lower punches move below the fi lling shoe and the accessory 
upper punches move above the fi lling shoe. In this case the fi lling volume ends at 
the upper edge of the die; however, to level off the surface, an additional scraper 
is necessary. In addition, many machines use the technique of lowering the lower 
punch and with it the powder bed before tableting [33] . This is most helpful to avoid 
dusting when the upper punch moves into the die. 
The newest fi lling technology is to fi ll the die by centrifugal forces [34] . The 
material moves through specially shaped radial channels which approach the die 
from the side. In this case the powder volume is determined by the positions of the 
lower and upper punches in the die. However, for centrifugal fi lling an absolute 
must is a free - fl owing powder. This is a great disadvantage for all non - free - fl owing 
powders. 
6.4.4.2 Compression 
The compression event is the central stage of tablet production. Compression not 
only depends on the machine but to a great deal on the material properties of a 
tablet formulation. The principal stages of compression have been described above. 
Principally, from the machine manufacturing side two different possibilities of compression 
exist. Either the lower punch is closing the die from the bottom side and 
the upper punch moves downward for compression or both upper and lower punches 
move simultaneously toward each other and the powder is compressed from both 
sides. In the fi rst case the surface hardness of the tablet is not the same on the upper 
TABLET MANUFACTURING PROCESS 1057

1058 TABLET PRODUCTION SYSTEMS 
and lower sides; in the second case the surface hardness is the same on the upper 
and lower sides. 
As already described, the tablet is not completely formed during compression 
[35, 36] . When the punches leave each other, the tablet relaxes during decompression, 
further relaxing when one punch leaves the tablet and continuing relaxation 
after ejection from the die. The process is called decompression as long as a force 
is measurable. Afterward the process is called elastic recovery or relaxation of the 
tablet. 
6.4.4.3 Ejection 
During ejection usually the lower punch moves upward to eject the tablet from 
the die and the upper punch has already left the die when the process of ejection 
starts. Only one machine is presently on the market which ejects the tablets downward 
at the bottom of the die [34] . After ejection from the die the tablets are 
collected. 
6.4.5 TABLETING MACHINES 
There exist tableting machines which operate in a different manner and can produce 
in between one single tablet and a million tablets per hour . The orientation of the 
particles in the machine and their rearrangement, densifi cation, and deformation 
depend not only on the material but also on the tableting machine used. 
The fi rst machines for the production of tablets were simple hydraulic or manually 
operated presses. Later, eccentric and rotary tableting machines were developed. 
Today, eccentric tableting machines are only used for research, in early 
development, or for special applications. 
The newest development for production is a machine which fi lls the dies by centrifugal 
force [34] . It is a special rotary tableting machine. Another innovation is a 
special machine which operates by ultrasound [37] . 
In the following the most important machine types and their working principles 
will be described. 
6.4.5.1 Single - Punch Tableting Machines 
Single - punch machines were the fi rst tableting machines used at the end of the 
nineteenth century. The upper punch is lowered by a lever arm on the powder bed 
in the die and by reciprocating this procedure single tablets can be produced. 
Another possibility is to lower the upper punch by means of a screw. These manually 
operated tableting machines are no longer used. 
Two other types are still in usage for special purposes. Hydraulically operated 
machines are used to produce tablets, for example, for Fourier transform infrared 
(FTIR) spectroscopy. In this case the upper punch moves hydraulically onto the 
powder bed and the tablet is formed in the die. The other possibility is an eccentric 
tableting machine, which will be described in the following. 

Eccentric Tableting Machines Eccentric tableting machines are still used in research, 
in early development, and for material characterization. They are the machines of 
choice when you want to use a single - punch tableting machine. As other single - punch 
machines, they work with one pair of punches. In principle, they have a mobile upper 
punch and a lower punch which is fi xed during compression. The lower punch only 
moves for ejection of the tablet. The densifi cation process is unilateral. 
An eccentric valve is driven by a motor and this eccentric valve is responsible 
for the movement of the upper punch (Figure 2 ). The eccentric movement determines 
the operation and speed of the upper punch. The upper punch is driven by 
the eccentric valve into the die, which is closed on the bottom side by the lower 
punch, and the tablet is compressed in the die. 
The punch forces for compression evolve due to contact with the powder. Thus 
the measured punch forces at the upper and lower punches result from the movement 
of the upper punch and the resistance of the powder bed toward deformation. 
From that it becomes clear that the measured upper punch force is usually higher 
than the lower punch force, and thus the upper punch force is the main force used 
for material characterization. The tablets show different hardness on upper and 
lower surfaces, as do all tablets produced by single - punch machines [38] . 
When the upper punch is lifted from the tablet surface, the lower punch moves 
the tablet upward for ejection out of the die . However, as is known nowadays, the 
lower punch also moves slightly before due to evolving forces. 
In eccentric machines, die fi lling is performed by a fi lling shoe which moves back 
and forth above the die. When the upper punch moves upward above the die, the 
die is fi lled, and when the upper punch starts downward movement, the powder bed 
is leveled off. After ejection of the tablet the fi lling shoe pushes the tablet down 
from the die table. During this fi lling process with a moving fi lling shoe, demixing 
of the product mixture is more easily possible than with a fi xed fi lling shoe. This is 
one major disadvantage of eccentric tableting machines. 
The production rate varies usually between 10 and 60 tablets/min and is determined 
by the number of eccentric movements. An example of an eccentric tableting 
machine is given in Figure 3 . Other machines are described by Ritschel and Bauer - 
Brandl [32] . 
FIGURE 2 Operation of eccentric tableting machine: ( a ) fi lling; ( b ) compression; ( c ) ejection; 
( d ) pushing from die table. 
(a) (b) (c) (d) 
TABLETING MACHINES 1059

1060 TABLET PRODUCTION SYSTEMS 
6.4.5.2 Rotary Tableting Machines 
Rotary tableting machines are commonly used for tablet production. The principle 
of all rotary machines is the same, with one exception, which will be discussed separately. 
According to Konkel and Mielck [39] , the information gained with eccentric 
and rotary machines complement each other. 
Rotary tableting machines work with a number of punch and die sets which move 
in a circle. The dies are fi xed in a round die table and the die table circulates. 
Together with the dies the lower and upper punches circulate on tracks. The lower 
punches close the dies. The densifi cation process is bilateral since both punches pass 
the compression wheels and the force is evolving on the upper as well as on the 
lower side of the powder bed. The produced tablets show the same hardness on the 
upper and lower surfaces. 
There are different stages of tablet production which happen simultaneously for 
several tablets. The central stage of tableting occurs when the punches pass the 
upper and lower compression wheels and the compression wheels determine the 
downward movement of the upper punch and the upward movement of the lower 
punch: The tablet is formed by the resulting punch forces. The compression wheels 
can be positioned either by a fl exible swing or more seldom by an eccentric valve 
[40] . 
FIGURE 3 Example eccentric tableting machine. (Courtesy of Korsch XP1.) 

In Figure 4 this principal stage and the other stages of tablet production are 
visible. Except for the compression stage the upper punch is always in an upward 
position. The upper punch is above the die and the lower punch closes the die, 
determining the fi lling depth; then the dies pass below the fi lling shoe and the 
powder fl ows into the die up to its upper edge where the powder bed is leveled off 
by a scraper. Whereas the upper punch is still in an upward position, the lower 
punch is lowered slightly on the track to keep the whole powder volume during 
compression and to prevent dusting [33] . Now the punches start to pass the compression 
wheels and the main compression event occurs. Forced by the wheels, both 
the upper and lower punches move toward each other, compress the tablet, and 
leave each other again. The upper punch lifts into an upward position and the lower 
punch moves upward to eject the tablet. After ejection the tablet passes a scraper 
and it is pushed down from the die table. An example of a rotary machine is given 
in Figure 5 . 
Additionally often rotary machines are equipped with precompression wheels 
(Figure 4 ). In this case the punches pass the precompression wheels before the main 
compression events starts. Precompression has the same stages of compression as 
the main compression, but the applied forces are lower. Precompression is deemed 
to be helpful to avoid, for example, dusting, capping, or lamination. After precompression 
only a lower main compression force is necessary. 
The number of dies and complementary punches of a rotary machine can vary 
between one and up to hundred. The number of punches and the rotation speed 
FIGURE 4 Operation of rotary tableting machine with precompression. (Courtesy of 
Fette.) 
TABLETING MACHINES 1061

1062 TABLET PRODUCTION SYSTEMS 
of the die table determine tablet production. Fast rotary machines have up to 
120 rpm of the die table [32] . However, the number of tablets produced per hour 
not only is determined by the number of punches and the rotation speed but also 
is limited by the deformation properties of the tableted material. Materials need 
some time for deformation, and if the time during one compaction cycle is not suf- 
fi cient for compression and compaction of the material, no tablets result from the 
process. 
In order to increase the production rate of rotary tableting machines, double - 
sided rotary machines were build which possess two pairs of compression wheels 
and two fi lling shoes. Thus during one rotation double the number of tablets are 
produced compared with a one - sided rotary machine. If these machines are equipped 
with precompression wheels one machine contains four pairs of wheels. 
Special Rotary Machines As already mentioned one special rotary machine works 
slightly different — called IMA Comprima (Figure 6 ) [34] . In this machine the material 
is fi lled by centrifugal force from the side directly into the die. The upper punch 
closes the die at the top and the lower punch closes the die at the bottom. However, 
when the given volume in the die is fi lled by the powder, both punches move downward 
until the die is completely closed. Then the compression process starts and the 
dies pass the compression wheels. After compression the tablet is ejected by the 
upper punch at the bottom of the die, contrary to all other rotary tableting machines 
which eject the tablet at the top of the die. 
High-Speed Rotary Tableting Machines High - speed rotary machines work with 
the same principles as all other rotary machines. They possess a huge number of 
punch and die sets and often two fi lling stations. Another possibility is to use punch 
and die sets which are able to produce several tablets simultaneously. Special tooling 
can be used for this purpose; however this is not the subject of this chapter. As 
FIGURE 5 Example rotary tableting machine: left, machine view; right, detail view into 
compression chamber. (Courtesy of Kilian Synthesis 500.) 

already mentioned production speed depends to a great extent on product properties. 
Excellent powder fl ow is essential since the dies have to be fi lled completely. 
This is also most essential for the IMA Comprima, which works with centrifugal 
force for die fi lling. Often this machine cannot be used since powder fl ow properties 
are not suffi cient. To improve powder fl ow on conventional rotary machines special 
fi lling devices have been developed (Section 6.4.11.1 ) . 
6.4.5.3 Application of Tableting Machines 
In summary, the single - punch tableting machines still being used are mainly eccentric 
tableting machines (mostly for research), whereas rotary machines with different 
production output are predominantly used for production; and for rotary 
machines in most cases it is not machine speed that determines the production rate 
but material fl ow and compression properties. 
6.4.6 TABLETING MACHINE SIMULATORS 
(COMPACTION SIMULATORS) 
Tableting machine simulators [41 – 48] have been developed in order to mimic tablet 
production systems with a very small amount of powder. Similar to eccentric tableting 
machines, tableting machine simulators use one pair of punches. Working only 
with a single pair of punches reduces the consumption of tableting materials and 
facilitates instrumentation for displacement measurement. 
6.4.6.1 Hydraulic Compaction Simulators 
The fi rst compaction simulators developed were hydraulic [41 – 45] . The hydraulic 
system is electronically controlled. An example is given in Figure 7 . Either compression 
force cycles or movement of the punches was freely adjustable. This allowed 
FIGURE 6 Working principle of Comprima tableting machine with centrifugal fi lling: left, 
operation mode; right, centrifugal fi lling. (Courtesy of IMA Comprima.) 
TABLETING MACHINE SIMULATORS (COMPACTION SIMULATORS) 1063

1064 TABLET PRODUCTION SYSTEMS 
much variation and the primary aim was to mimic the densifi cation process of a 
rotary machine and the mechanical factors infl uencing it. 
For example, theoretically, from machine geometries, the force – time profi le of a 
rotary tableting machine [49 – 52] can be deduced and calculated and the data are 
programmed into the compaction simulator. However, the force – time profi le of a 
tableting machine could not be calculated. Too many factors infl uence the measured 
force, for example, the tableted material, the geometries of the machine, the machine 
wear time, tableting speed, and tableting tools. Similarly, the displacement – time 
profi le of a tableting machine, especially a rotary tableting machine, is very diffi cult 
to calculate. It has been shown that calculation from machine geometries is only 
possible to a certain extent. Mainly the mechanics of a tableting machine cannot be 
completely simulated [53, 54] . Thus either an approximated displacement – time 
profi le can be used for programming the compaction simulator or approximation 
of real punch movement is only possible using recorded data from real tableting 
machines. 
Thus the simulation of tableting machines needs much effort and a real simulation 
is almost impossible because of the hydraulic control. Further the fi lling process 
of rotary tableting machines cannot be simulated since die fi lling is usually processed 
by a fi lling shoe moving forth and back. 
However, hydraulical compaction simulators are still used in research for basic 
material characterization. They show the advantage of controlling speed exactly and 
of using low and high punch travel speeds, between 10 and 300 mm/s. Mostly a simple 
displacement profi le is used for characterization (e.g., a saw tooth or a sine wave 
profi le), and the evolving forces at the lower and upper punches are measured. 
Further the speed of the punches can be controlled separately and both punches 
move freely and independently from each other. Time intervals in which the punches 
stand still can be freely set. Thus lots of freedom for material characterization is 
possible and these compaction simulators are important tools. 
Another advantage of compaction simulators is that only small amounts of material 
are necessary to produce a tablet. One single tablet can be produced at low as 
well as at high speed of the punches. This is important in order to evaluate defor- 
FIGURE 7 Example hydraulically working compaction simulator: left, machine view; right, 
detail view into compression chamber. (ESH compaction simulator, Courtesy of Huxley 
Bertram.)

mation properties of a formulation already in early dosage form development when 
only small quantities of the drug substance are available. Since nowadays the timelines 
for production of a new medicine are tight, this advantage of compaction 
simulators becomes more and more important. 
6.4.6.2 Mechanical Compaction Simulators 
More recently mechanical compaction simulators have been developed. The fi rst 
was the linear mechanical rotary tableting machine simulator Presster (Figure 8 ), 
which was introduced in 1998 [55, 56] . It can mimic the mechanics of different rotary 
tableting machines and is called a linear rotary tableting machine replicator. The 
name Presster was combined from press and tester. 
A single pair of punches moves linearly forth and back on a lower and an upper 
punch track. For tableting the punches pass the compression wheels which are 
equivalent in dimensions to those of rotary tableting machines used in practice. 
These compression wheels are exchangeable. Different machines are simulated by 
exchanging them. 
The machine speed can be varied and different tableting machines are simulated 
by using similar dwell times between 5 and 80 ms. Special tests exhibited that rotary 
tableting machines can be simulated with a precision of 1 – 5% [57, 58] . One major 
disadvantage of the Presster is that it works with a moving fi lling shoe, and thus the 
fi lling process of rotary machine cannot be simulated. 
In addition, the present model of the Presster possesses precompression wheels 
and thus, besides compression, precompression can be simulated. This is import 
FIGURE 8 Detail of Presster. (Courtesy of MCC Corp.) 
TABLETING MACHINE SIMULATORS (COMPACTION SIMULATORS) 1065

1066 TABLET PRODUCTION SYSTEMS 
when studying the effect of precompression on the fi nal tablet properties. However, 
the time between precompression and main compression is determined by the 
Presster geometries since the positions of the precompression and main compression 
wheels are fi xed. 
The newest development for compaction simulation is a mechanical tableting 
machine simulator which operates with a cam. Thus it is called Stylcam (Figure 9 ) 
[59] . The cam is positioned on the lower compression wheel and allows the simulation 
of different tableting machines and their dwell times due to different acceleration 
of the punches. It was introduced in 2005. 
With the Stylcam different dwell times are obtained by adjusting the speed of 
the compression wheels. Precompression is simulated by compressing a tablet twice. 
Thus the time interval between the precompression and main compression is freely 
adjustable. One further advantage of the Stylcam is that it works with a fi xed fi lling 
shoe, as on a conventional rotary tableting machine. However, data on the precision 
of this instrument are not yet available. 
6.4.6.3 Application of Tableting Machine Simulators 
Using mechanical compaction simulators allows us to simulate the tableting process 
of rotary tableting machines to a greater extent than when using hydraulical compaction 
simulators. Thus they will be mainly used in formulation development and 
scale - up. 
FIGURE 9 Detail of Stylcam. (Courtesy of MedelPharm.) 

However, for mechanical compaction simulators the movement of the punches 
is mechanically determined and, compared to hydraulic compaction simulators, not 
freely programmable. Thus, for basic material characterization and early formulation 
development, hydraulical compaction simulators can be advantageous. 
6.4.7 INSTRUMENTATION OF TABLETING MACHINES 
To describe the tableting process more precisely, tableting machines have been 
instrumented since the middle of the last century. Measured values are force, displacement, 
and temperature and they are always measured with dependence on 
time. Thus time is another variable. 
6.4.7.1 Force Measurement 
The fi rst instrumentation of a tableting machine for measurement of upper punch 
force was performed by Brake [60] . Thus it was for the fi rst time possible to visualize 
the compression process with regard to force development which results from 
the material stresses during tablet formation. Only shortly after that, similar measurements 
were published by another research group [61, 62] . 
Besides upper punch force, lower punch force, die wall force [63 – 65] , ejection 
force [66] , and tablet scraper force can be measured. Die wall force measurement 
will be discussed separately. 
For measurement strain gages are mostly used. These strain gages consist of 
constantan. They are applied in eccentric tableting machines at the upper or lower 
punch holder and in rotary tableting machines at the machine frame or the compression 
roll pin. Alternatively piezoelectric crystals can be used which have to be placed 
inside the punches [40] . 
The sensitivity of force measurement is dependent on the distance between the 
force transducer and where the force occurs. Thus for force measurement, instrumentation 
of the punches is more advantageous than instrumentation of the machine 
frame. However, since punch and die sets have to be exchanged between different 
runs of the machines, instrumentation of the punch holder, the machine frame, or 
the roller pin is most widely spread. 
The most often measured force is the upper punch force. For the eccentric 
machine it is the force which controls densifi cation; for rotary tableting machines 
upper and lower punch forces have ideally the same values. Schmidt et al. [67] 
measured force with a single punch of a rotary tableting machine. Ejection force is 
visible as a small lower punch signal which occurs shortly after the end of one compaction 
cycle. It is measured by lower punch instrumentation but needs more resolution. 
A review of force measurement is given by Bauer - Brandl [68] . 
Die Wall Force Measurement During compression of the powder the forces are 
evolving not only at the punches but also at the die wall [63 – 65] . Therefore die wall 
force measurement complements upper and lower force measurement. Since the 
compression process occurs axially, these radially evolving forces are smaller than 
the forces at the punches. Measurement of die wall force allows, for example, for 
indication on die wall friction, tablet capping, and lamination. Instrumentation for 
INSTRUMENTATION OF TABLETING MACHINES 1067

1068 TABLET PRODUCTION SYSTEMS 
die wall force measurement is diffi cult and different techniques have been developed 
[64, 69, 70] . The die can be instrumented axially or radially and strain gages 
or piezoelectric crystals can be used for measurement. Two main effects infl uence 
the measured signal: tablet height and tablet position. Related to this, the output 
signal can be nonlinear. Piezoelectric foils have been applied which possess the 
advantage of independence on tablet position [70] . 
One example most recently developed is a split die consisting of three sections 
(Figure 10 ) [64] . Integrating the sensing web in a thin middle layer isolates stress 
measurement to a narrow band around the tablet and gives much closer approximation 
to the true stress. Further die wall force measurement is linear and independent 
of tablet height and position as it is uncoupled from all other die wall stresses and 
strains. Further it is designed in the shape of a conventional die and can be mounted 
without modifi cation into a die table. 
6.4.7.2 Displacement Measurement 
The fi rst measurement of upper punch displacement was performed in the mid - 
1950s by Higuchi and co - workers [62, 71] with the aid of inductive displacement 
transducers. By the same instrumentation the movement of the lower punch can be 
visualized. Inductive transducers are mounted parallel to the punch and thus give 
information on punch position. Alternatively touchless measurement of displacement 
is possible. It is important that the transducers be positioned most closely to 
the punch in order to minimize the infl uence of machine deformation. 
A measurement of displacement on a rotary tableting machine was presented in 
1987 by Schmidt and Tenter [72] . Another possibility was presented by Matz and 
co - workes [73, 74] . Meanwhile touchless measurement systems for recording displacement 
were developed [73, 74] . For all measurements of displacement, correc- 
FIGURE 10 Construction details of split die. (Reproduced with permission from ref. 64.) 
dp dp 
ds dt 
wh 
wt 
Strain gauge 
dt 
do 
di: inner diameter 
do: outer diameter 
ds: screw hole diameter 
dp: alignment pin hole diameter 
wa: web height 
wt: web thickness 
= in. 
= in. 
= in. 
= in. 
= in. 
= in., in. 
38
785
32 
18181
16 
18

tions which take elastic punch and machine deformation into account are necessary. 
M u ller and Caspar [75] showed problems which occur when machine and punch 
deformation are not taken into account. Krumme and co - workers gave an extensive 
description on this issue for eccentric tableting machines [76, 77] . From punch displacement 
measurements tablet height can be calculated, and this height can further 
be related to tablet density and porosity. 
Again and again it was tried to derive displacement theoretically from machine 
geometry, punch geometry, and measured force [49 – 52] . However, until today this 
theoretical derivation has not been satisfying and thus experimental testing cannot 
be given up. 
6.4.7.3 Temperature Measurement 
Due to the forces evolving during tableting the compressed material can warm up. 
The evolving temperatures can be measured with different methods. In each case 
only approximated measurements are possible since either the measurement was 
not performed directly inside the tablet or additives were necessary, which can alter 
measurement. 
Several methods have been applied to determine the temperature increase with 
thermal sensors which are installed in the punches, inside the die, or in the powder 
bed (Table 1 ) [78 – 87] . 
Most recently an analysis technique was developed which allows measurement 
of tablet temperature directly after ejection of the tablet on the machine (Figure 
11 ) by an infrared sensor [85] . The temperature signal can be directly related to 
force and displacement measurement. 
6.4.7.4 Measurement of Time 
All measured variables can be determined with dependence on time. Thus the 
tableting process can be characterized for several variables with dependence on 
time. 
Two time defi nitions are important: contact time and dwell time. Contact time 
can be defi ned as the time during which a contact of powder and punches is measurable, 
for example, when the force exceeds a certain limit of 100 N. Dwell time can 
be defi ned predominantly for rotary machines as the time during which the punch 
heads are completely under the compression wheels and thus the applied force is 
constant. 
TABLE 1 Methods to Determine Temperature During and Shortly After Compaction 
Measurement in punches [78 – 80] 
Epoxide punches [81] 
Calorimetric measurement [82, 83] 
Infrared measurement [84, 85] 
Measuring conductivity during tableting with conductive materials [86] 
Energy calculations [78, 83, 84] 
Determination of melting of materials with certain melting point [87] 
INSTRUMENTATION OF TABLETING MACHINES 1069

1070 TABLET PRODUCTION SYSTEMS 
Further for description of the tableting process, the entering of the punch into 
the die, the compression start at the begin of contact time, and the lifting of the 
upper punch from the tablet are important. 
6.4.8 ANALYSIS OF TABLETING PROCESS 
All measured variables, namely force, time, displacement, and temperature, can be 
combined differently and can be analyzed afterward. From the functional relations, 
conclusions can be drawn about the compression and compaction behavior of the 
materials. 
The most basic analysis is the presentation of force versus time or displacement 
versus time. These curves are different for eccentric and rotary tableting machines. 
The data given in Figures 12 and 13 are valid for the contact time of the compaction 
cycle of one single tablet. Due to the eccentric - driven movement of the punches, 
the force – time curve can be described by a sharp peak at the maximum force evolving 
at the punches and the displacement – time curve can be described with a sharp 
peak at the minimum height of the powder bed. For curves of eccentric tableting 
FIGURE 11 Infrared sensor unit for measuring temperature directly after tableting [85] 
(Martin - Luther - University Halle - Wittenberg.) 
Upper 
punch 
Die 
holder 
IR sensor 
Tablet 
FIGURE 12 Force – time and displacement – time profi le for eccentric tableting machine. 
Force (kN) 
Time (ms) Time (ms) 
Displacement (mm) 
(a) (b) 

machines hardly any dwell time is measurable (Figure 12 ). In contrast, for rotary 
tableting machines, the force – time curve and the displacement – time curve are 
fl atter at the maximum peak. This is the case due to the dwell time when the punch 
heads move completely between the compression wheels. The dwell time is indicated 
in Figure 13 . For rotary tableting machines with precompression wheels 
additionally force – time curves and displacement – time curves for precompression 
can be recorded. They look similar to the curves from the main compression wheels 
with the exception that lower forces are applied. Precompression data will not be 
discussed in the following since the data can be treated similar to the data of the 
main compression event. 
Besides this presentation of force and displacement versus time, which are data 
directly derived from the tableting machine, other more advanced methods are 
possible. Extensive reviews on the methods used can be found in the literature 
[54, 88, 89] . In the following only the most important aspects will be discussed. 
6.4.8.1 Force – Time Analysis 
One method to analyze tableting data is the use of force – time or pressure – time 
diagrams. They are easily recordable since displacement measurement is not 
necessary. 
Some basic parameters can be directly read from the curves. For the force values 
upper and lower punch forces and ejection forces should be mentioned, and for the 
time values contact time should be mentioned. Deduced parameters such as pressure 
and normalized contact time can be calculated and further statistical data are 
often used for characterization (Table 2 ). Due to the different shapes of force – time 
curves from eccentric tableting machines compared with those from rotary tableting 
machines, some parameters can only be calculated from eccentric machine data and 
some can only be calculated from rotary machine data. 
FIGURE 13 Force – time and displacement – time profi le for rotary tableting machine. 
Force (kN) 
Displacement (mm) 
Time (ms) Time (ms) 
(a) (b) 
ANALYSIS OF TABLETING PROCESS 1071

1072 TABLET PRODUCTION SYSTEMS 
Only eccentric machine data allow us to calculate the R value (maximum upper 
punch force/maximum lower punch force), which is an indication of friction. They 
also allow us to calculate the time difference between the maximum upper punch 
force and the maximum lower punch force. Only dwell time and the minimum force 
during the dwell time can be calculated for rotary tableting machine data. The rise 
time of rotary machines is defi ned as the time during the compression phase, and 
peak offset time is defi ned as the time difference between maximum pressure and 
vertical alignment of the punches. Further the infl ection points during the compression 
and decompression phases are mostly only calculated for rotary machine 
data. 
In addition, for force – time diagrams different methods to characterize the tableting 
process were developed. These methods can be divided in those applicable to 
force – time curves from eccentric and rotary tableting machines [90] and those 
applicable only to data from eccentric or rotary tableting machines (Tables 3 – 5 ). 
One possibility to analyze the tableting process is to describe the areas under the 
curve during compression and decompression and to draw conclusions on plastic 
and elastic parts of deformation. Emschermann and M u ller [91] applied this method 
to data from eccentric machines (Figure 14 ). Similar area comparisons were performed 
by the research group of Schmidt [92 – 94] for rotary machines (Figure 15 ). 
They tried to gain information on elasticity by calculating differences between the 
area under the plot in the compression phase and the area under the plot in the 
decompression phase. A sophisticated technique to interpret area data under one 
TABLE 2 Parameters Directly Deduced from Force – Time Profi les 
Maximum upper punch force (pressure) 
Maximum lower punch force (pressure) 
Maximum ejection force (pressure) 
Contact time 
Normalized contact time 
Time at maximum upper compression force 
Time at maximum lower compression force 
Maximum upper precompression force (pressure) 
Maximum lower precompression force (pressure) 
Precompression contact time 
Normalized precompression contact time 
Time at maximum upper precompression force 
Time at maximum lower precompression force 
TABLE 3 Parameters Calculated for Eccentric and Rotary Tableting Machines 
Source Parameters 
Chilamkurti [90] Area under curve (AUC) 
Height of curve at maximum upper punch force 
Width of curve at half - maximum upper punch force 
Slope during compression 
Slope during decompression 

TABLE 4 Parameters Calculated for Eccentric Tableting Machines 
Source Parameters 
Emschermann [91] 
(Figure 14 ) 
Area under compression curve (compression area) 
Area under decompression curve (decompression area) 
Compression area/decompression area 
Pressure – time function 
(modifi ed Weibull 
function) [39, 96 – 98] 
(Figure 16 ) 
p p 
t t = 
.
. .
max, exp upper punch 
1 
1 
1 
. . 
. . 
where . = time parameter 
. = asymmetry parameter 
Modifi ed Fraser – Suzuki 
function [99] 
f t H 
A 
A t t 
S 
( ) exp 
. 
ln 
( ) 
. = . ... 
... 
. + . . ( ). ... 
... 
... 
... 
0 693 1 
1 177 2 
2 
r 
where A = asymmetry parameter 
t r = time parameter 
S = deviation of maximum 
TABLE 5 Parameters Calculated for Rotary Tableting Machines 
Source Parameters 
Tenter [72] Area center of gravity 
Vogel [92 – 94] (Figure 15 ) t 1 = compression time 
t 2 = time at the start of dwell time 
t 3 = time at the half of dwell time 
t 4 = time at the end of dwell time 
t 5 = time at the end of one compression cycle 
A 1 = area of densifi cation 
A 4 = area of decompression 
A 2 = partial area 
A 3 = partial area 
A 3 / A 2 = area quotient 
A 5 = partial area 
A 6 = partial area 
A 6 / A 5 = area quotient 
FIGURE 14 Pressure – time curve analysis [91] . 
Pressure 
Time 
1073

1074 TABLET PRODUCTION SYSTEMS 
compaction cycle was developed. The parameters of interest are given in Table 5 . 
Further advances were performed by Yliruusi and co - workers [95] . 
Another possibility of analysis is to fi t different functions to the force – time data. 
The research group of Mielck [39, 96 – 98] described the densifi cation behavior by 
the pressure – time function (Figure 16 ). The lower the values of the parameters . 
and . , the more plastically the material deforms; the higher the values, the more 
elastically the material deforms. The parameter . describes the asymmetry of the 
curve and . the time at maximum densifi cation. A similar function, the Fraser – 
Suzuki function, which originates from chemical analytics was applied to tableting 
data [99] . It can also be used to derive parameters that describe the deformation 
behavior of materials. Information on the reversible and irreversible deformation 
of the material can be deduced. 
FIGURE 15 Pressure – time curve analysis [92, 93] . 
Pressure 
A5 A6 
A1 A2 A3 A4 
t2 t1 t3 t4 t5 
Time 
FIGURE 16 Pressure – time curve analysis with pressure – time function [39, 96 – 98] . 
Time 
Pressure

6.4.8.2 Displacement – Time Analysis 
From displacement – time data a few parameters can be read. The most important 
data are given in Table 6 . In addition the data allow us to calculate fast elastic 
recovery. The increase in tablet height from the minimum tablet height in the die 
up to the lifting of the upper punch from the tablet is called fast elastic recovery. 
Since the travel of the punches is analyzed, the data also allow us to determine 
the speed of the punches at each point of the compaction cycle. Punch speed is an 
important parameter to compare different tableting events [100] . Maximum punch 
speed can be determined and used for characterization. 
Only a few authors have tried to relate displacement with time. Ho and Jones 
[101] determined the slope of porosity over time (rise time). This slope was also 
used by Tsardaka [102] for analysis. 
6.4.8.3 Force – Displacement Analysis 
The most extensively used method to characterize the tableting process is the use 
of force and displacement measurements. Usually upper punch force and upper 
punch displacement are used. Models which relate force and displacement directly 
can be distinguished from those which analyze pressure and volume. 
Further, some parameters can be directly read from the curves. The peak offset 
time of eccentric tableting machines is defi ned as the time difference between 
maximum displacement and maximum compression force [101, 103] . 
Relation between Force and Displacement The fi rst information on force – 
displacement analysis can be found in F u hrer [104] and Moldenhauer et al. [105] . 
Force – displacement diagrams (Figure 17 ) are used to calculate from the areas 
enclosed the work or energy necessary for tableting. The force – displacement profi le 
includes compression and decompression of the powder to the tablet. The area 
between compression and decompression is the area of the compaction energy, 
often called the energy of plastic deformation ( EP ) [106] . The area EE is the energy 
of elastic deformation. And the last area ( EF ) to complete the triangle start of compression 
( D0 ) – maximum force ( Fmax ) – displacement at maximum pressure ( DPM ) can 
be interpreted as the energy of friction ( EF ) [107, 108] . The sum of all three energies 
is the total energy ET of the tableting machine. The energies can be displaced as an 
absolute value or relative to the total energy. Based on these values, Stamm and 
Mathis [109] developed the determination of plasticity P as 
TABLE 6 Parameters Directly Deduced from 
Displacement – Time Profi les 
Maximum displacement 
Minimum height of tablet 
Minimum volume of tablet 
Maximum density of tablet 
Minimum porosity of tablet 
Maximum relative density of tablet 
Time at maximum displacement 
Time at minimum height of tablet 
ANALYSIS OF TABLETING PROCESS 1075

1076 TABLET PRODUCTION SYSTEMS 
P 
E 
E E 
= 
+
P 
P E 
(1) 
Other authors tried similar attempts [110, 111] and the developed methods were 
regarded to be very useful [27, 112 – 115] . Antikainen and Yliruusi [116] more 
recently tried to derive further parameters from the diagrams to enable a more 
complete characterization. An overview on the possibilities for force – displacement 
analysis is given by Ragnarson [117] . 
Relation between Volume and Pressure The oldest method of this type of analysis 
is to establish a relation between the volume of the tablet and the force necessary 
to produce this volume [62, 71] . For exact description the height of the tablet is 
determined by displacement measurement and the accuracy of this measurement is 
extremely important. Further displacement measurement has to be corrected precisely 
for elastic deformation of the punches and the machine in order to use correct 
tablet heights. 
From tablet height, volume, porosity, and relative density at different stages of 
densifi cation can be deduced. These variables are plotted as a function of pressure. 
For analysis, for example, the equations of Heckel [118 – 120] , Kawakita, [121, 122] , 
Cooper and Eaton [123] , Walker [124] , Bal ’ shin [125] , and S o nnergaard [126, 127] 
can be used. The equations are given in Table 7 . A further overview of these and 
other equations used can be found in Celik [88] . 
The Heckel equation describes the densifi cation process with fi rst - order kinetics. 
A linear equation is obtained with a slope which is inversely proportional to the 
yield strength. The slope of the Heckel equation provides information on the plastic 
deformation of the powder. It has also been published that the slope of the Heckel 
equation can be correlated with the elastic modulus (Young ’ s modulus). 
FIGURE 17 Force – displacement diagram for energy analysis [104 – 108] . 
Force (kN) 
Displacement (mm) 
D0 DPM 
EP 
EF 
Fmax 
EE 

TABLE 7 Parameters Calculated from Force – Time Profi les 
Source Parameters 
Heckel [118, 119] (Figure 18 ) 
. = 
. ( )= + ln ln . 1 
1 D 
Kp A 
where K = deformation parameter 
A = powder bed densifi cation 
Yield pressure [120] 
Yield pressure 
Heckel slope 
= 1 
Yield strength [118] 
Yield strength 
Heckel slope 
= 
. 
1 
3 
Kawakita [121, 122] 
p
C ab a p
= + 1 1 
where a = porosity of powder bed 
b = compression parameter 
Walker/Balshin [124, 125] 
100 100 V 
V 
V 
W p C rel= . =. + 
. 
log 
where W = compressibility coeffi cient 
S o nnergard [126] 
V V W p Vee 
p pm 
rel 
/ = . + . 
1 log 
where W = compressibility coeffi cient 
Cooper – Eaton [123] 
V aV 
V V 
V V 
a e a e i i 
i 
n 
k p k p * * / / = = . 
. 
= + 
= . 
. . .1 
0 
0 
1 2 
1 2 
where k 1 = deformation pressure for fraction part 1 
k 2 = deformation pressure for fraction part 2 
a 1 = fraction part 1 of deformation 
a 2 = fraction part 2 of deformation 
Cooper – Eaton (linearized) [123] 
ln ln ln V 
V V 
V V 
Q
p 
R * = . 
. 
= . + 
. 
0 
0 
where Q = extent of compressibility 
R = sum of fraction parts 
The equation of Kawakita describes volume reduction with pressure in the form 
of a hyperbolic equation. Walker and Bal ’ shin [125] postulated a logarithmic relation 
between applied pressure and volume reduction, which was further modifi ed 
by S o nnergard [126] . Cooper and Eaton [123] use an exponential function, which 
can also be linearized. Pressure thresholds for deformation mechanisms are determined. 
It should be noted that all of these equations and tableting models determine 
descriptive parameters. 
The equation of Heckel is the most extensively used model and the underlying 
porosity – pressure plot is called a Heckel plot (Figure 18 ). The equation for 
the linear compression process follows fi st - order kinetics (Table 7 ). Heckel 
ANALYSIS OF TABLETING PROCESS 1077

1078 TABLET PRODUCTION SYSTEMS 
distinguished measurements which determine the volume of the tablet without pressure 
(zero pressure) [119] from those measurements which determine the volume 
with pressure [118] . The fi rst method allows determination of the volume after 
release of the elastic energy; the second method allows a higher precision repeatability 
since it is often diffi cult to determine tablet height at a defi ned time after 
ejection. Some milliseconds can cause differences [35, 36] . Years later Sun and 
Grant [128] tried to determine the elastic part at pressure measurements. The 
experiments showed that deviations in Heckel plots at high pressures are dependent 
on the elasticity of the material. 
The equation of Heckel has been discussed again and again. One main issue of 
critique is that pharmaceutical powders are not purely plastically deforming materials 
and thus particle size and deformation mechanisms infl uence the derived parameters 
[129, 130] . Already very small errors in displacement determination or the 
measurement of true density can induce huge errors in the derived parameters 
[75 – 77, 129, 131, 132] . S o nnergaard [126] referred the equation of Walker and 
Bal ’ shin for his characterization of materials. He criticized further that the yield 
strength derived from the Heckel equation is directly dependent on the true density 
of the powders [127] . 
Despite this critique of the Heckel equation, the analysis of Heckel plots has 
been intensively used for the description of powder compression [128, 133 – 136] . 
Gabaude et al. [136] stated that the analysis is quite useful when defi ning preconditions 
exactly and apply correct displacement measurement. 
Since the development of the equation, it has been tried to derive further information 
from it. Rees and Rue [129] determined the area under the Heckel plot. 
Duberg and Nystr o m [137] used the nonlinear part for characterization of particle 
fracture. Paronen [138] deduced elastic deformation from the appearance of the 
Heckel plot during decompression. Morris and Schwartz [139] analyzed different 
phases of the Heckel plot. Imbert et al. [134] used, in analogy to Leuenberger and 
Ineichen [14] , percolation theory for the compression process as described by 
the Heckel equation. Based on the Heckel equation, Kuentz and Leuenberger 
[135, 140] developed a new derived equation for the pressure sensitivity of tablets. 
FIGURE 18 Heckel plot [118, 119] . 
Pressure (MPa) 
In(1/1 - Drel) 
3.0 
2.5 
2.0 
1.5 
1.0 
0.5 
0.0
0 50 100

Tsardaka and co - workers [102, 141, 142] presented the Heckel plot with dependence 
on time and analyzed deformation in combination with elastic recovery. Additional 
areas to describe plasticity were determined from two - dimensional (2D) plots [129] . 
Finally, the three - dimensional (3D) model [143, 144] was developed by fi tting a 
plane to a 3D data plot on the basis of normalized time, pressure, and porosity 
according to Heckel. 
6.4.8.4 Force – Displacement – Time Analysis 
Force, displacement, and time are the three most important parameters to characterize 
the compaction cycles of tableting materials. Even when Hoblitzell 
and Rhodes postulated a linear relationship between force – time and force – 
displacement data [145] , this could not been exactly proved until today. Thus it is 
im portant to analyze these three measured data together. 
From force – displacement – time curves some parameters can be directly deduced; 
for example, the power during tableting and the maximum power can be determined. 
Another important parameter which serves as a measure for viscoelasticity 
is the peak offset time [101, 103] . For single - punch tableting machines it is defi ned 
as the time difference between maximum displacement and maximum force. 
In addition, advanced models as those calculating viscoelasticity and the 3D 
model have been developed. They will be described in the following. 
Viscoelasticity Models For characterization with viscoelasticity models, simulation 
models have been developed on the basis of Kelvin, Maxwell, and Voigt elements. 
These elements come from continuum mechanics and can be used to describe 
compression. 
David and Augsburger [146] were the fi rst to try this method of analysis. Further 
tests, for example, determination of complex functions based on methods of numerical 
mathematics, were performed by the research group of M u ller [147, 148] . 
Although the results were helpful, for exact description the models became rather 
complicated and the derived parameters were complex. According to Bauer [149] , 
these models have to be three dimensional for a reasonable description. Another 
similar approach was used by the research group of Rippie [12, 13] . They described 
the structure evolution in the tablet during decompression by the aid of vectorial 
3D models and concluded that fracture and stress contribute to the fi nal structure 
of the tablet. 
Other research groups derived viscoelastic properties from creep experiments of 
the fi nal tablet [150 – 154] . As Tsardaka and Rees [142] determined, stress relaxation 
follows a hyperbolic equation. 
3D Model Most recently another technique which uses force, displacement, and 
time has been developed. 3D modeling is a very useful method to characterize the 
tableting process [3, 46, 47, 155 – 159] . Force is expressed as pressure, time has to be 
normalized, and from displacement data the porosity according to Heckel [119] is 
calculated. It is the only possibility to combine these variables during analysis. 
To describe the tableting process the three variables were presented in a 3D plot 
(3D data plot) and a plane was fi tted to the data twisted at t = tmax (Figure 19 a ). 
From the fi tting process the parameters d (time plasticity), e (pressure plasticity), 
ANALYSIS OF TABLETING PROCESS 1079

1080 TABLET PRODUCTION SYSTEMS 
FIGURE 19 ( a ) 3D data plot with fi tted plane twisted at t = t max and ( b ) 3D parameter plot 
of (  ) DCPD: dicalcium phosphate dihydrate, (  ) spray - dried lactose, (  ) MCC: microcrystalline 
cellulose, ( ) theophylline monohydrate, and (  ) HPMC: hydroxypropyl methylcellulose 
for data gained with an eccentric tableting machine [47] . 
Time (normalized) 
Pressure (MPa) 
In[1/(1-Drel)] 
2,5
2 
1,5
1 
0,5
0 
–0,5 
–1
0 
0,2 
0,4 
0,6 
0,8 
1 0 
50 
100 
150 
0.04 
0.02 
0.00
0 
1 
2 
0.000 
0.005 
0.010 
0.015 
0.72 
0.74 
0.90 
0.84 0.72 
0.89 
0.88 
0.72 
Increasing .rein, max 
d 
e (MPa–1 
) 
. 
(b) 
(a) 
and . (twisting angle, which indicates fast elastic decompression) can be derived 
(Table 8 ). 
The parameters of the fi tted plane (time plasticity d , pressure plasticity e , and 
twisting angle . ) were also exhibited in a 3D plot and this plot is called the 3D 
parameter plot. This plot exhibits the compression behavior of the powder. It gives 
a simple yet characteristic description of the tableting properties. An example is 
given in Figure 19 b . 

TABLE 8 Parameters Calculated from Force – Displacement – Time Profi les 
Source Parameters 
3D model [143, 
144, 155] 
(Figure 19 ) 
z 
D 
t t d p p ep f dt = 
. ( )= . + . + + + ln [( )( )] ( ) ( ) max max max 
1 
1 rel 
. 
d 
D 
t 
e 
D 
p 
f 
D 
= . = . = 
. ( ) . 
. 
. 
. 
ln[ ( )] ln[ ( )] 
ln 
1 1 1 1 1 
1 
/ / rel rel 
rel 
where D rel = relative density 
t = time 
p = pressure 
t max = time at maximum pressure 
p max = maximum pressure 
. = twisting angle at t max , indicates fast elastic decompession 
d = time plasticity 
e = pressure plasticity 
Time plasticity d describes the plastic deformation of the excipient according to 
time [160] . It is infl uenced by tableting speed [157] . With increasing time plasticity 
d the powder deforms faster during tableting. Therefore, with increasing densifi cation 
time plasticity increases. Pressure plasticity e describes the pressure - dependent 
increase of density. The pressure plasticity correlates with the slope of the 
Heckel equation [144] . With increasing pressure plasticity e the slope of the Heckel 
equation increases in the same direction and the necessary pressure for deformation 
(yield pressure [118] or yield strength [120] ) decreases. The twisting angle . 
is a measure of the material ’ s elasticity and the ratio between compression and 
decompression. Thus, it indirectly describes fast elastic decompression during the 
tableting process. When . increases, elasticity decreases. The twisting angle . correlates 
with the elastic modulus [144] . In conclusion, materials which deform fast 
show high d values, materials which deform easily and with low pressure show 
high e values, and those which relax a lot show a lot elasticity and thus low . 
values. 
Thus the 3D model allows us to characterize the tableting process completely 
and to distinguish time - dependent information from pressure - dependent deformation 
and elasticity in one step of the analysis. 
Temperature Analysis The results gained by determination of temperature during 
and shortly after tableting vary strongly and depend on the method used. 
A temperature increase of 5 K [79, 80, 161] could be determined with conventional 
punches; however, with epoxide punches the measured temperature increase 
was as high as 30 K [81] . By calorimetric measurement an increase of 10 up to 30 K 
[82] was determined and by infrared measurement the increase was 10 – 15 K 
[84] . 
Beissenhirtz [86] measured a temperature increase of 30 K indirectly by measuring 
conductivity which arose during tableting with conductive materials. Energy 
calculations indicate a temperature increase of more than 30 K caused by tableting 
ANALYSIS OF TABLETING PROCESS 1081

1082 TABLET PRODUCTION SYSTEMS 
[78, 84, 83] . Most recently, partial melting of drugs could be analyzed for materials 
whose melting temperature is as high as 94 ° C [87] , and the reversible transgression 
of a glass transition temperature of 80 ° C was determined [162] . 
All results indicated that temperature increase depends on the material. Further 
temperature increase during tableting can contribute to slight changes in material 
structure [85] . 
6.4.9 ANALYSIS OF FINAL TABLET FORMATION 
This characterization of the process of tablet formation has to be completed by 
analyzing the changes induced by tableting. 
Most important is the elastic recovery of the tablets which starts during decompression 
and is fi nished dependent on the material after several days. Elastic recovery 
can be defi ned as [163] . 
ER % t ( ) min 
min 
= . . 
100 
H H 
H 
(2) 
where H min is the minimum height of the tablet under load and H t the height of the 
tablet at different times t after tableting. 
Elastic recovery gives information on the remaining elasticity of the materials 
which is only slowly released. Further it can indicate structural changes inside the 
materials and tablets. Structural changes induced by tableting have to be analyzed 
by physicochemical techniques, such as spectroscopic and thermoanalytical methods, 
X - ray diffraction, scanning electron microscopy, and transmission electron microscopy 
[35, 36, 85, 164 – 166] . The analyzed changes will help to better understand the 
process of tablet formation and identify the reasons for compactibility of materials. 
However, these changes are not the subject of this chapter. 
6.4.10 COMPLETE DESCRIPTION OF PROCESS 
OF TABLET FORMATION 
On the whole, the process of tablet formation can fully be described by combining 
the analysis of the tableting process with the fi nal formation of the tablets. The 
methods which gives most detailed information of the whole process and simultaneously 
is a fast method is the 3D modeling technique in combination loith calculating 
the elastic recovery of the tablets. In addition, by combining both these methods 
and calculating general plasticity P from time plasticity d , pressure plasticity e , twisting 
angle . , and elastic recovery ER, a more general tool for analysis of the process 
of tablet formation is available [3, 4] . 
Finally the crushing force of the tablets after relaxation gives information on 
the formed bonds inside the material and the compactibility. Compactibility has 
been described by Leuenberger [167] . For the future it can be expected that a 
prediction of compactibility as a result of the process of tablet formation is 
possible. 

6.4.11 SPECIAL ACCESSORIES OF TABLETING MACHINES 
In tablet production it is essential to control tablet weight and tablet homogeneity 
in order to ensure a uniform dosage form. For patient safety pharmacopeias demand 
that tablet weight is between certain limits. Tablet producers often set their more 
narrow specifi cations to ensure that they meet pharmacopeial specifi cations. To 
control tablet weight and tablet homogeneity an optimal product mixture, complete 
fi lling of the die, exact tooling, and tightly controlled machine conditions are 
necessary. 
Another demand for patient safety and due to GMP regulations is to ensure the 
absence of impurities in the fi nal dosage form, for example, residues from the previous 
product or residues from detergents. Thus the process of cleaning of machines 
has to be standardized and controlled. In addition optimized short cleaning times 
of the machine increase operating time for production. During the last years one 
innovation for tableting machines was the development of special accessories for 
cleaning in order to reduce standing times. 
For special products (e.g., cytostatics or sterile products), it is necessary to 
produce tablets in a hermetically closed machine. For these products special containment 
solutions have been developed which allow the production in a hermetically 
closed machine or behind a wall. Most important is to separate the tablet 
production zone strongly from the mechanics of the machine. 
In the following tablet fi lling devices, possibilities to control tablet weight 
and mixing homogeneity as well as advances in cleaning technology will be 
discussed. 
6.4.11.1 Optimization of Die Filling 
The basics of fi lling have been explained above: The fi lling shoe is moving back and 
forth, the fi lling shoe is fi xed, or fi lling is centrifugally controlled. Two problems arise 
generally: Either the product demixes and tablet weight and content uniformity are 
no longer controlled or the die is not completely fi lled and thus tablet weight also 
varies. 
Optimal fi lling of the die is determined to a great extent by the material, but the 
speed of the machine is also important. At low machine speeds the die is usually 
completely fi lled; at high machine speeds this becomes more diffi cult. Thus special 
fi lling devices using one or more paddles have been developed to improve fi lling. 
One example for a paddle feeder is given in Figure 20 . Alternatively fi lling devices 
can be vibrated to improve feeding for materials with bad fl ow characteristics. An 
overview is given by Ritschel and Bauer - Brandl [32, 168] . 
Besides improved feeding, paddle feeders allow improved mixing uniformity 
since the formulation is mixed again shortly before feeding the die. This mechanical 
remixing is the only possibility to improve the homogeneity of the mixture. Demixing 
is a bigger problem for machines with a moving fi lling shoe than for those with 
a fi xed fi lling shoe. Thus moving fi lling shoes are equipped with paddles as a standard. 
Fixed fi lling shoes need paddles usually only for materials with bad fl owability 
or at high machine speeds. 
SPECIAL ACCESSORIES OF TABLETING MACHINES 1083

1084 TABLET PRODUCTION SYSTEMS 
6.4.11.2 Tablet Weight Control 
To control tablet weight different possibilities exist. The simplest method is to weigh 
at preset intervals (in - process control) a number of tablets manually and to adjust 
machine settings according to the results when necessary. For high - speed rotary 
machines automatically working weighing systems have been developed which 
determine tablet weight shortly after ejection [169, 170] . These systems can also 
determine tablet height and diameter, which are indirect measures for tablet 
uniformity. Simultaneously with tablet weight compression force drifts. A direct 
relation between compression force and tablet weight exists. Thus it is possible 
to monitor tablet uniformity also by control of compression force. For this purpose 
the machines are instrumented with strain gauges or piezoelectric force transducers. 
By control of compression, force changes in tablet weight can be directly 
detected. 
Automatically working control systems are able to eject those tablets separately 
which fail the requirements (rejection mechanism), and they adjust the machine 
for die fi lling or compression force and collect only those tablets which meet the 
requirements. Two alternative principles for automatic tablet weight control and 
adjustment are possible, depending on the application and selected machine type. 
The principle of control of compression force is based on measurement of the fi nal 
compression force under constant tablet height. This principle is used for all applications 
where tablet weight accuracy and constant tablet density are less critical. 
The principle of control of displacement is based on measurement of tablet 
height variations under constant force. This principle is more accurate than the force 
control system. It is used for all applications where constant density of the produced 
tablets is critical. 
Modern systems combine one of these control systems with automatic weighing 
of tablets. Weight control will automatically adjust the fi lling depth in order to keep 
tablet weight within specifi ed tolerance limits. 
6.4.11.3 Control of Mixing Homogeneity 
The systems to control weight uniformity are not able to control uniformity of the 
mixture. When during fi lling of the die the tableted material demixes, tablet weight 
FIGURE 20 Example paddle feeder. (Courtesy of Kilian.) 

usually tends to vary. However, these variations can be small and not easy to detect. 
To monitor mixing uniformity in the fi nal tablet, most recently spectroscopic techniques 
such as Raman spectroscopy and near - infrared (NIR) spectroscopy have 
been used [171 – 173] . Special online sensors have been build into the machine and 
they measure the spectrum for each tablet. When the mixture is homogeneous, the 
appearance of the spectrum will always be the same or between certain limits. If 
not, the production can be stopped and adjusted as far as possible. Thus a further 
step in quality assurance of tablets has been made. This was partially caused by the 
process analytical technique (PAT) initiative of the U.S. Food and Drug Administration 
(FDA) [174] . 
6.4.11.4 Cleaning 
To ensure product quality, cleaning is of utmost importance. Therefore different 
standardized cleaning technologies have been developed [175, 176] . Detailed information 
may be provided by manufacturers. 
Usually, the most effective way to implement cleaning is to design it into a 
process which has to be performed after tableting. It involves the addition of spray 
systems, tank cleaners, nozzles, and seals into the tableting machine in order to 
automate the cleaning process. The automation converts the batch processes to a 
continuous operation of tableting cycles and cleaning cycles. Cleaning or washing 
in place means an advanced wash liquid preparation system which handles all fi ltering, 
preheating, mixing, and pumping of water, detergents, and demineralized water 
and provides continuous monitoring and control of cleaning parameters. Another 
possibility for standardized cleaning is the wash - off - line procedure. In this case 
exchangeable compression modules are especially designed for a fast product 
changeover. Different techniques have been developed to exchange the modules. 
Either carries or arms or lifting systems are applied or additionally used. The wash - 
off - line procedure increases production time by cleaning the exchangeable compression 
modules after it is removed from the tablet press. Thus production time is 
increased; however, two compression modules are necessary. In this case a special 
separate washing system is necessary. 
6.4.12 IMPORTANT FACTORS DURING MANUFACTURING PROCESS 
To run a tablet production process effectively, robustly and smoothly several factors 
have to be kept in mind. Of utmost importance for the process are environmental 
humidity during tablet production and adhesion forces between machine punches 
and dies. These and other factors can contribute to problems during manufacturing. 
In the following the relevance of climatization during tableting, the necessity and 
methods of lubrication, and frequently occurring problems during manufacturing 
will be discussed. 
6.4.12.1 Climatization 
Relative humidity (RH) in the production room infl uences the water content of the 
materials. This has to be kept in mind during tablet production. 
IMPORTANT FACTORS DURING MANUFACTURING PROCESS 1085

1086 TABLET PRODUCTION SYSTEMS 
When the materials sorb water, they deform differently compared to the status 
before sorption, and compressibility changes [98, 177, 178] . Furthermore the compactibility 
of the materials changes and tablets with a different crushing force and 
friability result [179] . Even the release from the tablets can be infl uenced [180] . 
However, the infl uence of water content on tableting and tablet properties depends 
on the material; for example, hydrophilic polymers are mostly infl uenced by RH. 
Further, the infl uence of RH during production is most decisive when production 
conditions change extremely. Smaller differences up to 10% RH do not infl uence 
a robust formulation [3] ; however for critical formulations even these changes are 
of importance. Usually the conditions at one production site do not change from 
day to day, but great differences have been observed between different seasons of 
the year. Since tablet production and the fi nal tablet quality should be the same 
throughout the year, often a humidity interval between 40 and 60% RH is used for 
tablet production. This is a fi rst step. However, for characterization of material 
properties humidity control between 40 and 60% RH is not suffi cient. Material 
properties cannot be compared when obtained at different conditions. In this 
case humidity control at a certain humidity with a precision of 2 – 5% is absolutely 
necessary [3] . 
6.4.12.2 Lubrication 
Adhesion forces between the material and punches and dies result in sticking of the 
tablets at the punches and dies. These adhesion forces are further infl uenced by RH 
and this has also to be kept in mind. When the adhesion forces at the punches and 
dies are greater than the cohesion forces between the particles inside the tablet, the 
tablets stick at the punches and can cap. 
To overcome this problem, lubrication is the method of choice [181, 182] . Two 
alternatives for lubrication exist: internal lubrication and external lubrication. Internal 
lubrication is performed by mixing the tableted product shortly before the 
tableting process with a solid lubricant. Thus the lubricant is not only at the surfaces 
of the fi nal tablet but also inside the tablet. As a result, internal lubrication lowers 
bonding, and this is especially the case for plastically deforming materials [183 – 186] . 
The most frequently used and most effective material for internal lubrication is 
magnesium stearate [187 – 189] ; however, other hydrophobic or amphiphilic lubricants 
are also possible [190 – 196] . Magnesium stearate has one major disadvantage: 
It shows a low solubility and remains as a solid after dissolution. Thus the search 
for other lubricants is ongoing. 
When the lubricant should not be part of the tablet formulation, for example, 
when bonding properties of the drug are low, external lubrication is necessary [197] . 
For single - tablet production the punches and dies can be manually lubricated with 
a fl uid. In production several methods have been developed to place the fl uid on 
the surface of punches and dies [198] . Filaments applied at the punches to lubricate 
the die or special caps with fl uid lubricant are possible solutions. However, external 
lubrication also has disadvantages [199] . 
6.4.12.3 Occurring Problems during Manufacturing 
The most frequent problems occurring during the manufacture of tablets are high 
tablet weight variation, capping and lamination [200] , and further picking and stick

ing at punches and dies. Low product yield, low crushing force, and further tablet 
yams and chipping are other problems which have to be solved [33, 201] . 
High tablet weight variation can be reduced by using weight control systems. 
Further demixing of the tableted material has to be avoided, since demixing results 
in higher tablet weight variation and content uniformity can no longer be achieved. 
As already discussed, paddle feeders can be used to achieve mixing homogeneity 
for problematic products and further spectroscopic techniques can be used for 
control. 
The problem of capping and lamination can be solved by increasing RH or 
adding wetting agents. Further either external lubrication may help. Picking and 
sticking of the tablets at punches and dies can be avoided by using lubricants as 
discussed above. 
A low product yield is caused by loss of material during fast production processes. 
On rotary tableting machines this problem is solved by slightly lowering the 
lower punches before the compression event starts. 
A low crushing force is often caused by the composition of the special formulation. 
If the formulation itself is not the reason for a low crushing force, compression 
force can easily be increased. Another possible explanation can be low humidity of 
the tableted product. 
Finally tablets yam and chipping can occur before the tablets leave the die table 
of a rotary machine. Usually the lower punches or the tablet scraper of the tableting 
machine are not properly adjusted. Another reason can be low crushing force. 
In conclusion, for smooth and perfect machine runs, product properties and 
machine conditions have to be tightly controlled. 
6.4.13 FUTURE OF TABLET PRODUCTION SYSTEMS 
In principle tablet production systems have remained the same throughout the last 
century. However, major improvements in instrumentation, data acquisition, and 
analysis techniques have been made. Nowadays more sophisticated data acquisition 
and analysis techniques are available which facilitate and improve interpretation of 
tableting data. 
In order to facilitate scale - up, more sophisticated simulation systems can be 
thought of. They will be a real help for scale - up with small amounts of material as 
available in early development of formulations. 
Further quality control of the tablets during tableting or shortly after has become 
more important. Recent trends show improvements for production in a GMP environment 
by isolating the production unit from the machinery. For the near future 
the implementation of the PAT initiative of the FDA is conceivable. 
REFERENCES 
1. Brockedon , W. ( 1843 ), Shaping pills, lozenges, and black lead by pressure in dies, British 
Patent 9977. 
2. Leuenberger , H. ( 1997 ), Research in solid dosage forms — An obsolete topic? Pharm. 
Dev. Technol. , 2 , vii – viii . 
REFERENCES 1087

1088 TABLET PRODUCTION SYSTEMS 
3. Picker , K. M. ( 2002 ), New Insights in the Process of Tablet Formation — Ways to 
Explore Soft Tableting , Habilitationsschrift, Martin - Luther - University Halle - Wittenberg, 
Halle, Germany, and also G o rich und Weiersh a user Verlag, Marburg. 
4. Picker , K. M. ( 2004 ), Soft tableting: A new concept to tablet pressure sensitive drugs , 
Pharm. Dev. Technol. , 9 ( 1 ), 107 – 121 . 
5. Train , D. ( 1957 ), Transmission of forces through a powder mass during the process of 
pelleting , Trans. Inst. Chem. Eng. , 35 , 258 – 266 . 
6. Parrott , E. L. ( 1990 ), Compression in pharmaceutical dosage forms , in Lieberman , H. 
A. , Lachman , L. , and Schwartz , J. B. , Eds., Pharmaceutical Dosage Forms, Tablets. Bd.2 , 
Marcel Dekker , New York , pp. 201 – 243 . 
7. Guo , H. X. , Hein a m a ki , J. , and Yliruusi , J. ( 1999 ), Characterization of particle deformation 
during compression measured by confocal laser scanning microscopy , Int. J. Pharm. , 
186 , 99 – 108 . 
8. Hiestand , E. N. ( 1997 ), Mechanical properties of compacts and particles that control 
tableting success , J. Pharm. Sci. , 86 , 985 – 990 . 
9. Rumpf , H. ( 1958 ), Grundlagen und Methoden des Granulierens , Chem. Ing. Tech. , 30 , 
144 – 158 . 
10. Joneja , S. K. , Harcum , W. W. , Skinner , G. W. , Barnum , P. E. , and Guo , J. H. ( 1999 ), 
Investigating the fundamental effects of binders on pharmaceutical tablet performance , 
Drug. Dev. Ind. Pharm. , 25 , 1129 – 1135 . 
11. Adolfsson , A. , Gustafsson , C. , and Nystr o m , C. ( 1999 ), Use of tablet tensile strength 
adjusted to surface area and mean interparticulate distance to evaluate dominating 
bonding mechanism , Drug Dev. Ind. Pharm. , 25 , 753 – 764 . 
12. Rippie , E. G. , and Morehead , W. T. ( 1994 ), Structure evolution of tablets during compression 
unloading , J. Pharm. Sci. , 83 , 708 – 715 . 
13. Hoag , S. W. , and Rippie , E. G. ( 1994 ), Thermodynamic analysis of energy dissipation 
by pharmaceutical tablets during stress unloading , J. Pharm. Sci. , 83 , 903 – 908 . 
14. Leuenberger , H. , and Ineichen , L. ( 1997 ), Percolation theory and physics of compression 
, Eur. J. Pharm. Biopharm. , 44 , 269 – 272 . 
15. Hiestand , E. N. ( 1997 ), Principles, tenets and notions of tablet bonding and measurements 
of strength , Eur. J. Pharm. Biopharm. , 44 , 229 – 242 . 
16. Armstrong , N. A. , and Haines - Nutt , R. F. ( 1972 ), Elastic recovery and surface area 
changes in compacted powder systems , J. Pharm. Pharmacol. , 24S , 135P – 136P . 
17. Newton , J. M. , and Rowley , G. ( 1973 ), The infl uence of tablet weight on compaction 
pressure/tablet density relations , J. Pharm. Pharmacol. , 25 , 767 – 768 . 
18. van der Voort Maarschalk , K. ( 1997 ), Tablet relaxation, origin and consequences of stress 
relief in tablet formation , Ph.D. thesis, Riksuniversiteit Groningen. 
19. Aulton , M. E. , Travers , D. N. , and White , P. J. P. ( 1973 ), Strain recovery of compacts 
on extended storage , J. Pharm. Pharmacol. , 25 , 79P – 86P . 
20. York , P. , and Baily , E. D. ( 1977 ), Dimensional changes of compacts after compression , 
J. Pharm. Pharmacol. , 29 , 70 – 74 . 
21. van der Voort Maarschalk , K. , Zuurman , K. , Vromans , H. , Bolhuis , G. K. , and Lerk , 
C. F. ( 1997 ), Stress relaxation of compacts produced from viscoelastic materials , Int. J. 
Pharm. , 151 , 27 – 34 . 
22. F u hrer , C. ( 1975 ), Kristallographische Aspekte bei der Tablettenherstellung , Fortbildungs - 
kurs Arbeitsgemeinschaft f u r Pharmazeutische Verfahrenstechnik, Braunschweig. 
23. H u ttenrauch , R. ( 1978 ), Molekulargalenik als Grundlage moderner Arzneiformung , 
Acta Pharm. Technol. , 24 ( S6 ), 55 – 127 . 

24. Elamin , A. , Sebhatu , T. , and Ahlneck , C. ( 1995 ), The use of amorphous model substances 
to study mechanically activated materials in the solid state , Int. J. Pharm. , 119 , 
25 – 36 . 
25. Sebhatu , T. , Ahlneck , C. , and Alderborn , G. ( 1997 ), The effect of moisture content on 
the compression and bond - formation properties of amorphous lactose particles , Int. 
J. Pharm. , 146 , 101 – 114 . 
26. Sebhatu , T. , and Alderborn , G. ( 1999 ), Relationships between the effective interparticulate 
contact area and the tensile strength of tablets of amorphous and crystalline lactose 
of varying particle size , Eur. J. Pharm. Sci. , 8 , 235 – 242 . 
27. Moldenhauer , H. , Kala , H. , Zessin , G. , and Dittgen , M. ( 1980 ), Zur pharmazeutischen 
Technologie der Tablettierung , Pharmazie , 35 , 714 – 726 . 
28. Chan , H. K. , and Doelker , E. ( 1985 ), Polymorphic transformation of some drugs under 
compression , Drug Dev. Ind. Pharm. , 11 , 315 – 332 . 
29. Pirttim a ki , J. , Laine , E. , Ketolainen , J. , and Paronen , P. ( 1993 ), Effects of grinding 
and compression on crystal structure of anhydrous caffeine , Int. J. Pharm. , 95 , 93 – 
99 . 
30. Hiestand , E. N. ( 1991 ), Tablet bond. I. A theoretical model , Int. J. Pharm. , 67 , 
217 – 229 . 
31. Hiestand , E. N. , and Smith , D. P. ( 1991 ), Tablet bond. II. Experimental check of the 
model , Int. J. Pharm. , 67 , 231 – 246 . 
32. Ritschel , W. A. , and Bauer - Brandl , A. ( 2002 ), Die Tablette — Handbuch der Entwicklung, 
Herstellung und Qualit a tssicherung , Editio Cantor Verlag , Aulendorf Germany . 
33. Bogda , M. J. ( 2002 ), Tablet compression: Machine theory, design, and process troubleshooting 
, in Swarbrick , J. , and Boylan , J. C. , Eds., Encyclopedia of Pharmaceutical 
Technology , Marcel Dekker , New York , pp. 2669 – 2688 . 
34. Hausmann , R. , Kaufmann , H.-J. , and Richter , K. ( 1996 ), Neue Tablettiertechnologie 
mit zentrifugaler Matrizenf u llung und vollautomatischer Reinigung , Pharm. Ind. , 58 , 
842 – 846 . 
35. Picker , K. M. ( 2000 ), The automatic micrometer screw , Eur. J. Pharm. Biopharm. , 49 ( 2 ), 
171 – 176 . 
36. Picker , K. M. ( 2001 ), Time dependence of elastic recovery for characterization of tableting 
materials , Pharm. Dev. Technol. , 6 ( 1 ), 61 – 70 . 
37. Caraballo , I. , Millan , M. , Fini , A. , Rodriguez , L. , and Cavallari , C. ( 2000 ), Percolation 
thresholds in ultrasound compacted tablets , J. Controlled Release , 69 , 
345 – 355 . 
38. Aulton , M. E. ( 1981 ), Indentation hardness profi les across the faces of some compressed 
tablets , Pharm. Acta Helv. , 56 , 133 – 136 . 
39. Konkel , P. , and Mielck , J. B. ( 1997 ), Associations of parameters characterizing the time 
course of the tabletting process on a reciprocating and on a rotary tabletting machine 
for high - speed compression , Eur. J. Pharm. Biopharm. , 44 , 289 – 301 . 
40. Sucker , H. , Fuchs , P. , and Speiser , P. (1991), Pharmazeutische Technologie , Thieme 
Verlag, Stuttgart. 
41. Bateman , S. D. , Rubinstein , M. H. , Rowe , R. C. , Roberts , R. J. , Drew , P. , and Ho , A. 
Y. K. ( 1989 ), A comparative investigation of compression simulators , Int. J. Pharm. , 49 , 
209 – 212 . 
42. Celik , M. , and Marshall , K. ( 1989 ), Use of a compaction simulator system in tabletting 
research , Drug Dev. Ind. Pharm. , 15 , 759 – 800 . 
43. Celik , M. , and Lordi , N. G. ( 1991 ), The pharmaceutical compaction research laboratory 
and information center , Pharm. Technol. , 15 , 112 – 116 . 
REFERENCES 1089

1090 TABLET PRODUCTION SYSTEMS 
44. Rubinstein , M. H. , Bateman , S. D. , and Thacker , H. S. ( 1991 ), Compression to constant 
thickness or constant force: Producing more consistent tablets , Pharm. Technol. , 1 , 
150 – 158 . 
45. Celik , M. , Ong , J. T. , Chowhan , Z. T. , and Samuel , G. J. ( 1996 ), Compaction simulator 
studies of a new drug substance: Effect of particle size and shape, and its binary mixtures 
with microcrystalline cellulose , Pharm. Dev. Technol. , 1 , 119 – 126 . 
46. Picker , K. M. ( 2000 ), Three - dimensional modeling to determine properties of tableting 
materials on rotary machines using a rotary tableting machine simulator , Eur. J. Pharm. 
Biopharm. , 50 ( 2 ), 293 – 300 . 
47. Picker , K. M. ( 2003 ), The 3D model: Comparison of parameters obtained from and by 
simulating different tableting machines , AAPS PharmSciTech. , 4 ( 3 ) article 35 . 
48. Levin , M. ( 1999 ), Theory and practice of tablet press simulation for process scale - up , 
paper presented at the Arden House Conference Harsiman, New York, USA . 
49. Rippie , E. G. , and Danielsson , D. W. ( 1981 ), Viscoelastic stress/strain behaviour 
of pharmaceutical tablets: Analysis during unloading and postcompression periods , 
J. Pharm. Sci. , 70 , 476 – 482 . 
50. Oates , R. J. , and Mitchell , A. G. ( 1989 ), Calculation of punch displacement and work of 
powder compaction on a rotary tablet press , J. Pharm. Pharmacol. , 41 , 517 – 523 . 
51. Oates , R. J. , and Mitchell , A. G. ( 1990 ), Comparison of calculated and experimentally 
determined displacement on a rotary tablet press using both Manesty and IPT punches , 
J. Pharm. Pharmacol. , 42 , 388 – 396 . 
52. Oates , R. J. , and Mitchell , A. G. ( 1994 ), A new method of estimating volume during 
powder compaction and the work of compaction on a rotary tablet press from measurements 
of applied vertical forces , J. Pharm. Pharmacol. , 46 , 270 – 275 . 
53. Pudipeddi , M. , Venkatesh , G. , Faulkner , P. , and Palepu , N. ( 1993 ), Correlations between 
compaction simulator and instrumented Betapress , Pharm. Res. , 10S , S165 . 
54. Ruegger , C. D. ( 1996 ), An investigation of the effect of compaction profi les on the 
tableting properties of pharmaceutical substances , Ph.D. thesis, Rutgers University, 
Newark, NJ. 
55. Levin , M. , Tsygan , L. , and Dukler , S. ( 1998 ), U.S. Patent 6,106.262, International Patent 
Application No. PCT/US98/27421. 
56. MCC Corp . ( 2000 ), The Presster Binder , Technical Information, MCC Corp., East 
Hanover, New Jersey, USA . 
57. Lamey , K. ( 2000 ), Correlations between compaction simulators and rotary tableting 
machines , paper presented at the Compaction Simulator’s User Meeting, Loughbourough, 
UK. 
58. Guntermann , A. ( 2005 ), The Presster — A tablet press simulator , paper presented at 
TabletTech, Brussels. 
59. MedelPharm ( 2005 ), Technical information , Medel Pharm., Bourg - en - Bresee, France . 
60. Brake , E. F. ( 1951 ), Development of methods for measuring pressures during tablet 
manufacture , M.S. thesis, Purdue University, West Lafayette, IN. 
61. Higuchi , T. , Arnold , R. D. , Tucker , S. J. , and Busse , L. W. ( 1952 ), The physics of tablet 
compression. I. A preliminary report , J. Am. Pharm. Assoc. , 41 , 93 – 96 . 
62. Higuchi , T. , Nelson , E. , and Busse , L. W. ( 1954 ), The physics of tablet compression. III: 
Design and construction of an instrumented tableting machine , J. Am. Pharm. Assoc. , 
43 , 344 – 348 . 
63. Morehead , W. T. , and Rippie , E. G. ( 1990 ), Timing relationships among maxima of 
punch and die - wall stress and punch displacement during compaction of viscoelastic 
solids , J. Pharm. Sci. , 79 , 1020 – 1022 . 

64. Yeh , C. , Altaf , S. A. , and Hoag , S. W. ( 1997 ), Theory of force transducer design 
optimization for die wall stress measurement during tablet compaction: Optimization 
and validation of split - web die using fi nite element analysis , Pharm. Res. , 14 , 1161 – 
1170 . 
65. Khossravi , D. , and Morehead , W. T. ( 1997 ), Consolidation mechanisms of pharmaceutical 
solids: A multi-compression cycle approach, Pharm. Res. , 14 , 1039 – 1045 . 
66. Knoechel , E. L. , Sperry , C. C. , Ross , H. E. , and Lintner , C. J. ( 1967 ), Instrumented rotary 
tablet machines I , J. Pharm. Sci. , 56 , 109 – 115 . 
67. Schmidt , P. C. , Tenter , U. , and Hocke , J. ( 1986 ), Presskraft - und Weg - Zeit - 
Charakteristik von Rundlauftablettenpressen. 1. Mitt.: Instrumentierung von Einzel - 
stempeln zur Presskraftmessung , Pharm. Ind. , 48 , 1546 – 1553 . 
68. Bauer - Brandl , A. ( 1998 ), Qualifi zierung der Kraftmessung an Tablettenpressen , Pharm. 
Ind. , 60 , 63 – 69 . 
69. Hoag , S. W. , Nair , R. , and Muller , F. X. ( 2000 ), Force - transducer - design optimization for 
the measurement of die - wall stress in a compaction simulator , Pharm. Pharmacol. 
Commun. , 6 ( 7 ), 293 – 298 . 
70. Laich , T. , and Kissel , T. ( 1995 ), Axial die - wall force minimum. Infl uences and signifi cance 
for elastic behavior of single components and binary mixtures of excipients , Pharm. Ind. 
57 ( 2 ), 174 – 182 . 
71. Nelson , E. , Busse , L. W. , and Higuchi , T. ( 1955 ), The physics of tablet compression: VII. 
Determination of energy expenditure in the tablet compression process , J. Am. Pharm. 
Assoc. Sc. Ed., 44 , 223 . 
72. Schmidt , P. C. , and Tenter , U. ( 1987 ), Force and displacement characteristics of rotary 
tableting machines , Pharm. Ind. , 49 , 637 – 642 . 
73. Matz , C. ( 1999) , Evaluation einer IR - telemetrischen Kraft/Weg - Instrumentierung f u r 
Rundlauf tabletten pressen . Differenzierung des Verformungsverhaltens direktkomprimierbarer 
Tablettierhilfsstoffe, Dissertation, Universit a t Freiburg, Freiburg, 
Germany. 
74. Matz , C. , Bauer - Brandl , A. , Rigassi , T. , Schubert , R. , and Becker , D. ( 1999 ), On the 
accuracy of a new displacement instrumentation for rotary tablet presses , Drug Dev. Ind. 
Pharm. , 25 , 117 – 130 . 
75. M u ller , F. , and Caspar , U. ( 1984 ), Viskoelastische Ph a nomene w a hrend der Tablettierung 
, Pharm. Ind. , 46 , 1049 – 1056 . 
76. Krumme , M. ( 1992 ), Entwicklung rechnergest u tzter Verfahren zur Kompressions - und 
Festigkeitsanalyse von Tabletten , Dissertation, Freie Universit a t, Berlin. 
77. Krumme , M. , Schwabe , L. , and Fr o mming , K. H. ( 1998 ), Development of computerised 
procedures for the characterization of the tableting properties with eccentric machines. 
High precision displacement instrumentation for eccentric tablet machines , Acta Pharm. 
Hung. , 68 , 322 – 331 . 
78. Rankell , A. S. , and Higuchi , T. ( 1968 ), Physics of tablet compression. XV. Thermodynamic 
and kinetic aspects of adhesion under pressure , J. Pharm. Sci. , 58 , 574 – 577 . 
79. DeCrosta , M. T. , Schwartz , J. B. , Wigent , R. J. , and Marshall , K. ( 2000 ), Thermodynamic 
analysis of compact formation; compaction, unloading, and ejection. I. Design and development 
of a compaction calorimeter and mechanical and thermal energy determinations 
of powder compaction , Int. J. Pharm. , 198 , 113 – 134 . 
80. DeCrosta , M. T. , Schwartz , J. B. , Wigent , R. J. , and Marshall , K. ( 2001 ), Thermodynamic 
analysis of compact formation; compaction, unloading, and ejection. II. Mechanical 
energy (work) and thermal energy (heat) determinations of compact unloading and 
ejection , Int. J. Pharm. , 213 , 45 – 62 . 
REFERENCES 1091

1092 TABLET PRODUCTION SYSTEMS 
81. Bogs , H. , and Lenhardt , E. ( 1971 ), Zur Kenntnis thermischer Vorg a nge beim Tablettenpressen 
, Pharm. Ind. , 33 , 850 – 854 . 
82. Hanus , E. J. , and King , L. D. ( 1968 ), Thermodynamic effects in compression of solids , J. 
Pharm. Sci. , 57 , 677 – 684 . 
83. F u hrer , C. , and Parmentier , W. ( 1977 ), Zur Thermodynamik der Tablettierung , Acta 
Pharm. Technol. , 23 , 205 – 213 . 
84. Ketolainen , J. , Ilkka , J. , and Paronen , P. ( 1993 ), Temperature changes during tabletting 
measured using infrared thermoviewer , Int. J. Pharm. , 92 , 157 – 166 . 
85. Picker - Freyer , K. M. , and Schmidt , A. G. ( 2004 ), Does temperature increase induced by 
tableting contribute to tablet quality? J. Therm. Anal. Cal. , 77 , 531 – 539 . 
86. Beissenhirtz , M. ( 1974 ), Ermittlung der Verformungscharakteristik von Tablettierstoffen 
mit Hilfe elektrisch leitf a higer Zus a tze , Dissertation, Universit a t Bonn, Bonn, 
Germany. 
87. Schmidt , J. ( 1997 ), Direktablettierung niedrigschmelzender nichtsteroidaler Antirheumatika 
mit mikrokristallinen Cellulosen , Dissertation, Universit a t Halle - Wittenberg, 
Wittenberg, Germany. 
88. Celik , M. ( 1992 ), Overview over compaction analysis techniques , Drug Dev. Ind. Pharm. , 
18 , 767 – 810 . 
89. Alderborn , G. , and Nystr o m , C. ( 1996 ), Pharmaceutical Powder Compaction Technology , 
Marcel Dekker , New York . 
90. Chilamkurti, R. N. , Rhodes , C. T. , and Schwartz, J. B. (1982), Some studies on compression 
properties of tablet matrices using a computerized instrumented press , Drug Dev. 
Ind. Pharm. , 8 , 63 – 86 . 
91. Emschermann , B. , and M u ller , F. ( 1981 ), Evaluation of force measurements in tablet 
manufacture , Pharm. Ind. , 43 , 191 – 194 . 
92. Vogel , P. J. , and Schmidt , P. C. ( 1993 ), Force - time curves of a modern rotary tablet 
machine. Part 2. Infl uence of compression force and tableting speed of the deformation 
mechanisms of pharmaceutical substances , Drug Dev. Ind. Pharm. , 19 , 1917 – 1930 . 
93. Schmidt , P. C. , and Vogel , P. J. ( 1994 ), Force - time - curves of a modern rotary tablet 
machine. Part 1. Evaluation techniques and characterization of deformation behavior 
of pharmaceutical substances , Drug Dev. Ind. Pharm. , 20 , 921 – 934 . 
94. Schmidt , P. C. , and Leitritz , M. ( 1997 ), Compression force/time - profi les of microcrystalline 
cellulose, dicalcium phosphate dihydrate and their binary mixtures — A critical 
consideration of experiments and parameters , Eur. J. Pharm. Biopharm. , 44 , 303 – 313 . 
95. Yliruusi , J. K. , Merkku , P. , Hellen , L. , and Antikainen , O. K. ( 1997 ), A new method to 
evaluate the elastic behavior of tablets during compression , Drug Dev. Ind. Pharm. , 23 , 
63 – 68 . 
96. Dietrich , R. , and Mielck , J. B. ( 1985 ), Eignung der Weibull - Funktion zur Charakterisierung 
des zeitabh a ngigen Verformungsverhaltens von Tablettierhilfsstoffen , Acta Pharm. 
Technol. , 31 , 67 – 76 . 
97. Dietrich , R. , and Mielck , J. B. ( 1985 ), Parametrisierung des zeitlichen Verlaufs der Verdichtung 
bei der Tablettierung mit Hilfe der modifi zierten Weibull - Funktion , Pharm. 
Ind. , 47 , 216 – 220 . 
98. Picker , K. M. ( 1995 ), Hydrophile Matrixtabletten: Tablettierung und Freisetzung - unter 
besonderer Ber u cksichtigung der relativen Feuchte w a hrend der Herstellung , Dissertation, 
Universit a t Hamburg, Hamburg, Germany. 
99. Shlieout , G. , Wiese , M. , and Zessin , G. ( 1999 ), A new method to evaluate the consolidation 
behavior of pharmaceutical materials by using the Fraser - Suzuki function , Drug 
Dev. Ind. Pharm. , 25 , 29 – 36 . 

100. Armstrong , N. A. , and Palfrey , L. P. ( 1989 ), The effect of machine speed on the 
consolidation of four directly compressible tablet diluents , J. Pharm. Pharmacol. , 41 , 
149 – 151 . 
101. Ho , A. Y. K., and Jones , T. M. ( 1988 ), Rise time: A new index of tablet compression , 
J. Pharm. Pharmacol. , 40 , 74P . 
102. Tsardaka , E. D. ( 1990 ), Viscoelastic properties and compaction behavior of pharmaceutical 
particulate material , Ph.D. Thesis, University of Bath, Bath, England. 
103. Dwivedi , S. K. , Oates , R. J. , and Mitchell , A. G. ( 1991 ), Peak offset times as an indication 
of stress relaxation during tableting on a rotary tablet press , J. Pharm. Pharmacol. , 
43 , 673 – 678 . 
104. F u hrer , C. ( 1962 ), U ber den Druckverlauf bei der Tablettierung in Exzenterpressen , 
Dtsch. Apoth. Ztg. , 102 , 827 – 842 . 
105. Moldenhauer , H. , H u nerbein , B. , and Kala , H. ( 1972 ), Recording of pressure - path diagrams 
from an eccentric press using piezoelectric measurement , Pharmazie , 27 , 
417 – 418 . 
106. D u rr , M. , Hanssen , D. , and Harwalik , H. ( 1972 ), Kennzahlen zur Beurteilung der 
Verpressbarkeit von Pulvern und Granulaten , Pharm. Ind. , 34 , 905 – 911 . 
107. de Blaey , C. J. , and Polderman , J. ( 1970 ), Compression of pharmaceuticals. I. The 
quantitative interpretation of force - displacement curves , Pharm. Weekblad , 105 , 241 – 
250 . 
108. de Blaey , C. J. , and Polderman , J. ( 1971 ), Compression of pharmaceuticals. II. Registration 
and determination of force - displacement curves using a small digital computer , 
Pharm. Weekblad , 106 , 57 – 65 . 
109. Stamm , A. , and Mathis , C. ( 1976 ), Verpressbarkeit von festen Hilfsstoffen f u r die 
Direkttablettierung , Acta Pharm. Technol., 24 ( S1 ), 7 – 16 . 
110. D u rr , M. ( 1976 ), Bedeutung der Energiebilanz beim Tablettieren f u r die Entwicklung 
von Tabletten rezepturen , Acta Pharm. Technol. , 22 , 185 – 194 . 
111. Parmentier , W. ( 1978 ), Investigation on the interpretation of the Kraft - Weg diagram , 
Pharm. Ind. , 40 , 860 – 865 . 
112. Lammens , R. F. , Polderman , J. , and De Blaey , C. J. ( 1979 ), Evaluation of force displacement 
measurements during powder compaction. Part 1. Precision and accuracy of force 
measurements , Int. J. Pharm. Technol. Prod. Manuf. , 1 , 26 – 35 . 
113. Lammens , R. F. , Polderman , J. , Armstrong , N. A. , and De Blaey , C. J. ( 1980 ), Evaluation 
of force displacement measurements during powder compaction. Part 2. Precision 
and accuracy of powder height and displacement measurements , Int. J. Pharm. Technol. 
Prod. Manuf. , 1 , 26 – 35 . 
114. Krycer , I. , and Pope , D. G. ( 1982 ), The interpretation of powder compaction data — A 
critical review , Drug Dev. Ind. Pharm. , 8 , 107 – 347 . 
115. Ragnarsson , G. , and Sjogren , J. ( 1985 ), Force displacement measurements in tableting , 
J. Pharm. Pharmacol. , 37 , 145 – 150 . 
116. Antikainen , O. K. , and Yliruusi , J. K. ( 1997 ), New parameters derived from tablet 
compression curves. Part 2. Force - displacement curve , Drug Dev. Ind. Pharm. , 23 , 
81 – 93 . 
117. Ragnarson , G. ( 1996 ), Force - displacement and network measurements , in Alderborn , 
G. , and Nystr o m , C. , Eds., Pharmaceutical Powder Compaction Technology , Marcel 
Dekker , New York , pp. 77 – 97 . 
118. Heckel , R. W. ( 1961 ), An analysis of powder compaction phenomena , Trans. Metall. 
Soc. AIME , 221 , 1001 – 1008 . 
REFERENCES 1093

1094 TABLET PRODUCTION SYSTEMS 
119. Heckel , R. W. ( 1961 ), Density - pressure relationships in powder compaction , Trans. 
Metall. Soc. AIME , 221 , 671 – 675 . 
120. Hersey , J. A. , and Rees , J. E. ( 1970 ), Deformation of particles during briquetting , paper 
presented at Proc. 2nd Particle Size Analysis Conference of the Society of Analytical 
Chemistry, Bradford, 33 . 
121. Kawakita , K. , and L u dde , K. H. ( 1970/71 ), Some considerations on powder compression 
equations , Powder Tech. , 4 , 61 – 68 . 
122. L u dde , K. H. , and Kawakita , K. ( 1966 ), Die Pulverkompression , Pharmazie , 21 , 
393 – 403 . 
123. Cooper , A. R. , and Eaton , L. E. ( 1962 ), Compaction behaviour of several ceramic 
powders , J. Am. Ceram. Soc. , 5 , 97 – 101 . 
124. Walker , E. ( 1923 ), The properties of powders. Part VII. The infl uence of the velocity of 
compression on the apparent compressibility of powders , Trans. Faraday Soc. , 19 , 
614 – 620 . 
125. Bal ’ shin , M. Y . ( 1938 ), Contribution to the theory of powder metallurgical processes , 
Metalloprom. , 18 , 124 – 147 . 
126. S o nnergaard , J. M. ( 2000 ), Impact of particle density and initial volume on mathematical 
compression models , Eur. J. Pharm. Sci. , 11 , 307 – 315 . 
127. S o nnergaard , J. M. ( 1999 ), A critical evaluation of the Heckel equation , Int. J. Pharm. , 
193 , 63 – 71 . 
128. Sun , C. , and Grant , D. J. W. ( 2001 ), Infl uence of elastic deformation of particles on Heckel 
analysis , Pharm. Dev. Technol. , 6 , 193 – 200 . 
129. Rees , J. E. , and Rue , P. J. ( 1978 ), Time - dependent deformation of some direct compression 
excipients , J. Pharm. Pharmacol. , 30 , 601 – 607 . 
130. York , P. ( 1979 ), A consideration of experimental variables in the analysis of powder 
compaction behavior , J. Pharm. Pharmacol. , 31 , 244 – 246 . 
131. Vachon , M. G. , and Chulia , D. ( 1999 ), The use of energy indices in estimating powder 
compaction functionality of mixtures in pharmaceutical tableting , Int. J. Pharm. , 177 , 
183 – 200 . 
132. Krumme , M. , Schwabe , L. , and Fr o mming , K. H. ( 2000 ), Development of computerised 
prodedures for the characterisation of the tableting properties with eccentric machines: 
Extended Heckel analysis , Eur. J. Pharm. Biopharm. , 49 , 275 – 286 . 
133. Humbert - Droz , P. , Gurny , R. , Mordier , D. , and Doelker , E. ( 1983 ), Densifi cation behavior 
of drugs having bioavailability problems , Int. J. Pharm. , 4 , 29 – 35 . 
134. Imbert , C. , Tchoreloff , P. , Leclerc , B. , and Couarraze , G. ( 1997 ), Indices of tableting performance 
and application of percolation theory to powder compaction , Eur. J. Pharm. 
Biopharm. , 44 , 273 – 282 . 
135. Kuentz , M. , and Leuenberger , H. ( 2000 ), A new approach to tablet strength of a binary 
mixture consisting of a well and a poorly compactable substance , Eur. J. Pharm. Biopharm. 
, 49 , 151 – 159 . 
136. Gabaude , C. M. D., Guillot , M. , Gautier , J. C. , Saudemon , P. , and Chulia , D. ( 1999 ), Effects 
of true density, compacted mass, compression speed, and punch deformation on the 
means yield pressure , J. Pharm. Sci. , 88 , 725 – 730 . 
137. Duberg , M. , and Nystr o m , C. ( 1986 ), Studies on the compression of tablets. XVII. Porosity 
- pressure curves for the characterization of volume reduction mechanisms , Powder 
Technol. , 46 , 67 – 75 . 
138. Paronen , P. ( 1986 ), Heckel - plots as indicators of elastic properties of pharmaceuticals , 
Drug Dev. Ind. Pharm. , 12 , 1903 – 1912 . 
139. Morris , L. E. , and Schwartz , J. B. ( 1995 ), Isolation of densifi cation regions during powder 
compression , Drug Dev. Ind. Pharm. , 21 , 427 – 446 . 

140. Kuentz , M. , Leuenberger , H. , and Kolb , M. ( 1999 ), Fracture in disordered media and 
tensile strength of microcrystalline cellulose tablets at low relative densities , Int. J. 
Pharm. , 182 , 243 – 255 . 
141. Tsardaka , K. D. , Rees , J. E. , and Hart , J. P. ( 1988 ), Compression and recovery behaviour 
of compacts using extended Heckel plots , J. Pharm. Pharmacol. , 40 , 73P . 
142. Tsardaka , K. D. , and Rees , J. K. ( 1990 ), Relations between viscoelastic parameters and 
compaction properties of two modifi ed starches , J. Pharm. Pharmacol. , 42 , 77P . 
143. Picker , K. M. ( 2000 ), A new theoretical model to characterize the densifi cation behavior 
of tableting materials , Eur. J. Pharm. Biopharm. , 49 ( 3 ), 267 – 273 . 
144. Picker - Freger , K. M. ( 2007 ), The 3 - D Model: Experimental testing of the parameters d, 
e, and w and validation of the analysis , J. Pharm. Sci. , 96 ( 5 ), 1408 – 1417 . 
145. Hoblitzell , J. R. , and Rhodes , C. T. ( 1986 ), Preliminary investigations on the parity of 
tablet compression data obtained from different instrumented presses , Drug Dev. Ind. 
Pharm. , 12 , 507 – 525 . 
146. David , S. T. , and Augsburger , L. L. ( 1977 ), Plastic fl ow during compression of directly 
compressible fi llers and its effect on tablet strength , J. Pharm. Sci. , 66 , 155 – 159 . 
147. M u ller , F. , and Caspar , U. ( 1984 ), Viskoelastische Ph a nomene w a hrend der 
Tablettierung , Pharm. Ind. , 46 , 1049 – 1056 . 
148. M u ller , F. ( 1996 ), Viscoelastic models , in Alderborn , G. , and Nystr o m , C. , Eds., Pharmaceutical 
Powder Compaction Technology , Marcel Dekker , New York , pp. 99 – 132 . 
149. Bauer , A. D. (1991), Untersuchungen zur Prozessdatengewinnung, Viskoelastizit a t und 
Struktur von Tabletten, Dissertation, Universit a t Bonn, Bonn, Germany. 
150. Staniforth , J. N. , Baichwal , A. R. , and Hart , J. P. ( 1987 ), Interpretation of creep behavior 
of microcrystalline cellulose powders and granules during compaction , Int. J. Pharm. , 40 , 
267 – 269 . 
151. Staniforth , J. N. , and Patel , C. J. ( 1989 ), Creep compliance behavior of direct compression 
excipients , Powder Technol. , 57 , 83 – 87 . 
152. Tsardaka , K. D. , and Rees , J. E. ( 1989 ), Plastic deformation and retarded elastic 
deformation of particulate solids using creep experiments , J. Pharm. Pharmacol. , 41 , 
28P . 
153. Malamataris , S. , and Rees , J. E. ( 1993 ), Viscoelastic properties of some pharmaceutical 
powders compared using creep compliance, extended Heckel analysis and tablet strength 
measurements , Int. J. Pharm. , 92 , 123 – 135 . 
154. Rees , J. E. , and Tsardaka , K. D. ( 1994 ), Some effects of moisture on the viscoelastic 
behaviour of modifi ed starch during powder compaction , Eur. J. Pharm. Biopharm. , 40 , 
193 – 197 . 
155. Picker , K. M. , and Bikane , F. ( 2001 ), An evaluation of three - dimensional modeling of 
compaction cycles by analyzing the densifi cation behavior of binary and ternary mixtures 
, Pharm. Dev. Technol. , 6 ( 3 ), 333 – 342 . 
156. Picker , K. M. ( 2004 ), The 3D model: Explaining densifi cation and deformation mechanisms 
by using 3D parameter plots , Drug Dev. Ind. Pharm. , 30 ( 4 ), 413 – 425 . 
157. Picker , K. M. ( 2003 ), The 3D model: Does time plasticity represent the infl uence of 
tableting speed? AAPS PharmSciTech. , 4 ( 4 ), article 66 . 
158. Hauschild , K. , and Picker , K. M. ( 2003 ), The 3D model: Comparison with other data 
analysis techniques for tableting , AAPS PharmSci , 5(S1), W4304. 
159. Hauschild , K. , and Picker - Freyer , K. M. ( 2004 ), Evaluation of a new coprocessed 
compound based on lactose and maize starch for tablet formulation , AAPS PharmSci , 
6 ( 2 ), article 16 . 
REFERENCES 1095

1096 TABLET PRODUCTION SYSTEMS 
160. Picker , K. M. ( 2002 ), New insights in the process of tablet formation — Ways to explore 
soft tableting , Martin - Luther - University Halle - Wittenberg and G o rich und Weiersh a user 
Verlag, Marburg, Germany. 
161. Wurster , D. E. , Rowlings , C. E. , and Creekmore , J. R. ( 1995 ), Calorimetric analysis of 
powder compression: I. Design and development of a compression calorimeter , Int. J. 
Pharm. , 116 , 179 . 
162. Picker , K. M. ( 2003 ), The relevance of glass transition temperature for the process of 
tablet formation , J. Therm. Anal. Cal. , 73 ( 2 ), 597 – 605 . 
163. Armstrong , N. A. , and Haines - Nutt , R. F. ( 1972 ), Elastic recovery and surface area 
changes in compacted powder systems , J. Pharm. Pharmacol. , 24 , 135 – 136 . 
164. Picker - Freyer , K. M. ( 2005 ), Carrageenans: Analysis of tablet formation and properties 
(Part 1) , Pharm. Technol. Eur. , 17 ( 8 ), 37 – 44 . 
165. Picker - Freyer , K. M. ( 2005 ), Carrageenans: Analysis of tablet formation and properties 
(Part 2) , Pharm. Technol. Eur. , 17 ( 9 ), 32 – 44 . 
166. Schmidt , A. G. , and Picker - Freyer , K. M. ( 2006 ), Infl uence of mechanical activation on 
the micro structure of widely used excipients for solid dosage forms , paper presented at 
the 5th World Meeting of Pharmaceutics, Pharmaceutical Technology and Biopharmaceutics, 
Geneva. 
167. Leuenberger , H. , Hiestand , E. N. , and Sucker , H. ( 1981 ), Contribution to the theory of 
powder compression , Chemie Ing. Tech. , 53 ( 1 ), 45 – 47 . 
168. Bauer - Brandl , A. ( 2002 ), Tooling for tableting , in Swarbrick , J. , and Boylan , J. C. , Eds., 
Encyclopedia of Pharmaceutical Technology , Marcel Dekker , New York . 
169. Conte , U. , Colombo , P. , Caramella , C. , Ferrari , F. , Gazzaniga , A. , Guyot , J. C. , La Manna , 
A. , and Traisnel , M. ( 1988 ), Infl uence of tablet weight control systems during 
production on biopharmaceutical properties of the tablets , Acta Pharm. Technol. , 34 ( 2 ), 
63 – 67 . 
170. Cole , G. C. ( 1998 ), Pharmaceutical Production Facilities: Design and Applications , Taylor 
and Francis , London . 
171. Lyon , R. C. , Lester , D. S. , Lewis , E. N. , Lee , E. , Yu , L. X. , Jefferson , E. H. , and Hussain , 
A. S. ( 2002 ), Near - infrared spectral imaging for quality assurance of pharmaceutical 
products: Analysis of tablets to assess powder blend homogeneity , AAPS PharmSciTech , 
3 ( 3 ), article 17 . 
172. Lai , C. K. , Zahari , A. , Miller , B. , Katstra , W. E. , Cima , M. J. , and Cooney , C. L. ( 2004 ), 
Nondestructive and on - line monitoring of tablets using light - induced fl uorescence technology 
, AAPS PharmSciTech , 5 ( 1 ), article 3 . 
173. Lee , T. H. , and Lin , S. Y. ( 2004 ), Microspectroscopic FT - IR mapping system as a tool to 
assess blend homogeneity of drug - excipient mixtures , Eur. J. Pharm. Sci. , 23 ( 2 ), 
117 – 122 . 
174. U.S. Food and Drug Administration , Center for Drug Evaluation and Research, Offi ce 
of Pharmaceutical Science, Process Analytical Technology (PAT) Initiative, assessed on 
12/20/2007, available: www.fda.gov/cder/OPS/PAT.htm . 
175. Oeser , W. H. , and Sander , A. ( 1992 ), Pharma - Betriebsverordnung: Grundregeln f u r die 
Herstellung von Arzneimitteln (GMP) , Wissenschaftliche Verlagsgesellschaft Stuttgart, 
Germany. 
176. Hyde , J. M. ( 1985 ), New developments in CIP practices , Chem. Eng. Prog. , 81 ( 1 ), 
39 – 41 . 
177. Picker , K. M. , and Mielck , J. B. ( 1998 ), Effect of relative humidity during tableting on 
matrix formation of hydrocolloids: Densifi cation behaviour of cellulose ethers , Pharm. 
Dev. Technol. , 3 ( 1 ), 31 – 41 . 

178. Hauschild , K. , and Picker - Freyer , K. M. ( 2006 ), Evaluation of tableting and tablet properties 
of Kollidon SR: The infl uence of moisture and mixtures with theophylline monohydrate 
, Pharm. Technol. Dev. , 11 ( 1 ), 125 – 140 . 
179. Kachrimanis , K. , Nikolakakis , I. , and Malamataris , S. (2003), Tensile strength and disintegration 
of tableted silicifi ed microcrystalline cellulose: Infl uences of interparticle 
bonding , J. Pharm. Sci. , 92 ( 7 ), 1489 – 1501 . 
180. Steendam , R. , Eissens , A. C. E. , Frijlink , H. W. , and Lerk , C. F. ( 2000 ), Plasticization of 
amylodextrin by moisture. Consequences for drug release from tablets , Int. J. Pharm. , 
204 ( 1 – 2 ), 23 – 33 . 
181. Hanssen , D. , F u hrer , C. , and Schaefer , B. ( 1970 ), Evaluation of magnesium stearate as 
tablet lubricating agent by electronic pressure measurements , Pharm. Ind. , 32 ( 2 ), 
97 – 101 . 
182. Bolhuis , G. K. , and Lerk , C. F. ( 1977 ), Film forming of tablet lubricants during the mixing 
process of solids , Acta Pharm. Technol. , 23 ( 1 ), 13 – 20 . 
183. Zuurman , K. , Van der Voort Maarschalk , K. , and Bolhuis , G. K. ( 1999 ), Effect of magnesium 
stearate on bonding and porosity expansion of tablets produced from materials 
with different consolidation properties , Int. J. Pharm. , 179 ( 1 ), 107 – 115 . 
184. Van Der Voort Maarschalk , K. , and Bolhuis , G. K. ( 1998 ), Improving properties of 
materials for direct compaction. Part II , Pharm. Technol. Eur. , 10 ( 10 ), 28 – 36 . 
185. Shotton , E. , and Lewis , C. J. ( 1964 ), Effect of lubrication on the crushing strength of 
tablets , J. Pharm. Pharmacol. , 16S , 111 – 120 . 
186. Van Der Voort Maarschalk , K. , and Bolhuis , G. K. ( 1999 ), Improving properties of 
materials for direct compaction , Pharm. Technol. , 23 ( 5 ), 34 – 46 . 
187. Steffens , K. J. , Mueller , B. W. , and List , P. H. ( 1982 ), Tribological laws and results in tablet 
production. 7. Studies on magnesium stearate commercial products , Pharm. Ind. , 44 ( 8 ), 
826 – 830 . 
188. Hoelzer , A. W. ( 1984 ), Batch to batch variations of commercial magnesium stearates. 
Chemical, physical and lubricant properties , Labo - Pharma , 338 , 28 – 36 . 
189. Miller , T. A. , and York , P. ( 1985 ), Frictional assessment of magnesium stearate and palmitate 
lubricant powders , Powder Technol. , 44 ( 3 ), 219 – 226 . 
190. Lindberg , N. O. ( 1972 ), Evaluation of some tablet lubricants , Acta Pharm. Suec. , 9 ( 3 ), 
207 – 214 . 
191. Alpar , O. , Deer , J. J. , Hersey , J. A. , and Shotton , E. ( 1969 ), Possible use of poly(tetrafl uo 
roethylene) (Fluon) as a tablet lubricant , J. Pharm. Pharmacol. , 21S , 6 – 8 . 
192. Shah , N. H. , Stiel , D. , Weiss , M. , Infeld , M. H. , and Malick , A. W. ( 1986 ), Evaluation of 
two new tablet lubricants — Sodium stearyl fumarate and glyceryl behenate. Measurement 
of physical parameters (compaction, ejection and residual forces) in the tableting 
process and the effect of the dissolution rate , Drug Dev. Ind. Pharm. , 12 ( 8 – 9 ), 
1329 – 1346 . 
193. N’Diaye , A. , Jannin , V. , Berard , V. , Andres , C. , and Pourcelot , Y. ( 2003 ), Comparative 
study of the lubricant performance of Compritol HD5 ATO and Compritol 888 ATO: 
effect of polyethylene glycol behenate on lubricant capacity , Int. J. Pharm. , 254 ( 2 ), 
263 – 269 . 
194. Staniforth , J. N. ( 1987 ), Use of hydrogenated vegetable oil as a tablet lubricant , Drug 
Dev. Ind. Pharm. , 13 ( 7 ), 1141 – 1158 . 
195. Dawoodbhai , S. S. , Chueh , H. R. , and Rhodes , C. T. ( 1987 ), Glidants and lubricant properties 
of several types of talcs , Drug Dev. Ind. Pharm. , 13 ( 13 ), 2441 – 2467 . 
196. Roescheisen , G. , and Schmidt , P. C. ( 1995 ), Preparation and optimization of l -leucine as 
lubricant for effervescent tablet formulations , Pharm. Acta Helv. , 70 ( 2 ), 133 – 139 . 
REFERENCES 1097

1098 TABLET PRODUCTION SYSTEMS 
197. Jahn , T. , and Steffens , K.-J. ( 2005 ), Press chamber coating as external lubrication for high 
speed rotary presses: Lubricant spray rate optimization , Drug Dev. Ind. Pharm. , 31 ( 10 ), 
951 – 957 . 
198. Laich , T. , and Kissel , T. ( 1998 ), Automatic adaptation of lubricant quantity by control of 
an external lubrication. Tests on a reciprocating and on a rotary tablet press , Pharm. 
Ind. , 60 ( 10 ), 896 – 904 . 
199. Otsuka , M. , Sato , M. , and Matsuda , Y. ( 2001 ), Comparative evaluation of tableting compression 
behaviors by methods of internal and external lubricant addition: Inhibition of 
enzymatic activity of trypsin preparation by using external lubricant addition during the 
tableting compression process, AAPS PharmSci , 3 ( 3 ), article 20 . 
200. Jetzer , W. , and Leuenberger , H. ( 1984 ), Determination of capping tendency in pharmaceutical 
active ingredients and excipients , Pharm. Acta Helv. , 59 ( 1 ), 2 – 7 . 
201. Bauer , K. H. , Fr o mming , K. H. , and F u hrer , C. ( 2002 ) Lehrbuch der pharmazeutischen 
Technologie , Wissenschafteiche Verlagsgesellschaft, Stuttgart, Germany. 

1099 
6.5 
CONTROLLED RELEASE OF DRUGS 
FROM TABLET COATINGS 
Sacide Alsoy Altinkaya 
Izmir Institute of Technology, Urla - Izmir, Turkey 
Contents 
6.5.1 Introduction 
6.5.2 Tablet Coating Methods 
6.5.3 Characterization of Tablet Coatings 
6.5.4 Preparation of Asymmetric Membranes 
6.5.5 Methods for Optimization of Tablet Coating Formulations 
6.5.6 Applications 
6.5.6.1 Materials and Methods 
6.5.6.2 Results and Discussion 
6.5.7 Conclusion 
References 
Appendix 
6.5.1 INTRODUCTION 
Controlled - release technology for drug delivery applications is designed to target 
the drug to particular places or cells in the body, to overcome certain tissue and 
cellular barriers, and to control the duration and level of the drug in the body within 
a spefi cied therapeutic window. This usually implies achieving a prolonged, zero - 
order release rate over the desired duration of drug delivery. Controlled - release 
dosage forms provide sustained drug release and require less frequent drug loading 
than conventional forms. Thus, the toxic side effects of the drug are minimized and 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

1100 CONTROLLED RELEASE OF DRUGS FROM TABLET COATINGS 
patient ’ s convenience and compliance are improved. Controlled - release systems are 
usually classifi ed into four categories based on the rate - limiting step of the release 
process [1] : (1) diffusion - controlled systems, (2) chemically controlled systems, 
(3) swelling - controlled systems, and (4) magnetically controlled systems. Diffusion - 
controlled systems are formulated in two main geometries: reservoirs and matrices. 
In matrix systems, the drug is generally uniformly distributed or dissolved throughout 
a polymer. The release kinetics from these types of systems depend on the 
quantity and the diffusivity of drug in the matrix and the geometry of the system. 
The release rates from matrix systems usually decrease with time due to an increase 
in the path length for diffusion of drug and, thus, their release characteristics are 
not generally zero order. In the case of chemically controlled systems, the drug is 
either distributed through a biodegradable polymer or chemically bound to a 
polymer backbone chain. The drug release from the biodegradable polymer is controlled 
by degradation of the polymer through penetration of water or a chemical 
reaction [1] . The drug attached to the polymer is released by hydrolytic or enzymatic 
cleavage. In swelling - controlled systems, the drug is dissolved or distributed in a 
glassy polymer matrix. As water penetrates the dry matrix, the polymer swells and 
its glass transition temperature decreases below the temperature in the matrix. As 
a result, the glassy – rubbery transition occurs and it allows the release of the drug. 
The release rate of the drug out of these systems is mainly controlled by the rate 
and degree of swelling. Alternatively, the drug release can also be controlled magnetically 
by dispersing the drug and small magnetic beads within a polymer matrix 
and exposing them to an oscillating external magnetic fi eld. (The drug is released 
as the matrix is exposed to an oscillating external magnetic fi eld.) 
Reservoir systems consist of a drug - containing core surrounded by a polymer 
membrane. The release rate of drug is controlled by its diffusion through the membrane 
[1, 2] . In addition to diffusional release, osmotic pumping mechanisms 
contribute to the total drug release rate if either the drug is highly soluble or an 
osmotic agent is added to the active core [1 – 4] . Reservoir systems are prepared in 
different geometries, such as coated tablets, beads, particles, membrane - based 
pouches, and microcapsules. The main advantage of these systems is their ability 
to maintain zero - order release rates [1, 2] . This is usually achieved by loading the 
powdered form of drug at a level far above the solubility of drug. Then, the concentration 
of drug at the internal wall of the reservoir becomes its saturation 
concentration and zero - order release occurs until the drug is completely dissolved. 
In addition, drug loading can be higher for these systems compared to other controlled 
- release systems; thus, the cost of formulation is minimized and the drug is 
released at a higher rate for a longer period of time. The major disadvantage of 
the reservoir system is rupture of the rate - controlling membrane if it is subject to 
dose dumping. 
Numerous studies exist in the literature on the drug release from tablet coatings. 
In the majority of these studies, diffusional drug release takes place from the coatings 
[5 – 17] . Various factors such as the type [8, 10, 15] , and concentration of the 
coating material [7, 8, 11] , the type and amount of the plasticizer in the coating 
solution [10, 12, 15] , the thickness of the coating [5] , the composition of the tablet 
core [6, 14] , the particle size of the coating material [10, 18] , and the weight gain of 
the coating [7, 8, 11, 13, 16] were considered during the formulation of tablet coat

ings, and their effects on the drug release rates were investigated. The use of osmotic 
tablet coating systems are also described in the literature [3, 4, 17, 19 – 23] . Osmotic 
systems utilize osmotic pressure difference as a driving force, and in the simplest 
design they consist of an osmotic core containing drug with and without osmotic 
agents. Different studies have shown that the release rate of drugs from oral osmotic 
pump tablet coating systems is governed by various formulation variables such as 
solubility and osmotic pressure of core components [3, 4, 19 – 23] , number [17] and 
size of the delivery orifi ce [3, 4, 20, 21, 23] , drug loading [3, 4, 17] thickness of the 
coating [3, 4, 19, 21, 23] , composition of the coating solution [3, 4, 17, 20, 21, 23] , and 
weight of the coating [17, 20, 22] . The coatings prepared in most of these studies 
have dense structures with or without a hole drilled through the coating through 
which the drug is delivered. In some cases, drug delivery ports are formed by adding 
leachable materials to the coating [24, 25] . The main problem with these systems, in 
the absence of a hole, is an excessively prolonged drug release due to the low permeability 
of the coating. To increase permeability of the coatings, plasticizers and 
water - soluble additives were incorporated in the coating solution, and multilayer 
composite coatings [14, 15, 24, 26, 27] or multiple - compartment osmotic tablets were 
prepared [17, 22, 28, 29] . The permeability of the tablet coating systems were further 
improved by changing the structure of the coatings from dense to porous asymmetric 
ones [30 – 42] . Asymmetric membrane tablet coatings are characterized by 
a relatively thin, dense skin layer supported on a highly permeable, thicker, and 
porous sublayer. The permeabilities and the release rates of the drugs through the 
asymmetric - membrane capsule/tablet coatings were determined to be higher compared 
to conventional dense tablet coatings [30, 33, 34, 39] . The composition of the 
coating solution was found to have a signifi cant effect on the structure of these type 
of coatings and thus on the release rate of the drug [30, 33, 35, 36, 39, 40 – 42] . In 
addition, it was reported that the drying condition had a signifi cant impact on the 
structure of the asymmetric - membrane tablet coating and the release rate of the 
drug [41] . A review of the literature studies clearly indicates that asymmetric - type 
tablet coating is a new solution for developing controlled drug delivery systems, 
since the structure of these types of coatings can be varied easily by changing the 
preparation conditions without altering the coating material or signifi cantly varying 
the coating thickness. 
This work contains six sections in addition to the introduction. Sections 6.5.2 – 
6.5.5 present a brief review of manufacturing methods for the application of coating 
materials on tablets, characterization methods used to evaluate the uniformity and 
defects of the tablet coatings, the techniques commonly used for manufacturing 
asymmetric - type membranes, and modeling approaches employed for optimization 
of the tablet coating formulations. The aim of the last two sections is to demonstrate 
the advantages of asymmetric membrane tablet coatings with respect to their drug 
release properties and the factors that affect the morphology of these types of tablet 
coatings. To achieve this goal, the in vitro release of a model compound, theophylline, 
from asymmetric membrane tablet coatings is determined and the morphology 
of the coatings is examined. In addition, the dynamics of the phase inversion is 
quantifi ed in terms of ternary - phase diagrams coupled with composition paths 
determined from a mathematical model developed previously by our group [43] . 
To draw meaningful and objective conclusions from experimental data and derive 
INTRODUCTION 1101

1102 CONTROLLED RELEASE OF DRUGS FROM TABLET COATINGS 
an empirical expression for the release rate of drug, compositions of the coating 
solution are chosen using a statistical experimental design. 
6.5.2 TABLET COATING METHODS 
Four basic methods are commonly used for the application of coating materials on 
drug tablets: (1) pan coating, (2) fl uidized - bed coating, (3) compression (press) 
coating, and (4) melt/dry coating. 
Pan Coating The pan coating process is the oldest form of pharmaceutical coating 
for manufacturing small, coated particles or tablets. In this process, the particles 
are tumbled in a pan which is rotated at an angle of usually 45 ° to the horizontal 
surface at a speed between 20 and 50 rpm [44] . Coating fl uid is sprayed onto the 
particles from a above by means of an air jet. The hot air blown through the coater 
evaporates the fl uid and dries the fi lm coating. Pan coating is generally preferred 
to coat large tablets since they are exposed to mechanical damage in other coating 
operations. The uniformity of the coating applied to the tablets and defects on the 
coating are important issues from a practical point of view. Operating conditions 
such as drum speed, drum solids loading, the presence/absence of baffl es, air velocity, 
and temperature infl uence the movement of tablets within the moving bed, the 
circulation and surface exposure times spent by a tablet in the bulk of the moving 
bed and on the surface of the bed, respectively, and the rate of drying, which all in 
turn determine the uniformity of the coating in a pan coating process [45] . Different 
experimental tools such as light emission from a single luminous tablet, photographic 
and manual counting, positron emission particle tracking, magnetic resonance 
imaging, and near real - time video imaging techniques [45 – 51] were used to 
study the motion of tablets in the drum. Simulation of particle movement using 
discrete - element modeling has also been used to study the movement of tablets 
[52 – 55] . In addition, statistical experimental design was utilized to identify the critical 
processing variables that affect coating uniformity and loading of active agent 
coated on tablets [56] . Inlet airfl ow, pan speed, inlet air temperature, coating time, 
atomization pressure, and fan pressure were investigated as the process variables. 
Among these variables, atomization pressure, pan speed, and duration of coating 
were found to be critical process variables with respect to uniformity of the 
coating. 
Fluidized - Bed Coating The basic principle of fl uidized - bed coating is to suspend 
tablets in a moving hot - air stream in the upward direction during the coating 
process. The coating material is sprayed through a nozzle from the top, the side, or 
the bottom into the fl uidized bed. The solvent in the solution is removed from 
the coating by the hot - air stream, which also carries the coated tablets/particles. 
Fluidized - bed coating provides better coating uniformity due to good solid – fl uid 
mixing and minimizes formation of agglomerates. There are various types of 
fl uidized - bed coating equipment, the most commonly used confi guration being the 
Wurster column coater [44] , in which a draft tube insert is placed coaxially in the 
bed to aid the circulation of particles. This column is not suitable for coating of large 
particles and tablets due to high erosion of solids associated with the higher velocity 

needed to circulate them [57] . Typical operating variables that affect the performance 
of the fl uid bed coating in a Wurster column are airfl ow rate, air and bed 
temperature, spray rate, gap height between the draft tube and the air distributor 
plate, atomizing air pressure, humidity, and solids charge. The product coating uniformity 
in the Wurster column coating process is primarily determined by the variation 
in coating per pass and the circulation time distribution. Radioactive particle 
tracing, magnetic particle tracing, and controlled single - or multiple - pass coating 
evaluation techniques were commonly used for detecting particle circulation time 
and distribution [57, 58] . In addition, several models have been developed to predict 
the amount of material coated on the particles [59 – 65] . 
Compaction Coating In this process, the coating is compressed around a preformed 
core by using a special tablet press. Compaction coating is especially useful 
when the drug itself is unstable in the solution and precipitates from the solution 
in a less stable morphology. On the other hand, the large amount of coating material 
required limits the applicability of this technology. Another disadvantage of the 
process is poor adhesion at the coating – core interface which causes physical instability 
(i.e., friability) [66] . The Press coating process is not useful for coating relatively 
hard cores which provide essentially no compressibility. 
Melt/Dry Coating Film coating processes require water or organic solvent(s). The 
use of organic solvents causes environmental pollution and excessive cost of recovery 
while a long time is required to remove the aqueous solvent. Both hot - melt and 
dry coating techniques eliminate the use of solvents; as a result the processing times 
become much shorter and the cost of the process is reduced. Melt coating is possible 
for coating materials that have a low melting temperature and acceptable thermostability. 
The principal stages in the fi lm formation during dry coating are softening, 
melting, and curing [67 – 70] . The process requires larger amount of plasticizer to 
partially soften and dissolve the polymer. 
6.5.3 CHARACTERIZATION OF TABLET COATINGS 
Tablet coatings are applied to improve tablet swallowability, mask unpleasant 
tastes and odors, protect the tablet core against water and oxygen, which can 
degrade the drug in the core, and control the release rate of the drug. The rate of 
dissolution and bioavailability of a drug are primarily infl uenced by the quantity 
and quality of the coating applied on the tablet. Thus, the characterization of the 
coatings becomes essential in terms of the uniformity and integrity of the coating. 
A number of instrumental methods ranging from liquid chromatography [71] to 
various noninvasive spectroscopic probes have been introduced and evaluated as 
a means of monitoring the coating process. Among these methods, laser - induced 
breakdown spectroscopy (LIBS) has been used as a rapid technique for assessing 
the uniformity of the coating thickness [72] ; however, the destructive nature of the 
method has also been reported [73] . Near - infrared spectroscopy has been employed 
for determining the amount of coating applied [74 – 76] . The main disadvantage of 
this method has been identifi ed as imprecision in the calibration and validation 
models due to uneven distribution of coating from tablet to tablet [77] . Microscopy 
CHARACTERIZATION OF TABLET COATINGS 1103

1104 CONTROLLED RELEASE OF DRUGS FROM TABLET COATINGS 
techniques provide direct and accurate measurement of the coating thickness but 
require laborious sample preparation and are therefore impractical for real - time 
process monitoring. Recently, Raman spectroscopy combined with multivariate 
data analysis has been reported as a feasible noninvasive technique to quantify 
tablet coating thickness and uniformity in the presence of a fl uorescent ingredient 
in the coating formulation [77, 78] . Confocal laser scanning microscopy has been 
introduced as a novel technique for imaging the fi lm – core interface and surface 
defects of fi lm - coated tablets [79] . Surface roughness was also determined as an 
important factor in characterizing the tablet coatings using different imaging and 
roughness analytical techniques, including optical microscopy, scanning electron 
microscopy (SEM), laser profi lometry, and atomic force microscopy (AFM) 
[80 – 84] . 
6.5.4 PREPARATION OF ASYMMETRIC MEMBRANES 
Asymmetric membranes are usually produced by phase inversion techniques. In 
these techniques, an initially homogeneous polymer solution becomes thermodynamically 
unstable due to different external effects and the phase separates into 
polymer - lean and polymer - rich phases. The polymer - rich phase forms the matrix of 
the membrane, while the polymer - lean phase, rich in solvents and nonsolvents, fi lls 
the pores. Four main techniques exist to induce phase inversion and thus to prepare 
asymmetric porous membranes [85] : (a) thermally induced phase separation (TIPS), 
(b) immersion precipitation (wet casting), (c) vapor - induced phase separation 
(VIPS), and (d) dry (air) casting. 
Thermally Induced Phase Separation In the TIPS process, an initially homogeneous 
solution consisting of a polymer and solvent(s) phase separates due to a 
decrease in the solvent quality when the temperature of the casting solution is 
decreased. After demixing is induced, the solvent is removed by extraction, evaporation, 
or freeze drying. 
Immersion Precipitation (Wet Casting) A homogeneous polymer solution consisting 
of a polymer and solvent(s) is cast on a support and is immersed in a nonsolvent 
bath. During the immersion, casting solvent diffuses into the nonsolvent 
bath and, countercurrently, nonsolvent in the bath penetrates into the solution. The 
nonsolvent has a limited solubility in the polymer, and when it reaches its critical 
concentration in the solution, precipitation takes place. Then, the solvent and nonsolvent 
in the solution are extracted and fi lm is annealed. 
Vapor - Induced Phase Separation During the VIPS process, phase separation 
is induced by penetration of nonsolvent vapor, into the homogeneous polymer 
solution consisting of polymer and solvent(s). Mass transfer is usually much 
slower than that in the wet casting process; thus, the VIPS process has been 
used to obtain membranes with symmetric, cellular, and interconnected pores 
[86, 87] . 
Dry (Air) Casting In this process, the polymer dissolved in a mixture of a volatile 
solvent and a less volatile nonsolvent is cast on a support and exposed to an air 

APPLICATIONS 1105 
stream. During drying of the solution, fast solvent evaporation leads to a decrease 
in solubility of the polymer, then phase separation takes place. 
6.5.5 METHODS FOR OPTIMIZATION OF TABLET 
COATING FORMULATIONS 
Tablet coating formulation is composed of various formulation factors and process 
parameters. The formulation is usually divided into modeling and optimization 
phases. The modeling phase consists of preparing series of experimental formulations 
by varying the ingredients and process conditions systematically and measuring 
their properties. A detailed review of the literature indicates that the response 
surface method (RSM) has been widely used for modeling and choosing acceptable 
tablet coating and other pharmaceutical formulations. The RSM includes statistical 
experimental designs, multiple regression analysis, and optimization algorithms to 
search the best formulation for a given set of constraints. Full factorial, orthogonal, 
Box – Behnken, central composite, pseudorandom, and Plackett – Burman designs 
were used to investigate the effects of tablet core formulation, coating thickness, 
and process parameters such as mixing time and speed in the pan coating process, 
inlet airfl ow, pan speed, inlet air temperature, coating time, atomization pressure, 
and fan pressure in the fl uidized - bed coating process [88 – 92] . Artifi cal neural networks 
(ANNs) have also been investigated as an alternative method for modeling 
the pharmaceutical formulations [93 – 105] . In the RSM, the pharmaceutical responses 
are predicted based on the second - order polynomial equation, which is usually 
limited to low levels. When a nonlinear relationship between formulation factors 
and response variables is observed, the ANN approach was shown to give better 
estimations of optimal formulations [105] . 
6.5.6 APPLICATIONS * 
Previous sections have reviewed numerous studies in the literature investigating the 
release mechanisms from the tablet coating systems, their advantages/disadvantages 
as a control release system, and the methods used to characterize and optimize their 
formulation. In the following sections, the in vitro performance characteristics of 
the asymmetric membrane tablet coatings will be illustrated using release studies 
of a model compound theophylline. For this purpose, fi rst the method used for 
preparing asymmetric membrane tablet coatings will be explained, then the results 
of the dissolution studies will be discussed. 
6.5.6.1 Materials and Methods 
Preparation and Characterization of Tablet Coatings Tablet cores were prepared 
by compressing the drug without any excipient using a hydraulic press operated 
at 110 MPa. A stainless steel die with a diameter of 1.2 cm was used to produce 
400 - mg drug tablet cores. Asymmetric - membrane tablet coatings were applied 
* This article was published in Biochemical Engineering Journal, 28, Sacide Alsoy Altinkaya, 
Hacer Yenal, In vitro drug release rates from asymmetric - membrane tablet coatings: Prediction 
of phase - inversion dynamics, 131 – 139, Copyright Elsevier (2006). 

1106 CONTROLLED RELEASE OF DRUGS FROM TABLET COATINGS 
using a dip coating process (Dip Coater Nima, type D1L, serial no. 327). The tablets 
were dip coated in polymer solutions prepared by dissolving cellulose acetate 
(Aldrich) in a solution of acetone (Merck) and water. The rate of withdrawal of 
the tablets from the solution was adjusted to obtain similar fi nal coating thicknesses 
for each coating formulation. Immediately after coating, tablets were rotated for 
even distribution of the viscous membrane solution, transferred into an environmental 
chamber (Angelantoni Industrie, Italy, Challenge series, model number 
CH250), and kept there for 2 h to allow for evaporation of both the solvent (acetone) 
and nonsolvent (water). The temperature and relative humidity of air in the environmental 
chamber were adjusted to 25 ° C and 50%, respectively. Tablets were 
allowed to dry further for a minimum of 24 h at room temperature prior to dissolution 
experiments. Faster evaporation of acetone and resulting increase in the concentration 
of water in the coating leads to a decrease in solubility of the cellulose 
acetate (CA); then phase separation takes place. Consequently, asymmetric - membrane 
structure forms on the tablet core. Morphology of the coatings was examined 
using a scanning electron microscope (Philips, XL - 30SFEG). Samples were 
coated with gold palladium using a magnetron sputter coating instrument. The 
thickness of the dense skin layer, the overall porosity, and the average pore size 
were determined from image analysis of micrographs showing cross sections of the 
membranes. 
Dissolution Studies The release rate of theophylline from the tablets was determined 
by the U.S. Pharmacopeia (USP) XXIII dissolution methodology using a 
dissolution tester (Caleva 10ST). According to this standard, 900 ml of dissolution 
medium was placed in the vessel and the temperature was maintained constant at 
37 ° C using a constant - temperature bath. Then, the tablets were immersed in the 
vessel and the solution was stirred at a speed of 50 rpm. To simulate the actual 
dissolution environment in the body, the pH of the dissolution medium was 
kept at 3 during the fi rst 3.5 h by adding 8.5 vol % phosphoric acid to 900 mL distilled 
water and then increased to 7.4 by adding 5.3 M NaOH to the dissolution 
medium and kept at this value until the end of the experiment. To determine the 
quantity of drug released from the tablets, samples were taken periodically and 
assayed by ultraviolet (UV) spectrophotometry (Shimadzu UV - 1601) at a wavelength 
of 272 nm. Dissolution experiments were performed on three tablets and 
the release profi les were reported as the arithmetic average of the three experimental 
runs. 
Statistical Experimental Design To determine the infl uence of the composition 
of the coating solution on the release rate of drug, experiments were statistically 
designed using a commercial software package called Design - Expert [106] . The 
system studied in this chapter consists of three components with the following compositional 
restrictions: 
5 15 70 90 5 15 1 2 3 . . . . . . . . . (1) 
where . i is the weight percent of component i and 1, 2, 3 represent CA, acetone, 
and water, respectively. Any composition outside these limits will probably fail to 

APPLICATIONS 1107 
produce a successful asymmetric - membrane coating. In mixture experiments, the 
factors are the compositions of the mixture components, and the sum of the fractions 
of all components is equal to 1. Therefore the factor levels are mutually dependent. 
Thus, factorial experimental designs are not suitable for response surface modeling 
of mixtures since such designs require that the experimental treatment combinations 
be determined by independent adjustments of each component level. In addition, 
a standard response surface design cannot be used either due to the same 
constraints. Consequently using the constraint levels shown in Equation (1) , a D - 
optimal design was generated by the Design - Expert software package. The 14 experimental 
formulations determined are shown in Table 1 . The lower and upper limits 
on the weight fraction of each component are required to (a) obtain appropriate 
viscosity of the solution and coat the tablets uniformly and (b) induce phase separation, 
thus forming a porous membrane structure. These constraints were established 
based on preliminary dissolution experiments, available literature data, and the 
simulation results reported by Altinkaya and Ozbas [43] . 
Of the 14 formulations listed in Table 1 , six experimental runs were required to 
fi t the quadratic mixture model, four additional distinct runs were used to check for 
the lack of fi t, and fi nally four runs were replicated to provide an estimate of pure 
error. Design - Expert used the vertices, the edge centers, the overall centroid, and 
one point located halfway between the overall centroid and one of the edge centers 
as candidate points. Additionally, four vertices of the design region were used as 
check points [106] . 
Determination of Phase Diagrams and Composition Paths The dynamics of the 
membrane formation process is predicted by combining the kinetics and thermody- 
TABLE 1 Theophylline Release Rates from Asymmetric - Membrane Tablet Coatings, 
Results of Fitting Release Profi les to Zero - Order Kinetics, and Precipitation Times 
Determined from Model Predictions 
Compositon (wt %) 
Release Rate 
(mg/min) R 2 
Precipitation 
Time a (s) 
Cellulose 
Acetate Acetone Water 
15 80 5 0.036 0.9871 2671/2671 
15 80 5 0.036 0.9864 2671/2671 
5 90 5 0.45 0.9757 660/660 
5 90 5 0.36 0.9864 660/660 
15 70 15 0.036 0.9908 3374/3374 
15 70 15 0.027 0.9925 3374/3374 
5 80 15 0.36 0.9928 1000/1000 
5 80 15 0.36 0.9876 1000/1000 
5 85 10 0.27 0.9918 675/675 
10 80 10 0.054 0.9958 1751/1751 
15 75 10 0.054 0.9975 2314/2314 
10 85 5 0.036 0.9887 2554/2554 
12.5 77.5 10 0.036 0.9902 — 
10 75 15 0.063 0.9889 1484/1484 
a The fi rst number corresponds to the precipitation time at the tablet – coating interface. 

1108 CONTROLLED RELEASE OF DRUGS FROM TABLET COATINGS 
namics of the system simultaneously. An appropriate thermodynamic model is 
necessary to construct the ternary - phase diagram and to formulate the boundary 
conditions of the kinetic model. Phase separation is considered to occur when a 
mass transfer path touches the binodal curve. In this study, a robust algorithm developed 
previously by our group was used to construct the phase diagram [43] . The 
algorithm utilizes Flory Huggins thermodynamic theory with constant interaction 
parameters. The composition paths were determined from the kinetic model equations 
which consist of coupled unsteady - state heat and mass transfer equations, fi lm 
shrinkage, and complex boundary conditions. The details of both the thermodynamics 
and kinetic equations can be found in our previous study [43] . We have assumed 
that the kinetic model derived for a plane geometry can be used to predict the 
membrane formation process on a tablet surface. This assumption is fairly reasonable 
since the thickness of the coating is very small and, thus, the cylindrical geometry 
can be approximated as the plane geometry. 
6.5.6.2 Results and Discussion 
Effect of Composition of Coating Solution The effect of changing the composition 
of the casting solution is well documented for asymmetric membranes prepared 
for separation applications [107 – 114] . However, there are relatively few quantitative 
studies illustrating the relationship between the composition of the casting 
solution and the drug release rate from the asymmetric - membrane coated tablets/ 
capsules [30, 33] . To investigate such a relationship, the in vitro release profi les of 
the model drug theophylline were measured for the 14 formulations listed in Table 
1 and they are shown in the Appendix in Figures A1 – A10 . To fi nd out whether the 
drug release from the tablet coatings provides a zero - order release kinetics, each 
data set was fi tted to a linear equation. The quality of the fi tted model is determined 
by the coeffi cient of determination R 2 and it is defi ned as 
R y y 
y 
2 = 
. . . 
. 
. (2) 
where 
.y i 
i 
n 
n 
Y Y = . 
= . 
1 2 
1
( ) 
(3) 
. ( . ) .y i i 
i 
n 
n 
Y Y = . 
= . 
1 2 
1 
(4) 
denote the sample variance and the prediction error power, respectively. Additionally, 
n is the number of experimental data points, Y i is the experimental observation, 
Y is the average of the experimental data points, and .Y
i denotes the predicted value 
by the fi tted model. The quantity R 2 lies between 0 and 1, and if the value is 1, it 
can be said that the fi t of the model is perfect. High R 2 values listed in Table 1 for 
each data set indicate that there is an excellent linear relationship between the 
concentration of the drug and the release time; thus, all tablet coatings prepared 

APPLICATIONS 1109 
can provide zero - order or near - zero - order drug release. The release rates for each 
coating formulation were estimated from the slope of the average release profi les 
and they are also listed in Table 1 . 
The drastic change in the release rates with the composition of the coating solution 
is shown in Figure 1 . Within the experimental composition range covered, the 
highest release rate was observed in the case of the lowest CA (P: polymer) and the 
highest acetone (S: solvent) concentrations in the casting solution [polymer (P) – 
solvent (S) weight fraction ratio 5/90]. This is caused by the fi nal coating structure 
consisting of a very thin and dense top skin layer, and highly porous lower sublayer, 
as illustrated in Figure 2 . At the lowest level of polymer concentration (5%), the 
FIGURE 1 Release rate of theophylline as function of cellulose acetate(P) – acetone(S) 
weight fraction ratios. 
FIGURE 2 SEM of cross section of asymmetric - membrane made with 5% water and 
CA – acetone weight ratio of 5/90, magnifi cation 2500 . . 

1110 CONTROLLED RELEASE OF DRUGS FROM TABLET COATINGS 
thicknesses of the dense skin layers of the coatings are very small and similar to 
each other; thus, the structure of the lower sublayer becomes an important factor 
in determining the release rate of the drug. The porosity of the coating structure in 
the case of 90% acetone is so high that an increase in water concentration from 5 
to 10% is not suffi cient to produce a more porous fi nal structure and, thus, the 
release rate of the drug decreases. A further increase of water concentration to 15% 
results in an opposite effect, which makes the release rate increase to a level of 
0.36 mg/min due to the dominant effect of water concentration in increasing the 
porosity of the lower sublayer. 
An increment in the CA concentration from 5 to 10% resulted in signifi cantly 
lower release rates at all water concentrations because the structure of the coatings 
changed from porous to dense. Keeping the polymer concentration at 10% while 
changing the water concentration from 5 to 15% makes the release rates increase. 
This behavior is explained by the formation of more porous structures by adding 
more nonsolvent into the casting solution, which is in agreement with the observations 
of other groups [30, 33] . 
When the polymer concentration increases from 10 to 15%, no signifi cant 
changes in release rates were observed. As a matter of fact, in the cases of 5% 
and 10% water concentrations, the release rates did not change at all. This is due 
to the unusual transport characteristics of asymmetric membranes which are 
complex functions of the properties of the different regions of the membrane. In 
addition to the thickness of the dense skin layer and the porosity of the lower 
sublayer, structural factors such as tortuosity, pore size, shape, and connectivity 
of the pores also strongly affect the rate of transport through the coating. The 
SEM pictures taken at high magnifi cation which are shown in Figures 3 a and 3 b 
indicate that the tablet coating prepared with a P/S ratio of 10/85 has a uniform 
and narrow pore size distribution with regular elliptic pore shapes, while the 
other one (P/S: 15/80) has cylindrical pores with a wide pore size distribution, 
forming a connected pore network. As a result, even though the resistance of the 
dense skin layer of the coating made with P/S ratio of 15/80 is larger, its lower 
sublayer resistance is smaller due to the connected pore network. Consequently, 
the release rate of the theophylline becomes the same through both tablet coatings. 
Comparison of the scanning electron micrographs in Figures 4 a and 4 b indicates 
that the tablet coating made with the P/S ratio of 10/80 has a uniform pore 
size distribution, cylindrical pore shapes and high tortuosity while the tablet 
coating prepared with the P/S ratio of 15/75 has elliptic, irregular pore shapes 
and pores are isolated. Thus, the lower sublayer resistance of the coating made 
with the P/S ratio of 10/80 is larger due to the relatively higher tortuosity resulting 
in the same release rate with the coating prepared with the P/S ratio of 
15/75. 
At the 15% polymer concentration level, the release rate increases with the 
change in water concentration from 5 to 10%, which is mainly caused by the 
increased porosity. However, a further 5% increment in the water concentration 
(to 15%) made the release rate decrease back to the same level as in the case of 
5% water concentration. Even though higher water concentration favors forming 
a more porous structure and a concomitant higher release rate, the acetone level 
which decreased below a critical value destroyed this mechanism. 

APPLICATIONS 1111 
Results of dissolution studies along with morphological observations clearly 
indicate that the drug release rate is strongly infl uenced by the morphology of 
the membrane. Thus, if one wishes to control the drug release characteristics of the 
delivery system, a quantitative understanding of the dynamics and morphology of 
the phase inversion process is required. The dynamics of the phase inversion process 
can be quantifi ed in terms of the ternary - phase diagram coupled with the heat and 
mass transfer model equations. We have obtained information about the structure 
of the tablet coating by plotting the composition paths on the ternary - phase diagram 
and the polymer concentration profi le at the moment of precipitation. As an illustra- 
FIGURE 3 SEM of cross section of asymmetric - membrane made with 5% water and 
CA/acetone weight ratios of: ( a ) 10/85, magnifi cation 5000 . ( b ) 15/80, magnifi cation 
12,000 . . 
(a) 
(b)

1112 CONTROLLED RELEASE OF DRUGS FROM TABLET COATINGS 
tion, in Figure 5 , concentration paths in time for the tablet coating prepared with 
5% CA, 90% acetone, and 5% water are shown. According to this plot, the phase 
separation takes place since the concentration paths in time for the drug tablet – 
coating and the coating – air interface cross the binodal curve at the same time. In 
addition, the coating – air interface enters the phase envelope at a polymer volume 
fraction of 0.76, while the tablet – coating interface enters with a volume fraction of 
0.023. These two observations imply that the coating will be porous and asymmetric 
in which the top layer is more dense than the lower sublayer, which was confi rmed 
by the SEM picture in Figure 2 . The predictions, have shown that phase separation 
was achieved for all coating formulations, supporting the morphological observa- 
FIGURE 4 SEM of the cross section of the asymmetric - membrane made with 10% water 
and CA – acetone weight ratios of ( a ) 10/80, magnifi cation 10,000 . , and ( b ) 15/75, magnifi cation 
10,000 . . 
(a) 
(b)

APPLICATIONS 1113 
FIGURE 5 Ternary - phase diagram and concentration paths for coating made with 5% CA 
dissolved in 90% acetone and 5% water. 
FIGURE 6 Concentration profi le of CA in membrane at moment of precipitation for different 
CA – acetone weight fraction ratios. 
tions and the precipitation times calculated for each case (listed in Table 1 ). Model 
predictions can also be used to determine a rough thickness of the high - polymer - 
concentration region near the coating – air interface and the pore distribution of the 
sublayer structure when the polymer concentration profi les at the moment of phase 
separation are plotted. As an illustration, such a plot is shown in Figure 6 for coatings 
prepared with 10% CA in the casting solution. Examination of these profi les 

1114 CONTROLLED RELEASE OF DRUGS FROM TABLET COATINGS 
leads to the following conclusions regarding the effect of increased water concentration: 
(1) The polymer concentration at the coating – air interface slightly decreases. 
(2) More uniform porosity distribution throughout the lower sublayer is favored. 
Also, porosity increases, which is in complete agreement with our release studies 
and the observations of other groups [30, 33] . 
Effect of Evaporation Condition Previous studies on more traditional applications 
have investigated the effect of increased air velocity, that is, forced - convection 
conditions for a combination of dry/wet phase inversion techniques to produce 
defect - free, ultrahigh fl ux asymmetric membranes with ultrathin skin layers [115 – 
117] . To investigate the effect of evaporation condition on the release rate of drug, 
tablets were dip coated with CA solution containing 10% CA, 80% acetone, and 
10% water and allowed to dry by blowing air across the surface with a blower 
(forced convection). As a comparison, tablets coated with the same solution were 
air dried under natural free - convection conditions. 
As illustrated in Figure 7 , the release profi les of both tablet coatings show a linear 
behavior only at small times, and then exponential increases in concentrations were 
observed. Based on this behavior, the release profi les were fi tted to an empirical 
equation as 
C kt eb Ct = + . . 0 1 0 (5) 
where k 0 and b 0 are fi tting parameters. The accuracy of Equation (5) for correlating 
the release rate data in Figure 7 was confi rmed by high R 2 values, very close to 1 in 
both cases. Due to the presence of the second term in Equation (5) , the release rate 
FIGURE 7 Release of theophylline from tablet coatings made with 10% CA dissolved in 
80% acetone and 10% water. Coated tablets were dried under free - and forced - convection 
conditions. The lines correspond to prediction from Equation (5) using k 0 = 1.92 . 
10 . 5 mg/(mL·min); b 0 = 0.1179 mL/(mg·min) for forced convection and k 0 = 2.41 . 
10 . 5 mg/(mL·min); b 0 = 0.1174 mL/(mg·min) for free convection. 

APPLICATIONS 1115 
of the drug, dC/dt , is no longer constant and its dependency on the concentration 
of drug in the dissolution medium, C , can be expressed as 
dC 
dt 
k e bCt 
k b t e 
b Ct 
b Ct = + + . . 
. 
0 0 
0 0 
2 
1 1 
1 
0 
0 
( ) 
(6) 
Using Equation (6) , the release rate of theophylline from the tablets dried under 
forced - and free - convection conditions were determined as 0.047 and 0.078 mg/min, 
respectively. It shoud be noted that both of these values correspond to the arithmetic 
average of the release rates calculated at each average concentration level shown 
in Figure 7 . The difference in release rates can be explained by comparing the scanning 
electron micrographs shown in Figures 8 a and 8 b . It can be seen that the cross - 
sectional morphology of the tablet coating dried under the forced - convection 
condition is dense and nonporous while a porous and asymmetric structure is 
observed for the tablet coating dried under free - convection conditions. In the dense 
coating, diffusional resistance to transport of the drug occurs through the overall 
thickness and is larger than that in the asymmetric porous coating; hence, a lower 
drug release rate is observed. To understand the effect of air velocity on the formation 
of the coating structure, we have utilized our model predictions. The composition 
paths plotted in Figure 9 indicate that, when the speed of air in the drying 
atmosphere is signifi cantly increased, the rate of evaporation of solvent (acetone) 
increases dramatically and within a short time its concentration at the surface drops 
to zero. This situation leads to very strong diffusional resistance within the membrane 
solution and, thus, slow evaporation of the nonsolvent (water). Consequently, 
phase separation is never achieved and the resulting membrane structure becomes 
dense as supported by the SEM picture in Figure 8 a . 
Effect of Nonsolvent Type A few studies in the literature have shown that various 
membrane morphologies can be obtained by changing the type of nonsolvent in 
the casting solution [118 – 120] . To investigate the effect of nonsolvent type, tablet 
coatings were prepared from a casting solution of CA in acetone as a solvent and 
octanol, formamide, glycerol, and hexanol as nonsolvents. The drastic change in the 
release rates of theophylline and percentage of dense skin layer at the surface of 
the tablet coating with nonsolvent type is shown in Figure 10 . The decrease in the 
release rates is associated with the increase in the thickness of dense skin layer of 
the coating. The results illustrated in Figure 10 indicate that the membrane structure 
formation during the phase inversion process is strongly infl uenced by the type of 
nonsolvent, since each nonsolvent has different volatility, thermodynamic, and 
diffusion characteristics. The difference in the rates of evaporation and diffusion 
of nonsolvents and change in the miscibility gaps in the case of each nonsolvent 
lead to different mass transfer paths; consequently, the morphology of the resulting 
membrane structures signifi cantly varies from porous symmetric to dense asymmetric 
ones. 
Statistical Analysis of Experimental Design The effect of the composition of the 
coating solution on the release rate of drug was investigated in detail with the 14 
formulations listed in Table 1 . The drug release rate was chosen as an appropriate 

1116 CONTROLLED RELEASE OF DRUGS FROM TABLET COATINGS 
response variable since zero - order release was easily achieved for all tablet coatings 
prepared. The data in Table 1 were best fi t by the special cubic equation 
Release rate= + + . 
. . 
70 94 1 698 37 55 94 66 
354 3 49 
1 2 3 1 2 
1 3 
. . . . 
.
. . . .. 
. . . . 3 357 66 2 3 1 1 3 . . . . . + 
(7) 
Results of the regression analysis are given in Table 2 . An excellent fi t of the 
experimental release rate data to Equation (7) was confi rmed by the high R 2 value 
of 0.9801. In addition to the R 2 values, the signifi cance of Equation (7) and each 
FIGURE 8 SEM of cross section of asymmetric - membrane made with 10% CA dissolved 
in 80% acetone and 10% water: ( a ) coating solution was dried under forced - convection 
condition, magnifi cation 6500 . ; ( b ) coating solution was dried under free - convection condition, 
magnifi cation 5000 . . 
(a) 
(b)

APPLICATIONS 1117 
term in it to the prediction of the release rate of theophylline was evaluated by the 
F statistic or F value. The F statistic is viewed as a ratio that expresses variance 
explained by the model divided by variance due to model error or experimental 
error and is defi ned as 
F 
R k 
R n k 
= 
. . . 
2 
2 1 1 
/ 
/ ( )( ) 
(8) 
FIGURE 9 Ternary - phase diagram and concentration paths for coating made with 10% 
CA dissolved in 80% acetone and 10% water. Coating solution was dried under forced - 
convection condition. 
FIGURE 10 Release rates of theophylline and percentage of dense skin layer of membranes 
as function of nonsolvent type. Polymer: CA solvent – acetone. 

1118 CONTROLLED RELEASE OF DRUGS FROM TABLET COATINGS 
where k is the number of variables in the model. Usually the computed value of F 
is compared with the critical F value, F k , n . k , 1 . . , where . is a preselected signifi cance 
level. If the value of F is substantially greater than the critical F value ( F critic ), that 
is, if . F , the difference between F and F critic , is large, then the regression equation is 
considered useful in predicting the response. We have assessed the contribution of 
each interaction term by comparing the change in . F and R 2 values between the 
full model given in Equation (7) and reduced models. The reduced models were 
obtained by deleting a specifi c interaction term in the full model; for example, model 
2 includes all terms in Equation (7) except the term involving . 1 . 2 . 3 . The results 
in Table 2 indicate that all binary and ternary interaction terms in the full model 
are needed for accurate prediction of the release rate since the largest . F values 
are calculated for the full model for both signifi cance levels, . = 0.05 and . = 0.01. 
According to the criterion mentioned above, among all interactions, CA – acetone 
( . 1 . 2 ) was identifi ed as the most infl uential factor on the response since the largest 
decrease in both . F and R 2 values compared to those of the full model were 
observed when the term . 1 . 2 was deleted from the full model. This simply implies 
that changing the CA – acetone ratio in the coating formulation has the most signifi - 
cant effect on the release rate. Specifi cally, increasing the ratio of CA to acetone 
from 5/90 to 15/80 resulted in a decrease of the release rate from 0.45 to 0.036 mg/ 
min since the porosity of the membrane decreases and the thickness of the dense 
skin layer increases. The ratio of the composition of CA to water ( . 1 . 3 ) was also 
found to be an important parameter on the release rate of drug as indicated by the 
second largest decrease in R 2 and . F values compared to those of the full model. 
Decreasing this ratio from 15/5 to 5/15 caused an increase in the release rate by a 
factor of 10 since the thickness of the dense skin layer signifi cantly decreases. Based 
on the decrease in . F and R 2 values from those of the full model given in Table 2 , 
the relative importance of each interaction term can be ranked as follows: . 1 . 2 > 
. 1 . 3 > . 2 . 3 > . 1 . 2 . 3 . 
To further illustrate the simultaneous effects of the factors on the release rate of 
drug, a three - dimensional response surface plot based on Equation (7) was generated, 
as shown in Figure 11 . As can be clearly seen from this fi gure, the release rate 
can be signifi cantly varied just by tailoring the CA – acetone and CA – water ratios 
without changing the coating material. In addition, Figure 11 shows that a slight 
maximum in release rate is observed as the ratio of composition of acetone to water 
increases. 
In order to validate the predictive capability of the empirical expression, two 
formulations with compositions given in Table 3 were selected randomly from the 
TABLE 2 Statistical Analysis of Release Rate Data 
Model Number R 2 F values 
. = 0.05 . = 0.01 
F critic a . F F critic a . F 
1 (full model) 0.9801 42.21 4.21 38 8.26 33.95 
2 ( . 1 . 2 . 3 ) 0.955 25.13 3.87 21.26 7.19 17.94 
3 ( . 2 . 3 ) 0.953 23.57 3.87 19.7 7.19 16.38 
4 ( . 1 . 3 ) 0.946 20.64 3.87 16.77 7.19 13.45 
5 ( . 1 . 2 ) 0.881 8.65 3.87 4.78 7.19 1.46 
a Determined from statistical tables [121] . 

FIGURE 11 Three - dimensional response surface plot of release rate as function of composition 
of coating solution. 
0.40 
0.30 
0.20 
0.09 
–0.01 
.1 (0.25) 
.2 (0.70) 
.2 (0.90) 
.3 (0.25) 
.1 (0.05) 
.3 (0.25) 
Weight fraction 
Release rate (mg/min) 
experimental design region. Experimental release rates estimated from the slope of 
the release profi les and corresponding rates predicted from Equation (7) are also 
listed in Table 3 . According to the results, the maximum absolute percentage difference 
between the experimental and predicted release rates is 3.5%. This value lies 
within the residuals obtained in deriving Equation (7) . Based on this comparison, 
it is fair to conclude that the empirical expression derived in this study can be used 
as a tool to predict the release rate of theophylline for any composition within the 
experimental design region. 
6.5.7 CONCLUSION 
This chapter has considered the controlled release of drugs from tablet coating 
systems. These systems are still the preferred route of drug administration due to 
TABLE 3 Composition of Coating Solution Randomly Selected for Testing Predictive 
Capability of Empirical Expression 
Compositon (wt %) 
Experimental 
Release Rate 
(mg/min) 
Predicted 
Release Rate 
(mg/min) R 2 
Cellulose 
Acetate Acetone Water 
7.5 82.5 10 0.09 0.125 0.9816 
10 82.5 7.5 0.027 0.029 0.9858 
CONCLUSION 1119

1120 CONTROLLED RELEASE OF DRUGS FROM TABLET COATINGS 
their advantages of ease of administration, maintaining zero - order release rates, and 
better patient compliance; thus, they hold the major market share in the pharmaceutical 
industry along with the other formulations of oral drug delivery. In tablet 
coated systems, diffusion of drug through the coating is usually the rate - limiting step 
and the desired drug release rates are achieved by properly selecting the coating 
material and adjusting the morphology of the coating using suitable manufacturing 
methods. Coating characteristics such as glass transition temperature, crystalline 
content, and degree of cross - linking signifi cantly determine the release rate of the 
drug. For a selected coating material, the structure of the coating plays a critical role 
in achieving useful release rates. The morpholology of the tablet coatings can be 
varied by incorporating plasticizer or water - soluble additives in the coating solution, 
by blending the polymers, or by applying multilayer composite coatings. Another 
approach is to apply asymmetric - and porous - type coatings. The morphological 
characteristics of these types of coatings, such as fraction of dense top layer and 
porous sublayer and size and shape of the pores, can be varied by optimizing the 
composition of the coating solution, evaporation conditions (temperature, relative 
humidity, and velocity of air), and the type of solvent or nonsolvent used in the 
coating solution. Asymmetric - type coatings can be used to facilitate osmotic delivery 
of drugs with low solubilities since high water fl uxes can be achieved. These 
coatings allow us to control the release kinetics without altering the coating material 
or signifi cantly varying the coating thickness. 
Currently, considerable research efforts have been directed toward developing 
protein drug delivery systems due to discovery of numerous protein and peptide 
therapeutics. The delivery of protein drugs is usually limited to parenteral administration 
and frequent injections are required due to their short half - lives in the blood. 
In this regard, development of oral delivery systems is necessary for patient compliance 
and convenience. The challenge in the design of the oral delivery systems is 
that they should protect the incorporated drugs from chemical and enzymatic degradation 
until the drug reaches the delivery site. The protein drug should not be 
infl uenced by pH or bacteria and enzymes along the gastrointestinal (GI) tract and 
should be delivered at the desired site with a desired effi ciency. To achieve site - 
specifi c delivery for protein - and peptide - based drugs, one of the strategies that has 
often been investigated is to coat the drug core with polymers that can respond to 
the stimuli of local environments such as pH and enzymes. For water - soluble protein 
drugs, such as insulin, an additional protective coating is usually required to isolate 
the drug from the surrounding water. Composite tablet coating materials which 
combine the enzymatic susceptibility and protective properties of polymers can be 
another solution for this problem. A systematic comparison study among various 
polymer combinations is required to select the appropriate coating materials for 
specifi c drugs. Not only the selection of the coating material but also the manufacturing 
technique for the preparation of tablet coatings plays a critical role and still 
remains one of the most challenging subjects in the controlled drug delivery area. 
Proteins are very sensitive to conditions that can occur during the coating process. 
Mechanical stresses during the preparation, exposure to a hydrophobic organic 
solvent, intermediate moisture level during hydration, and interaction between 
protein and polymer can easily inactivate the protein - based drug. Therefore, more 
research focused on both optimization of coating materials and manufacturing 
methods for encapsulating the protein and peptide drugs is necessary. 

REFERENCES 
1. Langer , R. S. , and Peppas , N. A. ( 1981 ), Present and future applications of biomaterials 
in controlled drug delivery systems , Biomaterials , 2 , 201 – 214 . 
2. Ho , W. S. W. , and Sirkar , K. K. ( 1992 ), Membrane Handbook , Van Nostrand Reinhold , 
New York , pp. 915 – 935 . 
3. Liu , L. , Ku , J. , Khang , G. , Lee , B. , Rhee , J. M. , and Lee , H. B. ( 2000 ), Nifedipine 
controlled delivery by sandwiched osmotic tablet system . J. Controlled Release , 68 , 
145 – 156 . 
4. Liu , L. , Khang , G. , Rhee , J. M. , and Lee , H. B. ( 2000 ), Monolithic osmotic tablet system 
for nifedipine delivery , J. Controlled Release , 67 , 309 – 322 . 
5. Narisawa , S. , Nagata , M. , Ito , T. , Yoshino , H. , Hirakawa , Y. , and Noda , K. ( 1995 ), Drug 
release behavior in gastrointestinal tract of beagle dogs from multiple unit type rate - 
controlled or time - controlled release preparations coated with insoluble polymer - based 
fi lm , J. Controlled Release , 33 , 253 – 260 . 
6. Moussa , I. S. , and Cartilier , L. H. ( 1997 ), Evaluation of cross - linked amylose press - coated 
tablets for sustained drug delivery , Int. J. Pharm. , 149 , 139 – 149 . 
7. Khan , M. Z. I. , Prebeg , E. , and Kurjakovic , N. ( 1999 ), A pH - dependent colon targeted 
oral drug delivery system using methacrylic acid copolymers: I. Manipulation of drug 
release using Eudragit ® L100 - 55 and Eudragit ® S100 combinations , J. Controlled Release , 
58 , 215 – 222 . 
8. Kr o gel , I. , and Bodmeier , R. ( 1999 ), Floating or pulsatile drug delivery systems based 
on coated effervescent cores , Int. J. Pharm. , 187 , 175 – 184 . 
9. Macleod , G. S. , Fell , J. T. , and Collett , J. H. ( 1999 ), An in vitro investigation into the 
potential for bimodal drug release from pectin/chitosan/HPMC - coated tablets , Int. J. 
Pharm. , 188 , 11 – 18 . 
10. Thoma , K. , and Bechtold , K. ( 1999 ), Infl uence of aqueous coatings on the stability of 
enteric coated pellets and tablets , Eur. J. Pharm. Biopharm. , 47 , 39 – 50 . 
11. Fukui , E. , Uemura , K. , and Kobayashi , M. ( 2000 ), Studies on applicability of press - coated 
tablets using hydroxypropylcellulose (HPC) in the outer shell for timed - release preparations 
, J. Controlled Release , 68 , 215 – 223 . 
12. Crotts , G. , Sheth , A. , Twist , J. , and Ghebre - Sellassie , I. ( 2001 ), Development of an enteric 
coating formulation and process for tablets primarily composed of a highly water - 
soluble, organic acid . Eur. J. Pharm. Biopharm. , 51 , 71 – 76 . 
13. Ofori - Kwakye , K. , and Fell , J. T. ( 2003 ), Biphasic drug release from fi lm - coated tablets , 
Int. J. Pharm. , 250 , 431 – 440 . 
14. Goto , T. , Tanida , O. , Yoshinaga , T. , Sato , S. , Ball , D. J. , Wilding , I. R. , Kobayashi , E. , and 
Fujimura , A. ( 2004 ), Pharmaceutical design of a novel colon - targeted delivery system 
using two - layer - coated tablets of three different pharmaceutical formulations, supported 
by clinical evidence in humans , J. Controlled Release , 97 , 31 – 42 . 
15. Sundy , E. , and Danckwerts , M. P. ( 2004 ), A novel compression - coated doughnut - shaped 
tablet design for zero - order sustained release , Eur. J. Pharm. Sci. , 22 , 477 – 485 . 
16. Tarvainen , M. , Peltonen , S. , Mikkonen , H. , Elovaara , M. , Tuunainen , M. , Paronen , P. , 
Ketolainen , J. , and Sutinen , R. ( 2004 ), Aqueous starch acetate dispersion as 
a novel coating material for controlled release products , J. Controlled Release , 96 , 
179 – 191 . 
17. Thombre , A. G. , Appel , L. E. , Chidlaw , M. B. , Daugherity , P. D. , Dumont , F. , Evans , L. A. 
F. , and Sutton , S. C. ( 2004 ), Osmotic drug delivery using swellable - core technology , J. 
Controlled Release , 94 , 75 – 89 . 
REFERENCES 1121

1122 CONTROLLED RELEASE OF DRUGS FROM TABLET COATINGS 
18. Kim , I. H. , Park , J. H. , Cheong , I. W. , and Kim , J. H. ( 2003 ), Swelling and drug release 
behavior of tablets coated with aqueous hydroxypropyl methylcellulose phthalate 
(HPMCP) nanoparticles , J. Controlled Release , 89 , 225 – 233 . 
19. Okimoto , K. , Rajewski , R. A. , and Stella , J. V. ( 1999 ), Release of testosterone from an 
osmotic pump tablet utilizing (SBE)7m - . - cyclodextrin as both a solubilizing and an 
osmotic pump agent , J. Controlled Release , 58 , 29 – 38 . 
20. Khan , M. A. , Sastry , S. V. , Vaithiyalingam , S. R. , Agarwal , V. , Nazzal , S. , and Reddy , I. K. 
( 2000 ), Captopril gastrointestinal therapeutic system coated with cellulose acetate pseudolatex: 
Evaluation of main effects of several formulation variables , Int. J. Pharm. , 193 , 
147 – 156 . 
21. Lu , E - X. , Jiang , Z - Q. , Zhang , Q - Z. , and Jiang , X - G. ( 2003 ), A water - insoluble drug 
monolithic osmotic tablet system utilizing gum arabic as an osmotic, suspending and 
expanding agent , J. Controlled Release , 92 , 375 – 382 . 
22. Zhang , Y. , Zhang , Z. , and Wu , F. ( 2003 ), A novel pulsed - release system based on swelling 
and osmotic pumping mechanism , J. Controlled Release , 89 , 47 – 55 . 
23. Liu , L. , and Che , B. (2006), Preparation of monolithic osmotic pump system by coating 
the indented core tablet , Eur. J. Pharm. Biopharm. , 64 , 180 – 184 . 
24. Zentner , G. M. , Rork , G. S. , and Himmelstein , K. J. ( 1985 ), The controlled - porosity 
osmotic pump , J. Controlled Release , 1 , 269 – 282 . 
25. Baker , R. W. , and Brooke , J. W. ( 1987 ), Pharmaceutical drug delivery system, U.S. Patent 
4,687,660 . 
26. Theeuwes , F. , and Ayer , A. D. ( 1978 ), Osmotic device having composite walls, U.S. Patent 
4,077,407 . 
27. Theeuwes , F. ( 1978 ), Microporous - semipermeable laminated osmotic system, U.S. Patent 
4,256,108 . 
28. Cortese , R. , and Theeuwes , F. ( 1982 ), Osmotic device with hydrogel driving member, U.S. 
Patent 4,327,725 . 
29. Swanson , D. R. , Burday , B. L. , Wong , P. S. L. , and Theeuwes , F. ( 1987 ), Nifedipine gastrointestinal 
therapeutic system , Am. J. Med. , 83 , 3 – 10 . 
30. Herbig , S. M. , Cardinal , J. R. , Korsmeyer , R. W. , and Smith , K. L. ( 1995 ), Asymmetric 
membrane tablet coatings for osmotic drug delivery , J. Controlled Release , 35 , 127 – 
136 . 
31. Cardinal , J. R. , Herbig , S. M. , Korsmeyer , R. W. , Lo , J. , Smith , K. L. , and Thombre , 
A. G. ( 1997 ), Use of asymmetric membranes in delivery devices, U.S. Patent 
5,612,059 . 
32. Cardinal , J. R. , Herbig , S. M. , Korsmeyer , R. W. , Lo , J. , Smith , K. L. , and Thombre , A. G. 
( 1997 ), Asymmetric membranes in delivery devices, U.S. Patent 5,698,220 . 
33. Wang , D - M. , Lin , F - C. , Chen , L - Y. , and Lai , J - Y. ( 1998 ), Application of asymmetric 
TPX membranes to transdermal delivery of nitroglycerin , J. Controlled Release , 50 , 
187 – 195 . 
34. Thombre , A. G. , Cardinal , J. R. , DeNoto , A. R. , Herbig , S. M. , and Smith , K. L. ( 1999 ), 
Asymmetric - membrane capsules for osmotic drug delivery. Part I: Development of 
a manufacturing process , J. Controlled Release , 57 , 55 – 64 . 
35. Thombre , A. G. , Cardinal , J. R. , DeNoto , A. R. , and Gibbes , D. C. ( 1999 ), Asymmetric 
membrane capsules for osmotic drug delivery. Part II: In vitro and in vivo drug release 
performance , J. Controlled Release , 57 , 65 – 73 . 
36. Thombre , A. G. , DeNoto , A. R. , and Gibbes , D. C. ( 1999 ), Delivery of glipizide from 
asymmetric - membrane capsules using encapsulated excipients , J. Controlled Release , 
60 , 333 – 341 . 

37. Lin , Y - K. , and Ho , H - O. ( 2003 ), Investigations on the drug releasing mechanism from 
an asymmetric - membrane - coated capsule with an in situ formed delivery orifi ce , 
J. Controlled Release , 89 , 57 – 69 . 
38. Meier , M. M. , Kanis , L. A. , and Soldi , V. ( 2004 ), Characterization and drug - permeation 
profi les of microporous and dense cellulose acetate membranes: Infl uence of plasticizer 
and pore forming agent , Int. J. Pharm. , 278 , 99 – 110 . 
39. Prabakaran , D. , Singh , P. , Jaganathan , K. S. , and Vyas , S. P. ( 2004 ), Osmotically regulated 
asymmetric capsular systems for simultaneous sustained delivery of anti - tubercular 
drugs , J. Controlled Release , 95 , 239 – 248 . 
40. Wang , C - Y. , Ho , H - O. , Lin , L - H. , Lin , Y - K. , and Ming - Thau Sheu , M - T. ( 2005 ), Asymmetric 
membrane capsules for delivery of poorly water - soluble drugs by osmotic effects , 
Int. J. Pharm. , 297 , 89 – 97 . 
41. Altinkaya , S. A. , and Yenal , H. ( 2006 ), In vitro drug release rates from asymmetric - 
membrane tablet coatings: Prediction of phase - inversion dynamics , Biochem. Eng. J. , 28 , 
131 – 139 . 
42. Wang , G - M. , Chen , C - H. , Ho , H - O. , Wang , S - S. , and Sheu , M - T. ( 2006 ), Novel design 
of osmotic chitosan capsules characterized by asymmetric membrane structure for in 
situ formation of delivery orifi ce , Int. J. Pharm. , 319 , 71 – 81 . 
43. Altinkaya , S. A. , and Ozbas , B. ( 2004 ), Modeling of asymmetric - membrane formation 
by dry - casting method , J. Membr. Sci. , 230 , 71 – 89 . 
44. Mathiowitz , M. ( 1999 ), Encyclopedia of Controlled Drug Delivery , Wiley , New York , 
pp. 302 – 303 . 
45. Sandadi , S. , PAndey , P. , and Turton , R. ( 2004 ), In situ, near real - time acquisition of particle 
motion in rotating pan coating equipment using imaging techniques , Chem. Eng. 
Sci. , 59 , 5807 – 5817 . 
46. Prater , D. , Wilde , J. , and Meakin , B. ( 1980 ), A model system for the production of 
aqueous tablet fi lm coating for laboratory evaluation , J. Pharm. Pharmacol. , 32 (Suppl.), 
90 . 
47. Leaver , T. , Shannon , H. , and Rowe , R. ( 1985 ), A photometric analysis of tablet movement 
in a side - vented perforated drum (Accela - Cota) , J. Pharm. Pharmacol. , 37 , 
17 – 21 . 
48. Nakagawa , M. , Altobelli , S. , Caprihan , A. , Fukushima , E. , and Jeong , E. ( 1993 ), Non - 
invasive measurements of granular fl ows by magnetic resonance imaging , Exper. Fluids , 
16 , 54 – 60 . 
49. Parker , D. , Broadbent , C. , Fowles , P. , Hawkesworth , M. , and McNeil , P. ( 1993 ), Positron 
emission particle tracking — A technique for studying fl ow within engineering equipment 
, Nucl. Instrum. Methods Phys. Res. A , 326 , 592 – 607 . 
50. Parker , D. , Dijkstra , A. , Martin , T. , and Seville , J. ( 1997 ), Positron emission particle tracking 
studies of spherical particle motion in rotating drums , Chem. Eng. Sci. , 52 , 
2011 – 2022 . 
51. Wilson , K. , and Crossman , E. ( 1997 ), The infl uence of tablet shape and pan speed on 
intra - tablet fi lm coating uniformity , Drug Dev. Ind. Pharm. , 23 , 1239 – 1243 . 
52. Yamane , K. , Sato , T. , Tanaka , T. , and Tsuji , Y. ( 1995 ), Computer simulation of tablet 
motion in coating drum , Pharm. Res. , 12 , 1264 – 1268 . 
53. Yamane , K. , Nakagawa , M. , Altobelli , S. , Tanaka , T. , and Tsuji , Y. ( 1998 ), Steady particulate 
fl ows in a horizontal rotating cylinder , Phys. Fluids , 10 , 1419 – 1427 . 
54. Denis , C. , Hemati , M. , Chulia , D. , Lanne , J. , Buisson , B. , Daste , G. , and Elbaz , F. ( 2003 ), 
A model of surface renewal with application to the coating of pharmaceutical tablets 
in rotary drums , Powder Technol. , 130 , 174 – 180 . 
REFERENCES 1123

1124 CONTROLLED RELEASE OF DRUGS FROM TABLET COATINGS 
55. Pandey , P. , Song , Y. , Kayihan, F. , and Turton, R. (2006), Simulation of particle movement 
in a pan coating device using discrete element modeling and its comparison with video - 
imaging experiments , Powder Technol. , 161 , 79 – 88 . 
56. Rege , B. D. , Gawel , J. , and Kou , J. H. ( 2002 ), Identifi cation of critical process variables 
for coating actives onto tablets via statistically designed experiments , Int. J. Pharm. , 237 , 
87 – 94 . 
57. Turton , R. , and Cheng , X. X. ( 2005 ), The scale - up of spray coating processes for granular 
solids and tablets , Powder Technol. , 150 , 78 – 85 . 
58. Xu , M. , and Turton , R. ( 1997 ), A new data processing technique for noisy signals: Application 
to measuring particle circulation times in a draft tube equipped fl uidized bed , 
Powder Technol. , 92 , 111 – 117 . 
59. Sherony , D. F. ( 1981 ), A model of surface renewal with application to fl uid bed coating 
of particles , Chem. Eng. Sci. , 36 , 845 – 848 . 
60. Wnukowski , P. , and Setterwall , F. ( 1989 ), The coating of particles in a fl uidized bed 
(residence time distribution in a system of two coupled perfect mixers) , Chem. Eng. Sci. , 
44 , 493 – 505 . 
61. Choi , M. S. , and Meisen , A. ( 1997 ), Sulfur coating of urea in shallow spouted beds , Chem. 
Eng. Sci. , 52 , 1073 – 1086 . 
62. Maronga , J. , and Wnukowski , P. ( 1997 ), Modeling of the three - domain fl uidized bed 
particulate coating process , Chem. Eng. Sci. , 52 , 2915 – 2925 . 
63. Nakamura , H. , Abe , E. , and Yamada , N. ( 1998 ), Coating mass distributions of seed particles 
in a tumbling fl uidized bed coater. Part II. A Monte Carlo simulation of particle 
coating , Powder Technol. , 99 , 140 – 146 . 
64. Cheng , X. X. , and Turton , R. ( 2000 ), The prediction of variability occurring in fl uidized 
bed coating equipment. II: The role of nonuniform particle coverage as particle pass 
through the spray zone , Pharm. Dev. Technol. , 5 , 323 – 332 . 
65. KuShaari , K. , Pandey , P. , Song , Y. , and Turton , R. ( 2006 ), Monte Carlo simulations to 
determine coating uniformity in a Wurster fl uidized bed coating process , Powder 
Technol. , 166 , 81 – 90 . 
66. Waterman , K. C. , and Fergione , M. B. ( 2003 ), Press - coating of immediate release powders 
onto coated controlled release tablets with adhesives , J. Controlled Release , 89 , 
387 – 395 . 
67. Leong , K. C. , Lu , G. Q. , and Rudolph , V. ( 1999 ), A comparative study of the fl uidized - bed 
coating of cylindrical metal surfaces with various thermoplastic polymer powders , 
J. Mater. Proc. Technol. , 89 / 90 , 354 – 360 . 
68. Wulf , M. , Uhlmann , P. , Michel , S. , and Grundke , K. ( 2000 ), Surface tension studies of 
levelling additives in powder coatings , Prog. Org. Coat. , 38 , 59 – 66 . 
69. Belder , E. G. , Rutten , H. J. J., and Perera , D. Y. ( 2001 ), Cure characterization of powder 
coatings , Prog. Org. Coat. , 42 , 142 – 149 . 
70. Pfeffer , R. , Dave , R. N. , Wie , D. , and Ramlakhan , M. ( 2001 ), Synthesis of engineered 
particulates with tailored properties using dry particle coating , Powder Technol. , 117 , 
40 – 67 . 
71. McLaren , D. D. , and Hollenbeck , R. G. ( 1987 ), A high performance liquid chromatographic 
method for the determination of the amount of hydroxypropyl methylcellulose 
applied to tablets during an aqueous fi lm coating operation , Drug Dev. Ind. Pharm. , 13 , 
2179 – 2197 . 
72. Mouget , Y. , Gosselin , P. , Tourigny , M , and B ’ echard , S. ( 2003 ), Three - dimensional analyses 
of tablet content and fi lm coating uniformity by laser - induced breakdown spectroscopy 
(LIBS) , Am. Lab. , 35 , 20 – 22 . 

73. St - Onge , L. , Kwong , E. , Sabsabi , M. , and Vadas , E. B. ( 2002 ), Quantitative analysis of 
pharmaceutical products by laser - induced breakdown spectroscopy , Spectrochim. Acta, 
Part B , 57 , 1131 – 1140 . 
74. Kirsch , J. D. , and Drennen , J. K. ( 1995 ), Determination of fi lm - coated tablet parameters 
by near - infrared spectroscopy . J. Pharm. Biomed. Anal. , 13 , 1273 – 1281 . 
75. Kirsch , J. D. , and Drennen , J. K. ( 1996 ), Near - infrared spectroscopic monitoring of the 
fi lm coating process , Pharm. Res. , 13 , 234 – 237 . 
76. Anderson , M. , Josefson , M. , Langkilde , F. , and Wahlund , K. - G. ( 1999 ), Monitoring of a 
fi lm coating process for tablets using near infrared refl ectance spectrometry , J. Pharm. 
Biomed. Anal. , 20 , 27 – 37 . 
77. Romero - Torres , S. , P e rez - Ramos , J. D. , Morris , K. R. , and Grant , E. R. ( 2006 ), 
Raman spectroscopy for tablet coating thickness quantifi cation and coating characterization 
in the presence of strong fl uorescent interference , J. Pharma. Biomed. Anal. , 41 , 
811 – 819 . 
78. Romero - Torres , S. , P e rez - Ramos , J. D. , Morris , K. R. , and Grant , E. R. ( 2005 ), Raman 
spectroscopic measurement of tablet - to - tablet coating variability , J. Pharm. Biomed. 
Anal. , 38 , 270 – 274 . 
79. Ruotsalainen , M. , Hein a m a ki , J. , Guo , H. , Laitinen , N. , and Yliruusi , J. ( 2003 ), A novel 
technique for imaging fi lm coating defects in the fi lm - core interface and surface of 
coated tablets , Eur. J. Pharm. Biopharm. , 56 , 381 – 388 . 
80. Podczeck , F. ( 1998 ), Measurement of surface roughness of tablets made from polyethylene 
glycol powders of various molecular weight , Pharm. Pharmacol. Commun. , 4 , 
179 – 182 . 
81. Riippi , M. , Antikainen , O. , Niskanen , T. , and Yliruusi , J. ( 1998 ), The effect of compression 
force on surface structure, crushing strength, friability, and disintegration time of erythromycin 
acistrate tablets , Eur. J. Pharm. Biopharm. , 46 , 339 – 345 . 
82. Newton , M. , Petersson , J. , Podczeck , F. , Clarke , A. , and Booth , S. ( 2001 ), The infl uence 
of formulation variables on the properties of pellets containing a self - emulsifying 
mixture , J. Pharm. Sci. , 90 , 987 – 995 . 
83. Seitavuopio , P. , Rantanen , J. , and Yliruusi , J. ( 2003 ), Tablet surface characterisation by 
various imaging techniques , Int. J. Pharm. , 254 , 281 – 286 . 
84. Seitavuopio , P. , Rantanen , J. , and Yliruusi , J. ( 2005 ), Use of roughness maps in visualisation 
of surfaces , Eur. J. Pharm. Biopharm. , 59 , 351 – 358 . 
85. van de Witte , P. , Dijkstra, P. J. , Van den Berg , J. W. A. , and Feijen, J. (1996), Phase separation 
processes in polymer solutions in relation to membrane formation , J. Membr. Sci. , 
117 , 1 – 31 . 
86. Han , M. , and Bhattacharyya , D. ( 1995 ), Changes in morphology and transport characteristics 
of polysulfone membranes prepared by different demixing conditions , J. Membr. 
Sci. , 98 , 191 – 200 . 
87. Park, H. C. , Kim, Y. P. , Kim, H. Y. , and Kang , Y. S. (1999), Membrane formation by water 
vapor induced phase inversion , J. Membr. Sci. , 156 , 169 – 178 . 
88. Tobiska , S. , and Kleinebudde , P. ( 2001 ), A simple method for evaluating the mixing effi - 
ciency of a new type of pan coater , Int. J. Pharm. , 224 , 141 – 149 . 
89. Plumb , A. P. , Rowe , R. C. , York , P. , and Doherty , C. ( 2002 ), The effect of experimental 
design on the modeling of a tablet coating formulation using artifi cial neural networks , 
Eur. J. Pharm. Sci. , 16 , 281 – 288 . 
90. Rege , B. D. , Gawel , J. and Kou , H. J. ( 2002 ), Identifi cation of critical process variables 
for coating actives onto tablets via statistically designed experiments , Int. J. Pharm. , 237 , 
87 – 94 . 
REFERENCES 1125

1126 CONTROLLED RELEASE OF DRUGS FROM TABLET COATINGS 
91. Mueller , R. , and Kleinebudde , P. (in press), Infl uence of scale - up on the abrasion of 
tablets in a pan coater , Eur. J. Pharm. Biopharm. 
92. Wang , G - M. , Chen , C - H. , Ho , H - O. , Wang , S - S. , and Sheu , M - T. (press), Novel design 
of osmotic chitosan capsules characterized by asymmetric membrane structure for in 
situ formation of delivery orifi ce , Int. J. Pharm. 
93. Hussain , A. S. , Shivanand , P. , and Johnson , R. D. ( 1994 ), Application of neural computing 
in pharmaceutical product development: Computer aided formulation design , Drug 
Dev. Ind. Pharm. , 20 , 1739 – 1752 . 
94. Kesavan , J. G. , and Peck , G. E. ( 1995 ), Pharmaceutical formulation using neural networks 
. Proc.14th Pharm. Technol. Conf. (Barcelona) , 2 , 413 – 431 . 
95. Colbourn , E. A. , and Rowe , R. C. ( 1996 ), Modeling and optimization of a tablet 
formulation using neural networks and genetic algorithms , Pharm. Technol. Eur. , 8 , 
46 – 55 . 
96. Bourquin , J. , Schmidli , H. , van Hoogevest , P. , and Leuenberger , H. ( 1997 ), Basic concepts 
of artifi cial neural networks (ANN) modeling in the application to pharmaceutical 
development , Pharm. Dev. Technol. , 2 , 95 – 109 . 
97. Bourquin , J. , Schmidli , H. , van Hoogevest , P. , and Leuenberger , H. ( 1997b ), Application 
of artifi cial neural networks (ANN) in the development of solid dosage forms , 
Pharm. Dev. Technol. , 2 , 111 – 121 . 
98. Ebube , N. K. , McCall , T. , Chen , Y. , and Meyer , M. C. ( 1997 ), Relating formulation 
variables to in vitro dissolution using an artifi cial neural network , Pharm. Dev. Technol. , 
2 , 225 – 232 . 
99. Bourquin , J. , Schmidli , H. , van Hoogevest , P. , and Leuenberger , H. ( 1998 ), Comparison 
of artifi cial neural networks (ANN) with classical modeling techniques using different 
experimental designs and data from a galenical study on a solid dosage form , Eur. J. 
Pharm. Sci. , 6 , 287 – 300 . 
100. Bourquin , J. , Schmidli , H. , van Hoogevest , P. , and Leuenberger , H. ( 1998 ), Advantages 
of artifi cial neural networks (ANNs) as alternative modeling technique for data sets 
showing non - linear relationships using data from a galenical study on a solid dosage 
form , Eur. J. Pharm. Sci. , 7 , 5 – 16 . 
101. Bourquin , J. , Schmidli , H. , van Hoogevest , P. , and Leuenberger , H. ( 1998 ), Pitfalls of 
artifi cial neural networks (ANN) modeling technique for data sets containing outlier 
measurements using a study on mixture properties of a direct compressed dosage form , 
Eur. J. Pharm. Sci. , 7 , 17 – 28 . 
102. Chen , Y. , McCall , T. W. , Baichwal , A. R. , and Meyer , M. C. ( 1999 ), The application of 
an artifi cial neural network and pharmacokinetic simulations in the design of controlled - 
release dosage forms , J. Controlled Release , 59 , 33 – 41 . 
103. Plumb , A. P. , Rowe , R. C. , York , P. , and Doherty , C. ( 2002 ), The effect of experimental 
design on the modeling of a tablet coating formulation using artifi cial neural networks , 
Eur. J. Pharm. Sci. , 16 , 281 – 288 . 
104. Plumb , A. P. , Rowe , R. C. , York , P. , and Doherty , C. ( 2003 ), Effect of varying optimization 
parameters on optimization by guided evolutionary simulated annealing 
(GESA) using a tablet fi lm coat as an example formulation , Eur. J. Pharma. Sci. , 18 , 
259 – 266 . 
105. Takayama , K. , Fujikawa , M. , Obata , Y. , and Morishita , M. ( 2003 ), Neural network 
based optimization of drug formulations , Adv. Drug Deliv. Rev. , 55 , 1217 – 1231 . 
106. Montgomery , D. C. ( 1997 ), Design and Analysis of Experiments , 4th ed., Wiley , New 
York , pp. 611 – 616 . 

107. Lai , J. - Y. , Lin , F. - C. , Wang , C. - C. , and Wang , D. - M. ( 1996 ), Effect of nonsolvent additives 
on the porosity and morphology of asymmetric TPX membranes , J. Membr. Sci. , 
118 , 49 – 61 . 
108. Broadhead , K. W. , and Tresco , P. A. ( 1998 ), Effects of fabrication conditions on the 
structure and function of membranes formed from poly(acrylonitrile - vinylchloride) , 
J. Membr. Sci. , 147 , 235 – 245 . 
109. Won , J. , Park , H. C. , Kim , U. Y. , Kang , S. , Yoo , S. H. , and Jho , J. Y. ( 1999 ), The effect 
of dope solution characteristics on the membrane morphology and gas transport properties: 
PES/ - BL/NMP system , J. Membr. Sci. , 162 , 247 – 255 . 
110. Young , T - H. , Lin , D. - J. , Gau , J. - J. , Chuang , W. - Y. , and Cheng , L. - P. ( 1999 ), Morphology 
of crystalline Nylon - 610 membranes prepared by the immersion – precipitation 
process: Competition between crystallization and liquid – liquid phase separation , 
Polymer , 40 , 5011 – 5021 . 
111. Chuang , W. - Y. , Young , T. - H. , Chiu , W. - Y. , and Lin , C. - Y. ( 2000 ), The effect of polymeric 
additives on the structure and permeability of poly(vinyl alcohol) asymmetric - 
membranes , Polymer , 41 , 5633 – 5641 . 
112. Won , J. , Lee , H. J. , and Kang , Y. S. ( 2000 ), The effect of dope solution characteristics 
on the membrane morphology and gas transport properties. Part 2: PES/BL system , 
J. Membr. Sci. , 176 , 11 – 19 . 
113. Fan , S. - C. , Wang , Y. - C. , Li , C. - L. , Lee , K. - R. , Liaw , D. - J. , Huang , H. - P. , and Lai , J. - Y. 
( 2002 ), Effect of coagulation media on membrane formation and vapor permeation 
performance of novel aromatic polyamide membrane , J. Membr. Sci. , 204 , 67 – 79 . 
114. Mohamed , N. A. , and Al - Dossary , A. O. H. ( 2003 ), Structure – property relationships 
for novel wholly aromatic polyamide - hydrazides containing various proportions of para - 
phenylene and meta - phenylene units. Part III: Preparation and properties of semi - 
permeable membranes for water desalination by reverse osmosis separation performance , 
Eur. Polym. J. , 39 , 1653 – 1667 . 
115. Pinnau , I. , and Koros , W. ( 1991 ), Structures and gas separation property asymmetric 
polysulfone membranes made by dry, wet, and dry/wet phase - inversion , J. Appl. Polym. 
Sci. , 43 , 1491 – 1502 . 
116. Sharpe , I. D. , Ismail , A. F. , and Shilton , S. J. ( 1999 ), A study of extrusion shear and 
forced convection residence time in the spinning of polysulfone hollow fi ber membranes 
for gas separation , Sep. Purif. Technol. , 17 , 101 – 109 . 
117. Ismail , A. F. , Ng , B. C. , and Abdul Rahman , W. A. W. ( 2003 ), Effects of shear rate and 
forced convection residence time on asymmetric polysulfone membranes structure and 
gas separation performance , Sep. Purif. Technol. , 33 , 255 – 272 . 
118. Matsuyama , H. , Teramoto , M. , and Uesaka , T. ( 1997 ), Membrane formation and structure 
development by dry cast process , J. Membr. Sci. , 135 , 271 – 288 . 
119. Jansen , J. C. , Macchione , M. , and Drioli , E. ( 2005 ), High fl ux asymmetric gas separation 
membranes of modifi ed poly(ether ether ketone) prepared by the dry phase inversion 
technique , J. Membr. Sci. , 255 , 167 – 180 . 
120. Jansen , J. C. , Buonomenna , M. G. , Figoli , A. , and Drioli , E. ( 2006 ), Asymmetric membranes 
of modifi ed poly(ether ether ketone) with an ultra - thin skin for gas and vapour 
separations , J. Membr. Sci. , 272 , 188 – 197 . 
121. Kleinbaum , D. G. , Kupper , L. L. , and Muller , K. E. , ( 1987 ), Applied Regression Analysis 
and Other Multivariable Methods , 2nd ed., Duxbury , Belmont , pp. 658 – 659 . 
REFERENCES 1127

1128 CONTROLLED RELEASE OF DRUGS FROM TABLET COATINGS 
FIGURE A2 Release of theohylline from tablet coatings made with 10% CA dissolved in 
85% acetone and 5% water. 
FIGURE A1 Release of theohylline from tablet coatings made with 5% CA dissolved in 
90% acetone and 5% water. 
APPENDIX 
The in vitro release profi les of the model drug theophylline are shown below in 
Figures A1 through A10 . 

FIGURE A4 Release of theohylline from tablet coatings made with 5% CA dissolved in 
85% acetone and 10% water. 
FIGURE A3 Release of theohylline from tablet coatings made with 15% CA dissolved in 
80% acetone and 5% water. 
APPENDIX 1129

1130 CONTROLLED RELEASE OF DRUGS FROM TABLET COATINGS 
FIGURE A6 Release of theohylline from tablet coatings made with 15% CA dissolved in 
75% acetone and 10% water. 
FIGURE A5 Release of theohylline from tablet coatings made with 10% CA dissolved in 
80% acetone and 10% water. 

FIGURE A8 Release of theohylline from tablet coatings made with 10% CA dissolved in 
75% acetone and 15% water. 
FIGURE A7 Release of theohylline from tablet coatings made with 5% CA dissolved in 
80% acetone and 15% water. 
APPENDIX 1131

1132 CONTROLLED RELEASE OF DRUGS FROM TABLET COATINGS 
FIGURE A10 Release of theohylline from tablet coatings made with 12.5% CA dissolved 
in 77.5% acetone and 10% water. 
FIGURE A9 Release of theohylline from tablet coatings made with 15% CA dissolved in 
70% acetone and 15% water. 

1133 
6.6 
TABLET COMPRESSION 
Helton M. M. Santos and Jo a o J. M. S. Sousa 
University of Coimbra, Coimbra, Portugal 
Contents 
6.6.1 Introduction 
6.6.2 Theory of Particle Compaction 
6.6.3 Compactibility 
6.6.4 Tablet Compression 
6.6.5 Equipment for Tablet Compression 
6.6.6 Tablet Press Tooling 
6.6.7 Tablet Engraving 
6.6.8 Tablet Shape and Profi le 
6.6.9 Tablet Bisect 
6.6.10 Problems during Tablet Manufacturing 
6.6.10.1 Capping and Lamination 
6.6.10.2 Picking and Sticking 
6.6.10.3 Mottling 
6.6.10.4 Weight and Hardness Variation 
References 
6.6.1 INTRODUCTION 
Tablets are the most important pharmaceutical dosage from and their design has 
always been of great interest to pharmaceutical engineering. Since tablets are made 
by a process of die compaction, although commonly called tablet compression, many 
investigations have been involved in the task to describe the mechanisms involved 
in this process. Nevertheless, some considerations should be taken regarding the 
defi nitions of stages involved in tablet compression. Compression is one of two 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

1134 TABLET COMPRESSION 
stages involved in the compaction of a two - phase system due to the application of 
an external force. It is defi ned as the reduction in the bulk volume of a material as 
a result of gaseous phase [1, 2] . The second stage is consolidation, which is described 
as an increase in the mechanical strength of the material resulting in particle - 
to - particle interaction [1, 2] . 
It is suggested that four mechanisms are basically involved in the process of 
compression of particles: deformation, densifi cation, fragmentation, and attrition. 
The process of compression is briefl y described as follows: small solid particles are 
fi lled in a die cavity and a compression force is applied to it by means of punches 
and then the formed monolithic dosage form is ejected. The shape of the tablet is 
dictated by the shape of the die while the distance between the punch tips at the 
point of maximum compression governs the tablet thickness, and the punch tip 
confi guration determines the profi le of the tablet. The compression cycle in a conventional 
rotary tablet press will be described in detail in this chapter and is illustrated 
in Figure 1 . 
The physical and mechanical properties of tablets, such as density and mechanical 
strength, are signifi cantly affected by the process. Since tablet compression relies on 
the ability of particulates to be compacted, the need to control the critical properties 
of the materials with respect to readiness or ability to compact is an important issue 
to the formulator. 
In order to compress a powder or granulation product into a tablet of specifi c 
hardness, a defi ned compression force must be applied. As pointed out by Shlieout 
et al. [3] , by compressing a constant mass of powder, any variation in the applied 
force causes a change in the measured force. In addition, the substance itself plays 
an important role, that is, if it is of good compressibility, then the force needed for 
compression would be low. It is well known that this compressibility will depend on 
powder characteristics such as crystal habit and thermodynamic behavior. 
The structure and strength of tablets are often discussed in terms of the relationship 
between the properties of the particulate material and the properties of the 
formed tablet. The properties of a powder that control its evolution into a tablet 
FIGURE 1 Compression cycle. (Courtesy of Thomas Engineering.) 
(a) Creating thecavity 
Fill cam 
(b) Filling the cavity 
Suction 
gravity 
feeder 
(c) Metering thecavity 
Weight adjustment ramp 
and head 
scraper 
(d) Containing the fill 
Tail over die 
Pull down after 
weightcontrol 
Punch-holding device 
(e) Precompression 
Solid formation 
(f) Compressing 
Solid formation 
(g) Removing solid 
Ejection cam 

during compression, which will also relate to the fracture toughness and the tensile 
strength of the tablets, are the compression mechanics of the particles and their 
dimensions. Generally, all materials have the ability to store some elastic strain; 
however, its extent will greatly vary for different materials and will depend upon 
the intrinsic nature of the material. There are many instances where a brittle material, 
or its surface, reduces signifi cantly its cohesion or adhesion compared to that 
of a ductile material [4] . 
6.6.2 THEORY OF PARTICLE COMPACTION 
Basically, the process of tablet compression starts with the rearrangement of particles 
within the die cavity and initial elimination of voids. As tablet formulation is a 
multicomponent system, its ability to form a good compact is dictated by the compressibility 
and compactibility characteristics of each component. Compressibility 
of a powder is defi ned as its ability to decrease in volume under pressure, and compactibility 
is the ability of the powdered material to be compressed into a tablet of 
specifi c tensile strength [1, 2] . One emerging approach to understand the mechanism 
of powder consolidation and compression is known as percolation theory. In a 
simple way, the process of compaction can be considered a combination of site and 
bond percolation phenomena [5] . Percolation theory is based on the formation of 
clusters and the existence of a site or bond percolation phenomenon. It is possible 
to apply percolation theory if a system can be suffi ciently well described by a lattice 
in which the spaces are occupied at random or all sites are already occupied and 
bonds between neighboring sites are formed at random. 
The transitional repacking stage is driven by the particle size distribution and 
shape. This will determine the bulk density as the powder or granulation product is 
delivered into the die cavity. In this phase, the punch and particle movements occur 
at low pressure. The particles fl ow with respect to each other, with the fi ner particles 
entering the void between the larger particles, and thus the bulk density of the 
granulation is increased. Various techniques have been utilized to determine the 
degree of the two consolidation mechanisms in pharmaceutical solids (initial packing 
of the particles and elimination of void spaces), namely the rate dependency technique. 
By applying this technique, stress relaxation data based on the Maxwell 
model of viscoelastic behavior indicate virtually no rate dependency for elastic or 
brittle materials. There is also an increase in the calculated yield pressure with an 
increase in punch velocity for viscoplastic materials such as maize starch and polymeric 
materials. This is attributed to the reduction of time necessary for the plastic 
deformation process to occur [6] . For brittle materials such as magnesium and 
calcium carbonates there is no observed change in the yield pressure with increasing 
punch velocity [6] . 
When a force is applied to a material, deformation occurs. When this deformation 
completely disappears after cessation of the external force, further deformation 
occurs. Deformations that do not completely recover after release of the stress are 
known as plastic deformations. The force required to initiate a plastic deformation 
is known as the yield stress. When the particles are so closely packed that no further 
fi lling of the voids can occur, a further increase of the compressional force causes 
deformation at the points of contact. Both plastic and elastic deformation may occur, 
THEORY OF PARTICLE COMPACTION 1135

1136 TABLET COMPRESSION 
although one type will predominate for a given material. The ability of materials to 
be compressed relies on their deformation behavior. The known extreme cases are 
as follows. For elastic bodies, the force applied to consolidation will be fully given 
back (action equals reaction). This is expressed as a completely elastic deformation. 
For plastic bodies, the force applied will be saved as energy in the body and will 
express no elastic deformation at all. During tablet building, these two processes 
never occur alone but occur only in combination, as mentioned before. 
As the external force is increased, the stresses within the particles become great 
enough and cracks may occur. Fragmentation furthers densifi cation with the infi ltration 
of the smaller fragments into the void spaces being responsible for increasing 
the number of particles and formation of new and clean surfaces that are potential 
bonding areas. The mechanisms of fragmentation and plastic deformation are not 
independent since both phenomena modify particle size distribution, and larger 
particles do not act as small particles with respect to plastic deformation [7] . 
The bonding of particles is governed by different mechanisms. The three most 
considered theories are mechanical theory, intermolecular theory, and liquid surface 
fi lm theory. The fi rst theory assumes that under pressure the individual particles 
experience elastic, plastic, or brittle deformation and that the edges of the particles 
intermesh, forming a mechanical bonding. According to Parrot [2] , intermolecular 
theory states that under pressure the molecules at the points of true contact are 
close enough so that van der Waals forces interact to consolidate the particles. 
Liquid surface fi lm theory relies on the presence of a thin liquid fi lm, which may be 
the consequence of fusion or solution, at the surface of the particle, induced by the 
energy of compression. Even tough the applied force is not high, it is locally transmitted 
to small areas of true contact so that a very high pressure will exist at the 
contact surfaces. This high pressure plays an important role in the melting point and 
solubility of the material and proves to be essential to bonding. It follows that after 
releasing the pressure, solidifi cation of the fused material would form solid bridges 
between the particles. An important consideration has been proposed by Zuurman 
et al. [8] to explain the action of some excipients during this phase. One of these 
excipients is magnesium stearate, which is widely used as a lubricant in order to 
prevent tablets from sticking to the die and punches and minimize wear of tooling. 
It has been proven that magnesium stearate can have an adverse effect on bonding 
between particles. The decrease of tablet strength is always considered to be the 
result of reduction of interparticle bonding due to the addition of a lubricant. 
The production of tablets with the desired characteristics depends on the stresses 
induced by elastic rebound and the associated deformation processes during decompression 
and ejection. Ideally, if only elastic deformation occurred, with the sudden 
removal of axial pressure the particles would return to their original form, breaking 
any bonds that may have been under pressure. 
Finally, as the lower punch rises and pushes the tablet upward, there is a continued 
residual die wall pressure and considerable energy may be expanded due to the 
die wall friction. As the tablet is removed form the die, the lateral pressure is 
relieved, and the tablet undergoes elastic recovery with an increase in the volume 
of the portion removed from the die. 
The compression cycle profi les may be used to characterize the consolidation 
mechanisms of powders as they help to characterize the extent of pressure distribution 
within the powder bed as well as the formed tablet. The compression behavior 

of powder mixtures is usually characterized using the well - known Heckel equation 
[9, 10] , which describes the relationship between the porosity of a compact and the 
applied pressure and is based on the assumption that the densifi cation of the bulk 
powder in the die follows fi rst - order kinetics: 
ln 
r 
1 
1. 
= + 
. 
kP A 
(1) 
where . r is the relative density of the compact at pressure . , P is the applied pressure, 
and K and A are constants. The constants A and k are determined from the 
intercept and slope, respectively, of the extrapolated linear region of a plot of ln(1/1 
. . r ) versus . (compaction pressure). The Heckel constant k is related to the reciprocal 
of the mean yield pressure, which is the minimum pressure required to cause 
deformation of the material undergoing compression. The intercept obtained from 
the slope of the upper portion of the curve is a refl ection of the densifi cation after 
consolidation. A large value of k indicates the onset of plastic deformation at relatively 
low pressure. Thus, K appears to be a material constant. The correlation 
between k and the mean yield pressure P y gives Equation (2) . The constant A is 
related to the densifi cation during die fi lling and particle rearrangement prior to 
bonding [11] : 
k 
P 
= 1
y 
(2) 
A high . r value indicates that there will be a high volume reduction of the product 
due to particle rearrangement. The constant A has been shown to be equal to the 
reciprocal of the mean yield pressure required to induce plastic deformation. A 
larger value for A (low yield pressure) indicates the onset of plastic deformation at 
relatively low pressure, a sign that the material is more compressible. 
The Heckel plot allows an interpretation of the mechanism of bonding. A nonlinear 
plot with small value for its slope (a small Heckel constant) indicates that the 
material undergoes fragmentation during compression. When the plot is linear, it 
indicates that the material undergoes plastic deformation during compression. 
In addition to the Heckel approach, other techniques may be applied to the 
characterization of powder compression. One of these approaches was proposed by 
Cooper and Eaton [12] : 
V V 
V V 
a 
k 
P 
a 
k
P 
0 
0 
1 
1 
2 
2 .
. 
= . ( )+ . ( ) s 
exp exp 
(3) 
where V is the volume of the compact at pressure P (m 3 ), V 0 is the volume of 
compact at zero pressure (m 3 ), V s is the void - free solid material volume (m 3 ), a 1 , a 2 , 
k 1 , and k 2 are the Cooper – Eaton constants. 
The Kawakita equation [13] describes the relationship between volume reduction 
and applied pressure according to Equation (4) , where P is the applied pressure, V 0 
is the initial bulk volume, V is the volume at pressure P, a and b are the constants 
characteristic of the powder under compression, and C is the degree of volume 
reduction [Equation (5) ]: 
THEORY OF PARTICLE COMPACTION 1137

1138 TABLET COMPRESSION 
P
C 
P
a ab 
= + 1 (4) 
C 
V V 
V 
= . 0 
0 
(5) 
In the Kawakita equation the particle density is not introduced in the calculations 
since the model operates on the relative change in volume, which gives the same 
result whether the relative or the absolute volume is used. The problem in the calculation 
of this equation is to fi nd the correct initial volume V 0 . 
6.6.3 COMPACTIBILITY 
Compactibility of a powdered mixture is defi ned as the ability of the material to be 
compressed into a tablet of a specifi ed strength without changing its composition. 
Investigations have demonstrated that binary mixes of identical composition could 
have different organizations, depending on the surface energy and particle size of 
the fraction used. Actually, it has been demonstrated that it is possible to control 
the organization of binary mixes by modifying the particle sizes of the fractions 
blended if they have the appropriate surface energies [6] . 
Generally, only powders that form hard compacts under an applied pressure 
without exhibiting any tendency to cap or chip can be considered as readily 
compactible. The compactibility of pharmaceutical powders can be characterized 
by its tensile strength and indentation hardness, which can be used to determine 
three dimensionless parameters: strain index, bonding index, and brittle fracture 
index. 
To calculate the work of compaction during tableting, it is necessary to have 
accurate values of force and punch displacement. Differences in the dynamics of 
powder densifi cation between eccentric and rotary machines were pointed out by 
Palmieri et al. [14] after compression of microcrystalline cellulose, lactose monohydrate, 
and dicalcium phosphate dehydrate at different compression pressures. The 
effect of the longer dwell time of the rotary machine press on the porosity reduction 
after the maximum pressure is reached is more noticeable in a ductile material such 
as microcrystalline cellulose. It has been shown that Heckel parameters obtained in 
the rotary press are in some cases different from those recovered in the eccentric 
machine because of the longer dwell time, machine defl ection, and punch tilting 
occurring in the rotary press, although theoretically they could better describe the 
material densifi cation in a high - speed production rotary machine. 
Williams and McGinity [15] studied and compared the compaction properties of 
microcrystalline cellulose from six different sources using tableting indices. It was 
demonstrated that storage of compacts at elevated humidity conditions prior to 
determining the tableting indices decreased the magnitude of the tensile strength, 
dynamic indentation hardness, and bonding index. Based on the differences in 
physicomechanical properties observed for the tableting indices, the authors stated 
that microcrystalline cellulose products from different sources are not directly interchangeable 
and showed that the tensile strength, indentation hardness, bonding 
index, and brittle fracture index for compacts composed of microcrystalline cellu

lose in combination with either talc or magnesium stearate generally decreased as 
the amount of the lubricant was increased over the concentration range of 0 – 9%. 
Similar results were observed for admixtures of sodium sulfathiazole in combination 
with either talc or magnesium stearate. It was also demonstrated that the tensile 
strength, indentation hardness, and bonding index increased, and the brittle fracture 
index decreased as the percent of microcrystalline cellulose was increased in a 
mixture with sodium sulfathiazole. 
The results of a study conducted by Muller and Augsburger [16] suggest that 
the pressure – volume relationship determined during powder bed compression is 
affected by the instantaneous punch speed profi le of the displacement – time waveform 
for all materials studied, even though they deform by different mechanisms. 
It appears that the instantaneous punch speed profi le of the particular displacement 
– time waveform is a confounding factor of Heckel analysis. 
Moisute acts as a plasticizer and infl uences the mechanical properties of powdered 
materials for tablet compression. In the case of microcrystalline cellulose, at 
moisture levels above 5% the material exhibits signifi cant changes consistent with 
a transition from the glassy state to the rubbery state [17] . The possible infl uence of 
moisture on the compaction behavior of powders was also analyzed by Gupta et al. 
[18] . This work evaluates the effect of variation in the ambient moisture on the 
compaction behavior of microcrystalline cellulose powder. 
The work conducted by Gustafsson et al. [19] evaluated the particle 
properties and solid - state characteristics of two different brands of microcrystalline 
cellulose (Avicel PH101 and a brand obtained from the alga Cladophora sp.) 
and related the compaction behavior to the properties of the tablets. The difference 
in fi bril dimension and, thereby, the fi bril surface area of the two celluloses 
were shown to be the primary factor in determining their properties and 
behavior. 
The compaction properties of pharmaceutical formulations can be studied experimentally 
using a variety of techniques, ranging from instrumented production 
presses to compaction simulators, and methods of analysis. The results are usually 
plotted as porosity – axial stress functions, which is of interest to compare different 
materials. However, there are some drawbacks on this type of evaluation. As mentioned 
by Cunningham et al. [20] , this approach is defi cient once it considers only 
the average stress along the direction of compaction, ignoring radial stress transmission 
and friction. 
There have been some attempts to overcome the analysis of compaction 
problems, mostly by introducing numerical modeling approaches. The modeling 
approaches often used in compaction analysis are (a) phenomenological continuum 
models, (b) micromechanically based continuum models, and (c) discrete - element 
models. The parameters that should be analyzed when tableting is under development 
are as follows: 
1. Understanding the formulation and compositional effects on the compaction 
process, including axial loading and unloading along with ejection 
2. Determination of the stress distributions within the powder compact, including 
residual stresses 
3. Optimization of the tablet tooling design 
COMPACTIBILITY 1139

1140 TABLET COMPRESSION 
4. Estimation of the density distributions within a tablet that may infl uence dissolution 
or mechanical properties 
5. Estimation of the compaction force necessary to obtain tablets having given 
properties 
6. Taking into account the effect of the tablet material on the stress distribution 
on tooling to aid tool design 
7. Assessment of the origin of defect or crack formation 
8. Optimization of more complex compaction operations such as bilayer and 
trilayer tablets or compression - coated tablets 
The demonstration of the validity of the continuum - based modelling approach 
to tablet compaction requires familiarity with fundamental concepts of applied 
mechanics. Under the theory of such a mechanism, powder compaction can be 
viewed as a forming event during which large irrecoverable deformation takes place 
as the state of the material changes from loose packing to near full density. Moreover, 
it is important to defi ne the three components of the elastoplastic constitutive 
models which arose from the growing theory of plasticity, that is the deformation 
of materials such as powder within a die: 
1. Yield criterion , which defi nes the transition of elastic to plastic deformation 
2. Plastic fl ow potential , which dictates the relative amounts of each component 
of plastic fl ow 
3. Evolution of microstructure , which in turn defi nes the resistance to further 
deformation 
It is also known that the compression process can be described using static and 
dynamic models. In the case of static models, time is not considered, although it is 
a very important factor in the deformation process. The viscoelastic reactions are 
time dependent, especially for the plastic fl ow. 
Recently Picker [21] proposed a three - dimensional (3D) model to help explaining 
the densifi cation and deformation mechanism experienced by differently deforming 
materials during compression. According to the author, a single description of 
the processes during tableting is possible, and thus densifi cation and deformation 
properties can be clearly distinguished with a single model. This issue has been 
investigated over the last years, and a comprehensive approach has been developed 
for the analysis of compaction using continuum mechanics principles. This approach 
is based on the following components: 
1. Equilibrium equations (balance of forces transmitted through the material) 
2. Continuity equation (conservation of mass) 
3. Geometry of problem 
4. Constitutive behavior of powder (stress – strain behavior) 
5. Boundary conditions, including loading (e.g., displacement and velocity) and 
friction between tooling and powder 
6. Initial conditions (e.g., initial relative density of powder) 

Due to the signifi cant nonlinearity in material properties and contact stresses, a 
typical powder compaction problem cannot be solved analytically without major 
simplifi cations, and thus a numerical approach is required. 
The tableting properties of materials also depend on their deformation behavior. 
It is apparent that the tablet tensile strength is a strong function of the plastic work 
required for its formation but not a function of the elastic work recovered. Consequently, 
it is likely that strong and ductile interparticle functions, whose formation 
dissipated a signifi cant plastic work, result in strong and tough compacts [4] . 
It should be mentioned that the material parameters do change with compaction 
and the use of constant material values, which is often applied, is not necessarily 
appropriate given the evolving microstructure of the deforming powder. The experimental 
characterization and accompanying analysis allow these material properties 
to be evaluated within the comprehensive framework of continuum mechanics, 
which can be useful in analyzing and predicting the effects of constitutive behavior, 
friction, geometry, loading schedule, and initial condition, for example, initial relative 
density and powder fi ll confi guration. 
Ruegger and Celik [22] investigated the effect of punch speed on the compaction 
properties of pharmaceutical powders with one particular objective: to separate out 
differences between the effect of the compression and decompression events. Tablets 
were prepared using an integrated compaction research system. The loading and 
unloading speeds were varied independently of one another. In general, when the 
compression speed was equal to the decompression speed, the tablet crushing 
strength was observed to decrease as the punch velocity increased. When the 
compression speed was greater than or less than the decompression speed, the 
results varied, depending on the material undergoing compaction. The authors also 
stated that the reduction of the unloading speed had a similar effect on the direct - 
compression ibuprofen; however, even greater improvement in the crushing 
strength was observed when the loading speed was reduced. As a major conclusion, 
it was demonstrated that the strength of tablets can be improved and some tableting 
problems such as capping can be minimized or prevented by modifying the rates of 
loading/unloading. 
It is important to notice that, in the case of interacting materials, the compatibility 
of a binary mix will depend mostly on the compatibility of the percolating material 
[23] . Accordingly, several industrial applications can be made over these fi ndings. In 
the development phase, it is possible to modify the formulation of interacting 
systems to increase the drug content without losing the compatibility of the mix, 
whereas in the production phase, it is possible to increase the compatibility of a 
poorly compatible active ingredient by sieving or preferably by milling an excipient 
with good compression qualities without changing the composition of the mixture. 
6.6.4 TABLET COMPRESSION 
The process of tablet compression is divided into three stages: fi lling, compression, 
and ejection of the tablet (see Figure 1 ). 
During the fi rst stage of a compression cycle the lower punch falls within the die, 
creating a cavity which will contain the powder or granulation product that fl ows 
TABLET COMPRESSION 1141

1142 TABLET COMPRESSION 
from a hopper. The fi ll volume is determined by the depth to which the lower punch 
descends in the die. At this moment the particles of the powder or granulation 
product fl ow with respect to each other, thus resulting in a close packing arrangement 
and the physical characteristics of the material (particle size, particle size distribution, 
density, shape, and individual particle surface properties) associated with 
process parameters such as fl ow rate and compression rate, and the relationship 
between the die cavity and the particle diameter will defi ne the number of potential 
bonding points between the particles. The packing characteristic of the product to 
be compressed is greatly affected by the shape of the particles. Since the product to 
be compressed comprises components of different nature, the voidage of a closely 
packed system is considerably changed. 
When the upper punch goes down, its tip penetrates the die, confi ning the powder 
or granulation product, letting the particle rearrangement stage to continue and 
initiating the compression stage as the compression force is applied. As a result, 
forces resulting from the compression force are transmitted through the interparticulate 
points of contact created in the previous stage. The porosity of the powder 
bed is gradually decreased, the particles are forced into intimate proximity to each 
other, and stress is developed at the interparticulate points of contact. Once the 
particles have formed contacts, they will deform plastically under the applied load. 
Deformation of the particles will be characterized by elastic, plastic, fragmentation, 
or a combination of these phenomena, which will depend on the rate and magnitude 
of the external applied load, the duration of locally induced stress, and the physical 
properties of the product under compression. When the particles are in suffi ciently 
close proximity, they are bonded. Particles bond as a result of mechanical interlocking, 
which is described as entanglement of the particles, phase transition at the points 
of contact, and intermolecular forces, namely the van der Waals force, hydrogen 
bonding, and ionic bonding. 
After formation of the tablet by application of a compression force follows the 
decompression stage, where the compression force is removed and the upper punch 
leaves the die. Then, the formed tablet undergoes a sudden elastic expansion 
followed by a viscoelastic recovery during ejection when the lower punch moves 
upward. 
6.6.5 EQUIPMENT FOR TABLET COMPRESSION 
The equipment employed for tablet compression is generally categorized according 
to the number of compression stations and dislocation mode. Therefore, eccentric 
model presses have only one compression station (one die and one pair of punches, 
upper and lower) while rotary models have multiple compression stations (each 
station with one die and one pair of punches, upper and lower). The basic difference 
between the two types of compression equipment is that for eccentric models the 
compression force applied during compression is due to the upper punch whereas 
for rotary models it is mainly applied by the lower punch. 
A rotary tablet press machine (Figure 2 ) comprises a housing in which the compression 
set and subsets (upper and lower roller assemblies) are mounted, the turret 
head, the upper cams, the weight control assembly and the lower cams, the hopper, 

the feeder assembly, the take - off chute, the aspiration assembly, the gear box and 
the electrical unit, and the lubrication system. 
The compression zone is located on the back side of the equipment and employs 
a maximum load force limited by the type of tooling being used. It is of paramount 
importance to note that, if a load force is applied over the indicated limit, the press 
unit will not function properly, resulting in premature wear or possible damage to 
the tooling. The compression set comprises the hopper and feeder system, the die 
table, the upper and lower compression rollers, the upper and lower turrets, the 
excess - material scraper, the tablet stripper, the recirculation channel, and the aspiration 
system. 
The hopper is usually made of stainless steel and has the shape of a funnel to 
contain and deliver the product to be compressed. It may be provided with a window 
for the observation of the product level and may also be provided with low - level 
sensors that signal an alarm, shut off the engine, or activate the feeding mechanism 
to deliver the product when it falls below this level. The feeder system usually consists 
of three sections (in the case of force feeders) and is ideal for press performance 
at high speed. The fi rst section of a force feeder system is where the hopper is connected 
and is responsible for the fl ow of the product from the hopper to the next 
sections. The second section is where the die cavities are fi lled to their maximum 
capacities, and the third section is where the weight control adjustment takes place. 
These sections contain paddle systems which prevent packing of the product. The 
FIGURE 2 Rotary tablet press machine: ( a ) left-side view; ( b ) black - side view. (1) Cabinet, 
(2) compression, (3) turret, (4) gear, (5) weight control assembly and lower cams, (6) plate 
cams, (7) guarding, (8) hopper system, (9) feed frame assembly, (10) take - off chute, 
(11) aspiration assembly, (12) electrical system. 
(a) (b) 
EQUIPMENT FOR TABLET COMPRESSION 1143

1144 TABLET COMPRESSION 
speed of the paddles is adjustable and should be synchronized with the die table in 
order to prevent tablet weight variation. Better adjustment of the paddle speed 
could be achieved when keeping the lowest standard deviation of the compression 
force. The feeder system height above the die table is usually kept between 0.05 and 
0.10 mm. When the product to be compressed is of very fi ne particles, this height 
should be kept at 0.025 mm. 
Presses are commonly equipped with a powder aspiration system which is connected 
to a vacuum source in order to remove excess powder from the die table. 
This assembly is essential for a high - speed press working for extended periods of 
time. Special attention must be taken when the powder product comprises an active 
ingredient of fi ne particle size. In this case, aspiration should be minimal in order 
to prevent loss of the active ingredient. 
The compression subsets comprise the upper roller assembly and the lower roller 
assembly. The upper roller assembly is located on the roof plate of the press and 
utilizes an adjustment system for the regulation of the insertion depth. The lower 
roller assembly is located on the underside of the die table and utilizes a device for 
the regulation of the tablet edge thickness. 
The turret head is fi xed to the main shaft of the gear box. It is manufactured in 
two pieces (upper and lower) which guarantee the alignment between punches and 
dies. The gear box is mounted on the lower section under the die table and is responsible 
for transmitting the draft movement of the motor toward the turret head. 
The upper cams are responsible for guiding the upper punches around the circumference 
of the turret head. It comprises the fi lling stage track, which guides the 
upper punches in an up position during its passage over the feeder system; the upper 
lowering cam, which guides the upper punches down in order to keep their tips 
covering the cavities (precompression position) and directs the upper punches to 
their compression stage; the upper compression roller, which guides the upper 
punches to their compression position; and the upper fi lling cam, which guides 
the upper punches back to the fi lling track. 
The weight control assembly, which comprises the weight adjustment cam, is 
located in the lower section of the press and is regulated by an adjustment system. 
The lower cams are also located in the lower section of the press and comprise the 
preweight control (or fi ll cam), the weight adjustment cam, the lower lowering cam, 
and the ejection cam. The preweight control guides the lower punches to the full - fi ll 
position. The weight adjustment cam guides the lower punches up to the desired 
fi ll position. The lower lowering cam guides the lower punches to the precompression 
position. The ejection cam guides the lower punches and the formed tablets to 
the discharge position. It is recommended to operate the weight adjustment cam in 
the approximate center of the fi ll cam and because of this the fi ll cam is removable 
and available in different sizes having a range of approximately 10 mm with an 
increment range of 4 mm. The choice of the adequate fi ll cam for the operation of 
a tablet press with a particular product should be based on the density of the 
product. According to Figure 3 , the fi ll cam can be adequately chosen when taking 
into account the density of the material to be compressed and thus the material 
column height in the die cavity. 
Rotary tablet presses could be designed to be single, double, or triple sided. A 
single - sided press comprises one hopper, one set of compression rolls, and one take - 
off chute unit whereas double - and triple - sided presses comprise two and three each 

of these units, respectively. Irrespective of the design of the rotary tablet press, the 
compression cycle is described as follows (Figures 1 and 4 ). 
The powder or granulation product contained in the hopper fl ows to the feeder 
which spread the product through a large area over the die table in order to provide 
enough time to fi ll the die cavity. The die cavity is created when the fi ll cam guides 
the lower punches to the full - fi ll position and enters the feeder area. Note that the 
die cavity is fi lled with an excess of the product at this stage of the cycle. Right after, 
the weight adjustment ramp and head guide the lower punches to the desired fi ll 
position. The excess of the product is removed by the scraper and is pushed back 
by the excess product stripper when entering the recirculation channel. At this stage 
the lower punches are guided to the fi rst and second lower compression rols (precompression 
and main compression rolls, respectively) while the upper punches are 
guided by the upper lowering compression roll to the precompression position and 
to the compression position by the main compression roll. As the upper punch 
penetrates the die cavity until a predefi ned height, the main compression roll applies 
the compression load over the lower punch, compressing the product in the die. 
Soon after compression, the upper lifting cam allows the upper punch to leave the 
die cavity. Simultaneously, due to the ejection cam, the lower punch is pulled, 
FIGURE 3 Fill cam depth as function of product column height. 
Fill cam (mm) 
6 10 14 18 22 
Product 
column 
height 
(mm) 8 12 16 
1
2
3
4
5
6
7
8
9 
10 
11 
12 
13 
14 
15 
16 
17 
18 
19 
20 
21 
22 
EQUIPMENT FOR TABLET COMPRESSION 1145

1146 TABLET COMPRESSION 
ejecting the formed tablet to the die table. The ejected tablets are then stopped by 
a scraper and allowed to escape through a chute and collected. At this time the fi ll 
cam geometry makes the lower punch go down and a new compression cycle 
begins. 
6.6.6 TABLET PRESS TOOLING 
Punches and dies are essential tools in the tableting process and therefore are critical 
to the quality of the tablets produced. Both tools are designed for long life under 
normal conditions of working, but, in spite of this, they are not proof against careless 
handing. 
It is important for those working with a tablet press to be familiar with the terminology 
used in the industry concerning the punches and dies. Table 1 describes 
the commonly used terms related to press tooling. Some of the press tooling parts 
can be identifi ed in Figure 5 . 
When considering a tableting operation, it is important not only to select the 
appropriate press tooling in terms of dimensional data but also to consider the 
material of which tools are made. Performance of the press tooling will in part be 
a function of the material selected for its manufacture. Usually the material and 
hardness of the compression tooling are left to the manufacturer ’ s discretion. There 
are various types of steel available for the manufacture of press tooling. It is important 
to recognize the individual characteristics of the steel regarding its composition 
and the percentage of each constituent element. Usually only a small amount of 
alloying element is added to steels (usually less than 5%) for the purpose of improving 
hardness and strength corrosion resistance, stability at high or low temperatures, 
and control of grain size. Some of these elements are as follows: 
1. Carbon Principal hardening element. As the carbon content increases, its hardness 
increases. Increases the tensile strength of the steel. 
FIGURE 4 Compression cycle on rotary tablet press. (Courtesy of Thomas Engineering.) 
Upper lifting cam 
Upper lowering cam 
Feeder Scraper 
Ejection cam 
Tail over die 
Pull down after 
weight control cam 
Weight adjustment 
ramp and head 
Fill cam 
Main compression rolls 
Precompressionon 
rolls 
Main compression rolls 
Precompressionon 
rolls

TABLET PRESS TOOLING 1147 
TABLE 1 Tooling Terminology 
Band Area between opposing cup profi les formed by die wall 
Bakelite tip 
relief 
Undercut groove between lower punch tip straight and relief; assures 
sharp corner to assist in scraping product adhering to die wall 
Barrel/shank Surface controlled by turret punch guides to ensure alignment with die 
Barrel - to - stem 
radius 
Provides smooth transition from tip length to barrel 
Barrel chamfer Chamfers at ends of punch barrel, eliminates outside corners 
Barrel fl utes Vertical slots machined into punch barrel to reduce bearing surface 
and assist in removing product in punch guides 
Cup depth Depth of cup from highest point of tip edge to lowest point of cavity 
Die Component used in conjunction with upper and lower punches; accepts 
product for ocmpaction and is responsible for tablet ’ s perimeter size 
and confi guration 
Die bore Cavity where tablet is made, shape and size determine the tablet 
Die chamfer Entry of die bore 
Die groove Groove around periphery of die to allow die to be fi xed in press 
Die height/ 
overall length 
Overall height of die 
Die lock Mechanism used to lock die in position after it is installed in die table 
Die outside 
diameter 
Outside diameter of die, compatible with die pockets in press 
Die taper Gradual increase in die bore from point of compaction to mouth of 
bore, assists ejection 
Head End of punch which guides it through press cam track 
Head/dwell fl at Flat area of head that receives full force of compression rolls at time 
that tablet is being formed 
Inside head 
angle 
Area of contact with lower cam and upper cam 
Key Prevents rotational movement of punches ensuring alignment to 
shaped and multihole dies 
Keying angle Relationship of punch key to tablet shape; position will be infl uenced 
by tablet shape, take - off angle, and turret rotation 
Land Area between edge of punch cup and outside diameter of punch tip 
Neck Relieved area between head barrel which provides clearance for die 
Outside head fl at 
angle/radius 
Contact area with press cams and initial contact with pressure rolls 
Overall length Total punch length as measured from head fl at to end of tip 
Tip face/cup Portion of punch tip that determines contour of tablet face including 
tablet embossing 
Tip length Straight portion of stem effective inside die bore 
Tip relief Portion of punch stem which is undercut or made smaller than punch 
tip straight; most common for lower punches in order to reduce 
friction from punch tip and die wall 
Tooling set Complete set of punches and dies to accommodate all stations in tablet 
press 
Tooling station Upper punch, lower punch, and die which accommodate one station in 
tablet press 
Relief/undercut Mechanical clearance between stem and die bore, sharp edge between 
tip straight and undercut areas acts to clean die 
Stem Area of punch opposite head which begins at end of barrel and 
extends to tip 
Working length Length of punch from bottom of cup to head fl at; together, upper and 
lower working lengths control tablet thickness and weight; also 
known as overall length, bottom of cup (OLBC) 

1148 TABLET COMPRESSION 
FIGURE 5 Identifi cation of common parts of press tooling (upper and lower punches and 
die) according to ISO 18084, 2005: (1) upper punch, (2) lower punch, (3) die, (4) key, (5) land, 
(6) stem, (7) barrel - to - stem chafer, (8) cup depth, (9) tip face, (10) blended land, (11) face, 
(12) bore, (13) die grove, (14) protection radius or shoulder, (15) chamfer or radius, (16) outer 
diameter, (17) tip straight, (18) relief, (19) barrel - to - stem radius, (20) working length of tip, 
(21) overall length, (22) barrel, (23) working length, (24) barrel - to - neck radius, (25) neck - to - 
head radius, (26) inside head angle, (27) neck, (28) head, (29) head outer diameter, 
(30) outside head angle, (31) head fl at, (32) key orientation angle, (33) upper punch face 
key position, (34) barrel diameter. 
2. Manganese Increases ductility and hardenability of the steel. Also increases 
the rate of carbon penetration during carbonizing and imparts excellent wear 
resistance. 
3. Nickel Improves the toughness and impact resistance of the steel and mildly 
increases its hardness. 
4. Chromium Increases the hardness of the steel and improves its wear or abrasion 
resistance. It helps to limit grain size. If added in amounts greater than 
5%, it can impart corrosion and wear resistances. 
5. Molybdenum Improves hardenability and increases tensile strength of the 
steel. 
6. Vanadium Produces a fi ne grain size and improves fatigue strength of the steel, 
just like molybdenum. 

TABLET PRESS TOOLING 1149 
7. Tungsten Is used in tool steels to maintain hardness at elevated 
temperatures. 
8. Copper Increases corrosion resistance; nevertheless its content has to be 
controlled, otherwise the surface quality and hot - working behavior are 
compromised. 
The carbon steels comprise alloying elements not exceeding the defi ned limits of 
1% carbon, 0.6% cooper, 1.65% manganese, 0.4% phosphorus, 0.6% silicon, and 
0.05% sulfur. For the alloy steels, the limits exceed those for the carbon steels and 
may also include elements not found in carbon steels. The alloy steels have a specifi c 
designation according to the American Iron and Steel Institute (AISI). Such designation 
is a four - digit number where the fi rst digit stands for the class of the alloy 
(e.g., 1, carbon; 2, nickel - chromium; 3, molybdenum; 4, chromium), the second digit 
designates the subclass of the alloy, and the last two digits designate the amount of 
carbon in 0.01%. The stainless steels comprise at least 10% chromium with or 
without the addition of any other alloying element. The tool steels are carbon steel 
alloys with an excess fo carbides which impart hardness and wear resistance. According 
to the AISI, tool steels are grouped as water hardening (W), shock resisting (S), 
cold - work oil hardening (O), cold - work medium - alloy air hardening (A), cold - work 
high - alloy high chromium (D), low alloy (L), carbon tungsten (F), low - carbon mold 
steels (P1 – P19), other mold steels (P20 – P39), chromium - based hot work (H1 – H19), 
tungsten - based hot work (H20 – H29), molybdenum - based hot work (H40 – H59), 
high - speed tungsten based (T), and high - speed molybdenum based (M). 
The appropriate steel for press tooling should be selected based upon the toughness 
and wear resistance required by the application, and therefore it is mandatory 
to have satisfactory knowledge regarding the abrasiveness, corrosiveness, and lubricity 
of the product intended for compression as well as the desired dimensions of the 
punch tip. The toughness of the steel regards its ability to resist shock and its wear 
resistance regards the ability to resist physical damage or erosion due to product 
contact. 
For a clear understanding, the Thomas Engineering Press Tooling Manual [24] 
states the following: 
Punches manufactured from high carbon/high chromium steel may exhibit improved 
wear resistance characteristics, however under extreme compression force, the cup may 
crack due to the brittle nature of the steel. Steels with lower carbon and chromium 
levels will act conversely. While these steels may be useful in some applications, the 
majority will require a more moderate balance of toughness and wear resistance. 
Steel selection for dies is not as critical. In most cases, high wear resistance steel is 
preferred. 
The bulk of pharmaceutical tablet press punches are manufactured from S1, S5, S7, or 
408 (11% chromium, 8% nickel) tool steel. The S series steels provide a good combination 
of shock and wear resistance and have a proven record of performance in tableting 
operations. At one time, 408 or 3% nickel steel was the industry standard because of 
its superior shock resistance toughness . The S grades however, which have only a slight 
loss in ductibility by comparison, offer much improved wear characteristics and have 
all but replaced 408 as the preferred general purpose punch steel. A2, D2 and D3 are 
high carbon/high chromium steel used for their excellent wear resistance. Among all 
the steels commonly used for press tooling, D3 has the highest wear resistance. However, 

1150 TABLET COMPRESSION 
its low toughness rating typically limits its use to dies only. D2 rates slightly lower in 
abrasion resistance than D3 but its increased toughness makes it suitable for punch 
use, provided the cup design is not too fragile. A2 is a compromise between the general 
purpose S grades and D2 in both toughness and wear. It can be used for punches as 
well as dies. 
Tungsten carbide, while not actually a steel, is extremely wear resistant and is commonly 
used to line dies. Punch tips can be manufactured from tungsten carbide; 
however, the cost of tooling is quite high and restricted to applications where tip fracture 
due to high compression forces is not likely. 
Ceramic materials such as partially stabilized zirconia can also be used as die liners. 
Ceramics offer high wear and corrosion resistance and lower tablet ejection forces than 
either steel or carbide due to their low coeffi cient of friction. 
S1, S7 and 408 provide some protection against mildly corrosive materials. More severe 
corrosion problems however, demand the use of stainless steel (440C) tooling. From 
the standpoint of wear, 440C falls between the S and D grades of tool steel. Its low 
toughness rating (comparable to D3) requires a strong cup design if tip fracture problems 
are to be avoided. 
One measure of tool steel quality is the rate of inclusions. Inclusion are unwanted 
impurities or voids and are present to some degree in all steels. After heat treatment, 
inclusions give rise to localized areas of stress concentration where microscopic cracks 
can later develop. Remelting of the steel at the foundry will further reduce a tool steel ’ s 
level of impurities; therefore improving the quality of the steel and subsequently its 
performance in the tooling environment. In cases where punch tip fracture is a problem, 
tooling suppliers may recommend a remelted or premium grade of particular steel as 
a means of eliminating the problem. 
Concerning the confi guration of compression tooling, the most commonly used 
are the so - called B (19 mm, or 3/4 in.) and D (21 mm, or 1 in.) tooling types. Additionally, 
these two types are classifi ed into three specifi cations: the North American 
TSM ( Tableting Specifi cation Manual ) [25] , the European Union (EU) standard, 
and the Japan Norm (JN). The North American TSM is used in the United States 
and is the only standard offi cially supported by the governing body and published 
by the American Pharmacists Association. The EU standard and the JN are generally 
used in Europe and the Far East, respectively. In spite of the existence of these 
standard specifi cations, there are tablet press manufacturers that use their own 
confi gurations for tooling which have the disadvantage of being restricted to a specifi 
c tablet press. Figures 6 and 7 depict the three standard confi gurations of compression 
tooling. In addition, the International Organization for Standardization 
(ISO) standard 18084:2005 [26] comprises specifi cations of the main dimensions, 
including tolerances and characteristics of punches and dies. 
Regarding the importance of compression tooling to the performance of the 
tablet press and the quality of the tablets, it is of paramount importance that 
punches and dies are handled with care. The fi rst criterion is the identifi cation 
of the tooling; that is, punches and dies should be identifi ed according to the standard 
and be designated by “ upper punch without key, ” “ upper punch with key, ” 
“ lower punch with key, ” “ lower punch without key, ” or “ die, ” the reference of the 
standard (e.g., TSM, EU, JN, ISO), and the punch or die diameter. Punches and 
dies should also have a marking that includes at least the manufacturer ’ s identifi cation, 
the number of the punch in the series, and/or the identifi cation number. Upon 

TABLET PRESS TOOLING 1151 
FIGURE 6 Tooling standards confi gurations. 
receipt, after manufacturing, and prior to inspection, the punches and dies should 
be carefully and thoroughly cleaned and dried. Then, tooling should be lightly oiled, 
packed, and stored in a dry, cool place. 
Damage to the punches and dies should be avoided. Therefore, they should not 
be transported from place to place without protective package. During transportation, 
installation, and removal of tooling from the tablet press, cleaning, inspection, 
and storage, care must be taken to avoid hitting the tips of the punches. 
The visual and dimensional inspection of punches and dies should be carried out 
periodically. Visual inspection should be performed each time punches and dies are 
installed in and removed from the tablet press. Under normal conditions, slight wear 
is to be expected. When abnormal or excess wear is detected, the cause should be 
immediately investigated, inspecting the cams or components which touch the 
affected area of the tool. The importance of visual inspection resides on the fact 
that it may ensure the optimum life of the punches and dies, performance of the 
tablet press, and consistency and appearance of the tablets. In addition to the visual 
inspection, it is also recommended that dimensional inspection be performed at 
specifi c intervals throughout the life span of the punches and dies. The dimensional 
inspection not only ensures the consistency of hardness, weight, and thickness of the 
tablets, but also proves to be critical in diagnosing potential and real problem areas 
with regard to the tableting process and press. A typical schedule for the dimensional 
inspection may be as follows: 50%, 75%, 85%, 90%, and 95% of the historical 
or projected life cycle of the punches and dies. Therefore, the history or data base 
should be maintained for each set of tooling. Nevertheless, there is no general 
agreement on what dimensions of the punches and dies should be included in a 

1152 TABLET COMPRESSION 
FIGURE 7 Tooling head confi gurations. 
Standard TSM (B-type punches) TSM domed (B-type punches) 
Standard TSM (D-type punches) TSM domed (D- type punches) 
EU standard Japan norm 
dimensional inspection. Some believe that a 100% inspection should be carried out 
while others defend that only critical dimensions (e.g., working length, cup depth, 
and overall length) should be inspected, believing that measuring any other dimension 
is either unnecessary, since it rarely if ever changes and therefore is not worthy 
of the time and expense of measuring, or cannot be properly measured with current 
equipment and is better served by a visual inspection. What is important when 
inspecting compression tooling is that the dimensional values are consistent within 
the set and tolerances and within specifi cations. Before proceeding with inspection, 
the measuring instruments should be calibrated to be certain that the dimensional 
values obtained are accurate and true. 

6.6.7 TABLE ENGRAVING 
Engraving is the most common method for tablet surface marking identifi cation. 
The engraving method could be embossed (letters or symbols are raised on the 
tablet surface and cut into the punch tip face) or debossed (letters or symbols are 
cut into the tablet surface and raised on the punch tip face). For engraving on the 
tablet surface some specifi cations should be considered: stroke width, angle of 
engraving, radius, depth, spacing, and engraving area. 
Generally, for uncoated tablet application, a stroke width between 15 and 20% 
of the letter height having an engraving angle of 30 ° is recommended. The radius 
should be between 50.8 . m (0.002 in.) and a value derived from dividing the stroke 
width by 2 times the cosine of the engraving angle. It is important to note that radii 
smaller than 50.8 . m or exceeding the maximum value are diffi cult to machine since 
it may decrease depth and defi nition of engraving. The depth is a function of the 
engraving angles, stroke, and radius for a given tablet size and, as a general rule, the 
depth should not exceed 50% of the stroke width, or no less than 88.9 . m (0.0035 in.). 
Spacing between letters or symbols should be a minimum of one stroke width. The 
available engraving area is based upon letter distortion due to the curvature or 
radius of the cup and thus, as a general rule, letter distortion is defi ned by the ratio 
of the outside depth of the engraving to the specifi ed depth. Generally, distortion 
is present when this ratio exceeds 1 : 3. 
When engraving is considered for fi llm - coated tablets, the recommended stroke 
width should be the same as recommended for uncoated tablets. The recommended 
engraving angle should be 35 ° . However, engraving angles up to 40 ° can be used 
in extreme applications to allow coating solution fl ow. For stroke widths of 203.2 . m 
(0.008 in.) or less, a 30 ° angle is recommended to maintain minimum engraving 
depth. The recommended radius should be between 76.2 and 152.4 . m and a value 
derived by dividing the stroke width by 2 times the cosine of the engraving angle. 
It is important to note that radii will be determined by the fl owability of the coating 
solution and coating process. As a general rule, stroke depth should be at least 
177.8 . m. However, shallower depths can be used, provided that the fi lm coating 
process is properly developed taking this factor into account. The spacing between 
the letters and symbols and the available engraving area considerations are the same 
as for uncoated tablets. The following equations can be used to determine stroke 
width and engraving radii: 
S H H = - 0 15 0 20 . . (6) 
R R 
S 
min max m 
cos 
= = 50 8 
2 
. . 
. 
(7) 
6.6.8 TABLET SHAPE AND PROFILE 
The more popular standard geometric shapes of tablets are the round and the caplet 
shapes. Other tablet shapes include the oval, elliptical, square, diamond, rectangular, 
and polygonal. The shape of tablets plays an important role in terms of aesthetics, 
process (printing, fi lm coating, packaging, and shipping), and acceptability by the 
consumer (identifi cation, help with swallowing). 
TABLET SHAPE AND PROFILE 1153

1154 TABLET COMPRESSION 
In terms of design, the profi le of a tablet also plays an important role in the aesthetics, 
packaging, orientation for printing processes, and handling. The profi le of a 
tablet is important in the fi lm coating process and even in helping with the swallowing. 
Applying a bisect score onto the tablet surface enables the tablet to be easily 
divided into smaller dosage amounts. 
Nowadays it is common for tooling suppliers to use software to provide 2D and 
3D technical drawings of tablets and tooling. Such software may provide accurate 
details of tablets and tooling using only tablet dimensions as input and therefore 
enables fast evaluation by the manufacturing department prior to ordering prototypes. 
Figure 8 illustrates some tablet shapes and profi les. 
Flat - face, bevelled - edge tablets have many advantages due to their fl atness, which 
provides the most compact tablet weight per volume weight, uniform hardness since 
the compression force is exerted evenly on the cup face, and engraving with no distortion. 
This tablet profi le proves to be ideal for small tablets, especially when 
engraved, although the engraving area may be limited by the 381 . m (0.015 in.) 
radius on the bevel. On the other hand, compression tooling displays an inherent 
weakness in the punch cup design at the point where the bevel edge meets the cup 
fl at. Attention must be paid since these types of tablets cannot be coated as they 
will stick together, or twin. 
FIGURE 8 Common tablet shapes and profi les: (A) standard convex, (B) compound cup, 
(C) convex beveled, (D) fl at faced plain, (E) fl at faced bevel edged, (F) fl at faced radius 
edged, (G) lozenge, (H) modifi ed ball, (I) core rod with hole in center, (J) capsule, (K) modi- 
fi ed capsule, (L) oval, (M) bullet, (N) arrow head, (O) triangle, (P) arc triangle, (Q) square, 
(R) pillow or arc square, (S) rectangle, (T) modifi ed rectangle, (U) diamond, (V) pentagon, 
(W) hexagon, (X) octagon, (Y) almond. 
A B C D E 
F G H I J 
K L M N O 
P Q R S T 
U V W X Y 

Shallow and standard concave tablets have the great advantage of displaying a 
maximum allowable area available for engraving without distortion as a result of 
the moderate curvature of the cup profi les and the absence of a bevel. The shallow 
and standard cup confi gurations are the strongest profi les per punch tip diameter. 
In addition, such profi les allow consistent distribution of the compression force over 
the cup face due to the slight curves involved in the cup, thus contributing to the 
production of tablets of uniform hardness. Nevertheless, caution must be taken 
concerning the cup depth since when it approaches the cup edge it may be less than 
the depth of the engraving. The major disadvantage of these profi les may be due to 
the angle of the cup profi le to the tablet sidewall, which may lead to chipping at the 
tablet edge during fi lm coating or handling. 
Caplet - shaped tablets are easier to swallow, aesthetically pleasing, and chipping 
at the tablet edge generally does not occur during fi lm coating or handling due to 
the angle of the cup profi le to the tablet sidewall. Nevertheless, the increased curvature 
of the cup reduces compression force by approximately 50% compared to 
the shallow and standard concave profi les. Distortion of engraving may also be a 
problem because of the more extreme curvature. During fi lm coating caplet tablets 
have the potential of sticking together, or twinning, as the tablet sidewalls are parallel. 
This problem could be alleviated by applying a 76.2 mm (0.003 in.) drop (15.24 – 
20.32 cm, or 6 – 8 in. radius) to the sidewalls. 
The concave oval profi le displays a maximum allowable area for engraving and 
a uniform distribution of the compression force over the cup face. Such an advantage 
is a consequence of the mild curvature of the cup profi le and absence of a bevel. 
Structurally, concave oval tablets are the strongest of the non - round - shaped tablets. 
However, due to the angle of the cup profi le to the tablet sidewall, chipping at the 
tablet edge may occur during fi lm coating or handling. 
The compound cup profi le could be used to provide round or oval tablets. This 
profi le provides a good tablet weight per volume but simultaneously presents a 
weak cup edge, thus being the weakest of all cup confi gurations. Because of this, the 
maximum compression force is limited to the minor cup radius on the round shapes 
and the minor cup radius on the minor side for the oval shapes. In addition, the 
available engraving area is limited to the blending point of the two radii. 
6.6.9 TABLET BISECT 
Usually known as score or break line, the tablet bisect has the purpose of easily 
breaking the tablet in predetermined small dosages. According to the TSM, the 
bisect types range from the most functional (the pressure sensitive, or type G) to 
the least functional (partial, or type H). Each bisect type has its own characteristic, 
as can be seen in Figure 9 . Generally, the bisect is placed on the upper punch, especially 
when its depth exceed 40% of the cup depth, in order to avoid problems 
during ejection of tablets. Nevertheless, the bisect can be placed on the lower punch 
either when the upper punch is supposed to contain embossed characters or printing 
that makes diffi cult the existence of the bisect or when its depth does not exceed 
40% of the cup depth. When it is desired to apply a bisect to the upper tablet ’ s 
surface but there is interference of engraving or printing, then a modifi ed bisect 
design should be considered. 
TABLET BISECT 1155

1156 TABLET COMPRESSION 
When considering applying a bisect to a tablet ’ s surface, careful attention should 
be taken with respect to the tablet ’ s cup depth, band thickness, and hardness. Considering 
these aspects, the specifi cations for the bisect size are determined taking 
into account the tablet ’ s size, engraving or printing, and desired bisect design. 
The TSM acknowledges two different confi gurations of bisect for concave tablets: 
protruding and cut fl ush. The protruding confi guration follows the curvature of a 
radiused cup and extends past the tip edge of the punch. The cut fl ush confi guration 
FIGURE 9 Tablet bisect for concave tooling (according to TSM): (A, B, C) pressure - 
sensitive (type G), (D, E, F) cut through (type D) or European style, (G, H, I) decreasing 
(type C), (J, K, L) standard protruding (type A), (M, N, O) standard (type E), (P, Q, R) short 
(type B), (S, T, U) partial (type H). 
Top view Profile (end) view Profile (side) view 
A B C 
D E F 
G H I 
J K L 
M N O 
P Q R 
S T U 
50% max of 
band thickness 
95% of 
cup depth 
75% of 
cup depth 
Cup radius
Bisect 
radius 
Break 
radius 
Break 
radius

is the most popular bisect confi guration since one may experience problems with 
the protruding confi guration. This is explained by the fact that the protruding bisect 
may run into the tip edge of the lower punch if they become too close during the 
compression cycle of the press. 
Among the bisect styles acknowledged by the TSM, the cut - through, also known 
as the European style, can only be applied on radiused cup designed tablets. Other 
styles are the standard, the short, and the partial bisects. Compared to the standard 
style, the cut - through style is said to have the advantage of letting patients better 
break the tablet into smaller subunits. On the other hand, because the cut - through 
is wider at the center, it decreases the available tablet surface area for engraving or 
printing. 
6.6.10 PROBLEMS DURING TABLET MANUFACTURING 
Due to either formulation or equipment, some problems can arise during the tablet 
compression process, such as capping and lamination, picking and sticking, mottling, 
double printing, weigh variation, and hardness variation. It is the early detection 
and accurate diagnosis of any of these fl aws that can avoid tablet compression 
process failure and consequently improve its reliability, safety, reduce process downtime 
and the overall operating cost. 
Often, some of the above - mentioned problems are not detected during the development 
of a particular tablet formulation, only appearing during scale - up as the 
processing speed is increased. Some of the problems experienced during tableting 
can be solved by shifting the formulation or alleviated by altering the tableting 
conditions. 
6.6.10.1 Capping and Lamination 
Capping and lamination are common problems that can be experienced during 
tableting. Capping is defi ned as the splitting of one or both lids of a tablet from its 
body. Lamination is a precursor to capping since it involves the occurrence of layers 
in a compact parallel to the punch face. Sometimes capping is noticed not during 
the process but during physical testing, such as friability and hardness. 
An incipient theory proposed by Train [27] related lamination to radial elastic 
recovery of the compacted material during ejection. A once - accepted theory formulated 
that capping and lamination are the result of air entrapped in the tablet 
under pressure which tries to escape during ejection [28] . This theory is no longer 
widely accepted. Disagreement arises from the fact that some formulations cap or 
laminate even at low press speeds. Today, it is believed that the entrapped air may 
be related to capping but does not affect lamination. 
A widely accepted theory for lamination presented by Long [29] and reformulated 
by Ritter and Sucker [30] attributes capping to the residual die wall pressure. 
This pressure is said to cause internal shear stresses in the tablet causing the propagation 
of cracks, which results in lamination or capping. The propagation of cracks 
can be prevented by plastic relaxation of shear stresses. Therefore, materials having 
suffi cient plasticity may not be susceptible to lamination. Some properties of the 
powder mixture, such as moisture content, type and amount of the binder, and 
PROBLEMS DURING TABLET MANUFACTURING 1157

1158 TABLET COMPRESSION 
particle size, are important formulation variables that could be assessed in order to 
impart plasticity, thus diminishing capping and lamination tendencies. 
Normally, drugs such as paracetamol, mannitol, ibuprofen, phenazone, and mefenamic 
acid have poor compression properties and produce tablets that are weak 
and frequently exhibit capping. Materials that deform elastically or exhibit time 
dependence are more susceptible to capping and lamination and/or strength reduction, 
especially as tableting rate is increased. The effect of punch velocity is most 
marked when transferring a material from an eccentric to a rotary press or when 
scaling up to larger production size tablet presses. 
In addition to the possible causes of capping and lamination discussed previously, 
one should also consider the possibility that shape of the tooling and tooling defects 
are sources of capping. In such cases the problem can simply be alleviated by repairing 
or altering press tooling. 
Usually the process of capping can be evidenced as an increase in tablet height 
within a few seconds after tablets are ejected from the die. 
A technique generally applied to characterize and prevent the capping and lamination 
of a material intended to be compacted is using the brittle fracture index 
(BFI). The BFI was designed by Hiestand et al. [31] and measures the ability of a 
material to relieve stress by plastic deformation around a defect. It is obtained by 
applying Equation (8) and compares the tensile strength of a tablet with a hole in 
its center ( T 0 ), which acts as a built - in stress concentrator defect, with the tensile 
strength of a similar tablet without a hole ( T ), both at the same relative density: 
BFI = ( ). ... 
... 
0 5 1 0 . 
T
T 
(8) 
It is said that a material showing a moderate to high BFI value ( > 0.5) is prone to 
laminate and cap during the process. A low value of BFI is desirable to minimize 
lamination and capping during tablet production. 
Indentation hardness is another measure which fi nds wide application in the 
pharmaceutical industry for the assessment of capping and lamination tendency. The 
indentation hardness measurement employs an indentation hardness tester and is 
defi ned as the hardness of a material determined by either the size of an indentation 
made by an indenting tool under a fi xed load or the load necessary to produce 
penetration of the indenter to a predefi ned depth. An instrumented indentation 
hardness tester can be employed for that purpose since it has the ability to measure 
the intender penetration ( H ) under the applied force ( F ) throughout the testing 
cycle and is therefore capable of measuring both plastic and elastic deformation of 
the material under test. 
Another technique for the assessment of capping and lamination tendency which 
has been increasingly employed in the research - and - development phase of tablet 
manufacturing is acoustic emission. This technique relies on the fact that an abrupt 
change in stress within a material to be compacted generates the release of a transient 
strain energy designated as acoustic emission which results in a mechanical wave that 
propagates within and on the surface of a structure [5, 32] . Thus, this technique can 
discriminate between capped and noncapped tablets based on comparing the measured 
level of acoustic emission energy against a decision threshold. 

If it is desirable to overcome capping and lamination during the tableting process, 
the use of ultrasound - assisted presses could be a reliable solution. However, use 
this technique is still very recent since reports in the scientifi c literature extend only 
over the last decade [33] . 
In general, when capping and lamination are possible problems during tablet 
manufacture, an option could be the slower removal of force during decompression. 
This could be useful since capping tendency increases with increasing rates of 
decompression. However, better improvements could be achieved if the compression 
and decompression events are treated separately. By determining the effect of 
reducing either the loading or unloading speeds on the individual materials, it could 
be possible to increase crushing strength and eliminate or minimize the incidence 
of capping and lamination to greater extents. Thus, there is the need for a machine 
that is capable of customizing compaction profi les so that each formulation can be 
manufactured under an optimum set of conditions. 
6.6.10.2 Picking and Sticking 
Picking refers to adherence of powder to the punch surface. It is more problematic 
when the punch surfaces are engraved with logos or letters such as B, A, or O in 
order to produce debossed tablets. Sticking occurs when powder tends to adhere to 
the die leading to the development of an additional pressure to surpass friction 
between the formed compact and the die wall. As a result, the produced tablets 
show a rough surface at their edges. Furthermore, sticking can cause picking or 
damage the press punches by blocking the free movement of the lower punches 
leading to an increase of compaction pressure. 
Various approaches can be used to solve picking and sticking problems during 
tablet manufacturing, namely optimization of press tooling, process parameters, and 
formulation. Generally, it is important to fi nd the optimal combination of formulation 
and process parameters, particularly when market image tablets are to be 
produced. 
In relation to formulation adjustment, an antisticking agent (talc is commonly 
used for this purpose) can be added to the powdered formulation in order to 
eliminate picking and sticking during manufacturing. Colloidal silicon dioxide may 
be the right choice when picking is evident since this excipient can impart smoothness 
to the punch surfaces. However, when adding colloidal silicon dioxide to the 
powder formulation, it would be necessary to add an extra lubricant in order to 
avoid sticking and facilitate ejection of tablets from the dies. In addition to the 
need for an extra excipient in the powder formulation, press tooling may need to 
be adjusted to improve tableting. For the production of market image tablets, 
logo or letters on the punches should be as big as possible. Additionally, punch 
tips may be plated with chrome in order to give a smooth and nonadherent 
surface. 
When a lubricant such as stearic acid or propylene glycol or any other raw material 
of low melting point is present in the powder formulation, the heat generated 
during tableting may cause softening of these ingredients, thus leading to sticking. 
To overcome this problem, it may be needed to refrigerate the powder load to be 
tableted or to equip the press machine with a cooling unit. 
PROBLEMS DURING TABLET MANUFACTURING 1159

1160 TABLET COMPRESSION 
6.6.10.3 Mottling 
Mottling is defi ned as an uneven coloration of tablets or nonuniformity of color 
over the tablet surface. One of the possible causes of mottling may be the difference 
in color between the active principle and excipients, but sometimes it may be the 
result of degradation of the active ingredient which imparts spot zones over the 
surface of the tablets. 
Nonetheless, when colored compressed tablets are needed for aesthetic reasons, 
the foremost cause of mottling is dye migration to the periphery of granules 
during the drying process [34] . To overcome this problem, one should consider 
changing the solvent used for wet granulation or the binder agent, using a low drying 
temperature, or decreasing the particle size of the excipient. Another way to overcome 
mottling was demonstrated by Zagrafi and Mattocks [35] and suggests the 
inclusion of an adsorbent agent such as wheat or potato starch to the formulation. 
The adsorbent agent is said to adsorb the dye, retarding its migration then decreasing 
mottling. 
6.6.10.4 Weight and Hardness Variation 
Weight and hardness variation are common problems experienced when tableting. 
Tablet weight is mainly affected by factors such as powder variation, tablet press 
condition and tooling, and fl ow of powder on the tablet press. 
Inconsistent powder or granulate density and particle size distribution are 
common sources of weight variation during tablet compression. Problems related 
to the density of the powder or granulate are often associated with overfi lling of 
the die and recirculation of the product on the tablet press. A variation of particle 
size distribution of the powder or granulate can be the result of segregation due to 
transfer or static electricity. It might also vary because the product cannot withstand 
the handling and mechanical stress it undergoes before reaching the tablet press. 
Weight variation can arise as a result of a poorly prepared or operated tablet 
press. To solve this problem, one should inspect the press performance. Attention 
must be taken when dealing with a new die table on a load tablet press. In such a 
case, operation of the tablet press must regard the up - and - down motion of the 
punches within 76.2 . m of the setting without neglecting the conditions of the pressure 
rolls and cams. 
Inspection of the critical dimensions of tablet press tools is recommended. At 
least three dimensions of the upper and lower punches should be inspected: the 
working length, the cup depth, and the overall length. The working length is the key 
factor affecting tablet weight. Therefore, the length of each punch must be correct 
and identical. The cup depth and the overall length are not critical with regard to 
controlling tablet weight. Therefore comprehensive inspection and evaluation of the 
press tooling are essential to minimize deviation of tablet thickness, weight, and 
hardness. 
During the course of a compression operation it is also important to not neglect 
the level of the product in the hopper. Head pressure is a critical factor related to 
the amount of product in the hopper. The more product present in the hopper, the 
greater the head pressure, and vice versa. Therefore, when the head pressure varies, 
so does the weight of the tablets. So, in order to maintain a constant head pressure, 

thus reducing a potential variation of weight, compression should be conducted 
within a narrow range of the powder or granulate product in the hopper. 
The fi ll cam is another factor that can have a profound effect on tablet weight. 
The choice of an adequate fi ll cam regarding some characteristics of the powder or 
granulate product allows the die cavity to be properly overfi lled. Usually, in order 
to maintain consistent tablet weight during compression, it is recommended to 
overfi ll the cam by 10 – 30% of its volume. Basically, any tablet press part that is 
ultimately related to the powder product fl ow can have a mild or profound impact 
on weight control. It is important to remember that the scraper blade tends to 
become worn by die table rotation and powder product abrasion. Therefore, periodic 
inspection of its condition and replacement are recommended. Nevertheless, 
the scraper blade proper condition is important but also its adjustment since if it is 
not set up correctly, powder product may accumulate on the die table, leading to 
problems with weight control. 
Tablet hardness variation is intimately related to weight variation and, accordingly, 
to the infl uence of compression variables such as dwell time, tablet thickness, 
and working length of the punches. Thus, to solve a hardness variation, consistency 
of the tablet weight must be checked fi rst. If the predefi ned weight is achieved but 
hardness is out of limits, then precompression and compression forces should be 
adjusted while keeping tablet thickness within target limits. Although dwell time 
might be a source of hardness variation, adjustment of this parameter may be detrimental 
to the whole process since the compression rate is slowed. Occasionally, 
when the tablet weight target is kept within limits but hardness varies, the problem 
may be due to the formulation. As mentioned previously, the correct use of punches 
and dies is of paramount importance and periodic inspection is mandatory in order 
to ensure the compression process has not been compromised. So, when it becomes 
hard to achieve tablet hardness, it is recommended to fi rst verify tablet weight and 
thickness consistency and then try to adjust the precompression. The choice to 
increase the tablet weight even if it is within limits or to reduce the tablet press 
speed is not convenient and should be used only when there are no more options. 
REFERENCES 
1. C elik , M. ( 1994 ), Compaction of multiparticulate oral dosage forms , in Ghebre - Sellassier , 
I. , Ed., Multiparticulate Oral Drug Delivery , Marcel Dekker , New York , pp. 181 – 216 . 
2. Parrot , E. ( 1990 ), Compression , in Lieberman , H. L. , and Schwartz , J. B. , Eds., Pharmaceutical 
Dosage Form , 2nd ed., Marcel Dekker , New York , pp. 201 – 244 . 
3. Shlieout , G. , Wiese , M. , and Zessin , G. ( 1999 ), A new method to evaluate the consolidation 
behavior of pharmaceutical materials by using the Fraser - Suzuki function , Drug. Dev. Ind. 
Pharm. , 25 ( 1 ), 29 – 36 . 
4. Mohammed , H. , Briscoe , B. J. , and Pitt , K. G. ( 2006 ), A study on the coherence of compacted 
binary composites of microcrystalline cellulose and paracetamol , Eur. J. Pharm. 
Biopharm. , 63 ( 1 ), 19 – 25 . 
5. Leuenberger , H. , and Leu , R. ( 1992 ), Formation of a tablet: A site and bond percolation 
phenomenon , J. Pharm. Sci. , 81 ( 10 ), 976 – 982 . 
6. Khossravi , D. , and Morehead , W. T. ( 1997 ), Consolidation mechanisms of pharmaceutical 
solids: A multi-compression cycle approach, Pharm. Res. , 14 ( 8 ), 1039 – 1045 . 
REFERENCES 1161

1162 TABLET COMPRESSION 
7. Masteau , J. C. , and Thomas , G. ( 1999 ), Modelling to understand porosity and specifi c 
surface area changes during tabletting , Powder Technol. , 101 ( 3 ), 240 – 248 . 
8. Zuurman , K. , Van der Voort Maarschalk , K. , and Bolhuis , G. K. ( 1999 ), Effect of magnesium 
stearate on bonding and porosity expansion of tablets produced from materials with 
different consolidation properties , Int. J. Pharm. , 179 ( 1 ), 107 – 115 . 
9. Heckle , R. W. ( 1961 ), An analysis of powder compaction phenomena , Trans. Metall. Soc. 
AIME , 221 , 1001 – 1008 . 
10. Heckle , R. W. ( 1961 ), Density - pressure relationship in powder compaction , Trans. Metall. 
Soc. AIME , 221 , 671 – 675 . 
11. Shivanand , P. , and Sprockel , O. L. ( 1992 ), Compaction behavior of cellulose polymers , 
Powder Technol. , ( 69 ), 177 – 184 . 
12. Cooper , A. R. , and Eaton , L. E. ( 1962 ), Compaction behavior of several ceramic powders , 
J. Am. Ceram. Soc. , 45 ( 3 ), 97 – 101 . 
13. Kawakita , K. , and Ludde , K. H. ( 1970 ), Some considerations on powder compression 
equations , Powder Technol. , 4 , 61 – 68 . 
14. Palmieri , G. F. , Joiris , E. , Bonacucina , G. , Cespi , M. , and Mercuri , A. ( 2005 ), Differences 
between eccentric and rotary tablet machines in the evaluation of powder densifi cation 
behaviour , Int. J. Pharm. , 298 ( 1 ), 164 – 175 . 
15. Williams , R. O. 3rd, and McGinity , J. W. (1989), Compaction properties of microcrystalline 
cellulose and sodium sulfathiazole in combination with talc or magnesium stearate , 
J. Pharm. Sci. , 78 ( 12 ), 1025 – 1034 . 
16. Muller , F. X. , and Augsburger , L. L. ( 1994 ), The role of the displacement - time waveform 
in the determination of Heckel behaviour under dynamic conditions in a compaction 
simulator and a fully - instrumented rotary tablet machine , J. Pharm. Pharmacol. , 46 ( 6 ), 
468 – 475 . 
17. Amidon , G. E. , and Houghton , M. E. ( 1995 ), The effect of moisture on the mechanical 
and powder fl ow properties of microcrystalline cellulose , Pharm. Res. , 12 ( 6 ), 923 – 929 . 
18. Gupta , A. , Peck , G. E. , Miller , R. W. , and Morris , K. R. ( 2005 ), Infl uence of ambient moisture 
on the compaction behavior of microcrystalline cellulose powder undergoing uni - 
axial compression and roller - compaction: A comparative study using near - infrared 
spectroscopy , J. Pharm. Sci. , 94 ( 10 ), 2301 – 2313 . 
19. Gustafsson , C. , Lennholm , H. , Iversen , T. , and Nystrom , C. ( 2003 ), Evaluation of surface 
and bulk characteristics of cellulose I powders in relation to compaction behavior and 
tablet properties , Drug. Dev. Ind. Pharm. , 29 ( 10 ), 1095 – 1107 . 
20. Cunningham , J. C. , Sinka , I. C. , and Zavaliangos , A. ( 2004 ), Analysis of tablet compaction. 
I. Characterization of mechanical behavior of powder and powder/tooling friction , 
J. Pharm. Sci. , 93 ( 8 ), 2022 – 2039 . 
21. Picker , K. M. ( 2004 ), The 3D model: Explaining densifi cation and deformation mechanisms 
by using 3D parameter plots , Drug. Dev. Ind. Pharm. , 30 ( 4 ), 413 – 425 . 
22. Ruegger , C. E. , and Celik , M. ( 2000 ), The effect of compression and decompression speed 
on the mechanical strength of compacts , Pharm. Dev. Technol. , 5 ( 4 ), 485 – 494 . 
23. Barra , J. , Falson - Rieg , F. , and Doelker , E. ( 1999 ), Infl uence of the organization of binary 
mixes on their compactibility , Pharm. Res. , 16 ( 9 ), 1449 – 1455 . 
24. Press Tooling Manual ( 2003 ), Thomas Engineering Inc. Hoffman Estates, IL , USA . pp. 
1 – 38 . 
25. Tableting Specifi cation Manual , 7th ed., American Pharmacists Association, p. 130. 
26. International Organization for Standardization (ISO) , 18084: 2005 , 1st ed, ISO , Geneva , 
pp. 1 – 13 . 

27. Train , D. ( 1956 ), An investigation into the compaction of powders , J. Pharm. Pharmacol. , 
8 ( 10 ), 745 – 761 . 
28. Burlinson , H. ( 1968 ), Tablets and Tabletting , Heinemann , London . 
29. Long , W. M. ( 1960 ), Radial pressures in powder compaction, Powder Metall. , 6 , 73 – 86 . 
30. Ritter , A. , and Sucker , H. B. ( 1980 ), Studies of variables that effect tablet capping , Pharm. 
Tech. , ( 3 ), 57 – 65 , 128. 
31. Hiestand , E. N. , Bane , J. M. , Jr ., and Strzelinski , E. P. ( 1971 ), Impact test for hardness of 
compressed powder compacts , J. Pharm. Sci. , 60 ( 5 ), 758 – 763 . 
32. Joe Au , Y. H. , Eissa , S. , and Jones , B. E. ( 2004 ), Receiver operating characteristic analysis 
for the selection of threshold values for detection of capping in powder compression , 
Ultrasonics , 42 ( 1 – 9 ), 149 – 153 . 
33. Rodriguez , L. , Cini , M. , Cavallari , N. , Passerini , N. , Saettone , M. F. , Monti , D. , and Caputo , 
O. ( 1995 ), Ultrasound - assisted compaction of pharmaceutical materials , Farm Vestn. , ( 46 ), 
241 – 242 . 
34. Armstrong , N. A. , and Palfrey , L. P. ( 1989 ), The effect of machine speed on the consolidation 
of four directly compressible tablet diluents , J. Pharm. Pharmacol. , 41 ( 3 ), 149 – 151 . 
35. Zografi , G. , and Mattocks , A. M. ( 1963 ), Adsorption of certifi ed dyes by starch , J. Pharm. 
Sci. , 52 (Nov.), 1103 – 1105 . 
REFERENCES 1163


1165 
6.7 
EFFECTS OF GRINDING IN 
PHARMACEUTICAL TABLET 
PRODUCTION 
Gavin Andrews , David Jones , Hui Zhai , Osama Abu Diak , and 
Gavin Walker 
Queen ’ s University Belfast, Belfast, Northern Ireland 
Contents 
6.7.1 Introduction 
6.7.2 Milling Equipment 
6.7.2.1 Ball Mill 
6.7.2.2 Fluid Energy Mill 
6.7.2.3 Hammer Mill 
6.7.2.4 Cutting Mill 
6.7.3 Powder Characterization Techniques 
6.7.3.1 Powder Sampling 
6.7.3.2 Particle Density and Voidage 
6.7.3.3 Particle Surface Area 
6.7.3.4 Particle Shape 
6.7.4 Effect of Particle Size Reduction on Tableting Processes 
6.7.4.1 Wet Granulation Processes 
6.7.4.2 Mixing Processes 
6.7.4.3 Flowability of Pharmaceutical Powders 
6.7.4.4 Compression Processes 
References 
6.7.1 INTRODUCTION 
The importance of size reduction in relation to pharmaceutical active agents and 
excipients is well known, and the aim of this chapter is to identify methods for particle 
size reduction, discuss how particle size and shape are characterized, and 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

1166 EFFECTS OF GRINDING IN PHARMACEUTICAL TABLET PRODUCTION 
recognize the importance of controlling particle characteristics to ensure the success 
of pharmaceutical powder processing and the manufacture of elegant pharmaceutical 
products. An initial overview of the implications of size reduction within 
pharmaceutics and the importance of comminution in relation to variability of 
active pharmaceutical ingredient (API) surface area, effi cacy, and ultimately dosing 
regimen required to maintain optimum therapeutic effects will be addressed. This 
will encompass examples from a diverse range of dosage forms, including oral, 
parenteral, and topical systems. The effects of particle size on the essential characteristics 
of powders intended for compression (tablets, capsules) such as fl uidity and 
compressibility will be addressed. The need for uniformity of size and the effects 
of particle size distribution on the homogeneity of mixing/blending and in essence 
on the uniformity of APIs within the fi nal manufactured dosage form will be 
highlighted. 
6.7.2 MILLING EQUIPMENT 
There are many factors that must be taken into consideration in choosing milling 
equipment. Some of these factors are related to required product specifi cations such 
as particle size distribution, but additionally, physical and chemical properties of 
the material such as particle shape and moisture content must also be taken into 
consideration. Furthermore, other factors that are related to production requirements 
(mill capacity and the required production rate) must be carefully balanced 
to ensure the correct choice of milling equipment. 
6.7.2.1 Ball Mill 
A ball mill consists of a hollow cylinder mounted such that it can be rotated on its 
horizontal longitudinal axis (Figure 1 ). The length of the ball mill is slightly greater 
than its diameter. A ball mill reduces particle size by subjecting particles to impact 
and attrition forces generated by moving steel balls or pebbles (grinding medium) 
that typically occupy 30 – 50% of the total volume of the mill. It is common for a 
ball mill to contain balls of different diameters that aid size reduction. Generally, 
larger diameter balls have a higher tendency to act upon coarse feed materials 
FIGURE 1 Ball mill in operation showing correct cascade action. 
(a) (b) (c)

MILLING EQUIPMENT 1167 
whereas smaller diameter balls facilitate the formation of fi ne product by reducing 
void spaces between the balls. 
The most important factors governing the performance of the mill and the 
achievement of the desired particle size are as follows: 
1. Amount of material required for subsequent testing (sample volume) 
2. Speed of rotation of ball mill 
A high volume of powder feed produces a cushioning effect whereas small 
sample volumes cause a loss of effi ciency and abrasive wear of the mill parts. The 
amount of material to be milled in a ball mill may be expressed as a material - to - void 
ratio (ratio of the volume of material to that of the void in the ball charge). As the 
amount of material is increased, the effi ciency of a ball mill is increased until the 
void space in the bulk volume of ball charge is fi lled; then, the effi ciency of milling 
is decreased by further addition of material. 
Rotational speed is the most signifi cant factor controlling the particle size speci- 
fi cation. The optimum speed of rotation is dependent on mill diameter. At low 
angular velocities the balls move with the drum until the force due to gravity 
exceeds the frictional force of the bed on the drum, and the balls then slide back 
to the base of the drum. This sequence is repeated, producing very little relative 
movement of balls so that size reduction is minimal. At high angular velocities the 
balls are thrown out onto the mill wall by centrifugal force and no size reduction 
occurs. At about two - thirds of the critical angular velocity where centrifuging occurs, 
a cascading action is produced. Balls are lifted on the rising side of the drum until 
their dynamic angle of repose is exceeded. At this point they fall or roll back to the 
base of the drum in a cascade across the diameter of the mill. By this means, the 
maximum size reduction occurs by impact of the particles with the balls and by 
attrition. 
The critical speed of a ball mill is the speed at which the balls just begin to centrifuge 
with the mill. Thus, at the critical speed, the centrifugal force is equal to the 
weight of the ball. At and above the critical speed, no signifi cant size reduction 
occurs. The critical speed n c is given by the equation 
n 
D c = 76 6 . 
where D is the diameter of the mill. 
A larger mill reaches its critical speed at a slower revolution rate than a smaller 
mill. Ball mills are operated at from 60 to 85% of the critical speed. Over this range, 
the output increases with the speed; however, the lower speeds are for fi ner grinding. 
An empiric rule for the optimum speed of a ball mill is 
n D = . 57 40log 
where n is the speed in revolutions per minute and D is the inside diameter of the 
mill in feet. 

1168 EFFECTS OF GRINDING IN PHARMACEUTICAL TABLET PRODUCTION 
In practice, the calculated speed should be used initially in the process and modi- 
fi ed as required. 
The use of a ball mill is advantageous in that it may be used for both wet and 
dry milling and additionally can be successfully employed in batch and continuous 
operation. Also, the installation, operation, and labor costs involved in ball milling 
are extremely low in comparison to other techniques, which makes this technique 
economically favorable. 
6.7.2.2 Fluid Energy Mill 
Fluid energy milling acts by particle impaction and attrition that are generated by 
a fl uid, usually air (Figure 2 ). Fluid energy mills can reduce the particle size to 
approximately 1 – 20 . m. A fl uid energy mill consists of a hollow toroid that has a 
diameter of 20 – 200 . m, depending on the height of the loop, which may be up to 
2 m. Fluid is injected as a high - pressure jet through nozzles at the bottom of the 
loop with the high - velocity air, giving rise to zones of turbulence into which solid 
particles are fed. The high kinetic energy of the air causes the particles to impact 
with other particles with suffi cient momentum for fracture to occur. Turbulence 
ensures that the high levels of particle – particle collision produce substantial size 
reduction by impact and attrition. 
The design of fl uid energy mills provides an internal classifi cation system according 
to their particle size in which the fi ner and lighter particles are discharged and 
the heavier, oversized particles, under the effect of centrifugal force, are retained 
until reduced to a signifi cantly smaller size. 
FIGURE 2 Fluid energy mill. 
Centrifuging action 
throws coarser 
particles outward 
Classifier removes 
fine particles 
and fluid
Solids inlet 
Fluid inlet jets 
Zone of 
turbulence

MILLING EQUIPMENT 1169 
6.7.2.3 Hammer Mill 
The main mechanism of size reduction produced by a hammer mill is by impaction 
that is generated from a series of four or more hammers hinged on a central shaft 
and enclosed within a rigid metal case (Figure 3 ). During milling the hammers swing 
out radially from the rotating central shaft. The angular velocity of the hammers 
produce strain rates up to 80 s . 1 , which are so high that most particles undergo brittle 
fracture. As size reduction continues, the inertia of particles hitting the hammers 
reduces markedly and subsequent fracture is less probable, so that hammer mills 
tend to produce powders with narrow particle size distributions. Particle retention 
within the mill is achieved using a screen, which allows only suffi ciently milled particles 
(defi ned particle size) to pass through. Particles passing through a given mesh 
can be much fi ner than the mesh apertures, as particles are carried around the mill 
by the hammers and approach the mesh tangentially. For this reason, square, rectangular, 
or herringbone slots are often used. According to the purpose of the operation, 
the hammers may be square faced or tapered to a cutting edge or have a 
stepped form. 
The particle size achieved may be controlled variation in the speed of the hammers 
and additionally by careful selection of the size and type of screen. During the 
operation of a hammer mill the speed of rotation is critical such that below a critical 
impact speed the rotor turns so slowly that a blending action rather than milling is 
obtained. Such operating conditions result in signifi cant rises in temperature. Moreover, 
at very high speeds, there is the probability of insuffi cient time between successive 
passes of the hammers for a signifi cant mass of material to fall from the 
grinding zone. 
The hammer mill is particularly useful in achieving particles in the approximate 
size range of 20 – 40 . m and additionally in producing a particle size distribution that 
is extremely narrow. The equipment offers ease of use and high levels of fl exibilty 
(speed and screen may be rapidly changed allowing rapid variation in achievable 
particle size), is easy to clean, and can be operated as a closed system, thus avoiding 
operator exposure to potent dusts and potential explosion hazards. 
FIGURE 3 Hammer mill. 
Hammers 
Feed 
Screen 
Product

1170 EFFECTS OF GRINDING IN PHARMACEUTICAL TABLET PRODUCTION 
6.7.2.4 Cutting Mill 
Particle size reduction using a cutting mill involves successive cutting or shearing a 
sample using a series of knives attached to a horizontal rotor (Figure 4 ). This rotary 
motion pushes the sample against a series of stationary knives that are attached to 
the mill casing. Size reduction occurs by fracture of particles between the two sets 
of knives, which have a clearance of approximately a few millimetres. As with a 
hammer mill a screen is fi tted at the base of the mill casing and acts to retain material 
until a suffi cient degree of size reduction has occurred. 
6.7.3 POWDER CHARACTERIZATION TECHNIQUES 
6.7.3.1 Powder Sampling 
Powdered materials are used in a wide range of industries, no more so than in the 
pharmaceutical industry wherein powders are used for the manufacture of a wide 
range of dosage forms, the two most common being tablets and hard gelatin capsules. 
Orally administered solid dosage forms are the preferred and most patient 
convenient, primarily because of the ease of administration and the convenience of 
handling. Pharmaceutically, orally administered solid dosage forms are generally 
more favorable because of increased stability in comparison to their liquid counterparts 
(suspensions, syrups) and the increased control they offer in manipulating 
drug dissolution in vivo to suit end - use requirements. Solid dosage forms administered 
via the oral route are an intricate blend of pharmaceutical excipients (diluents, 
FIGURE 4 Cutter mill. 
Screen Product 
Feed 
Stationary knives 
Rotating knives

binders, disintegrants, glidants, lubricants, and fl avors) and APIs. In order to successfully 
manufacture acceptable pharmaceutical products, these materials must 
be adequately mixed and/or granulated to ensure that the resultant agglomerates 
possess the required fl uidity and compressibility and, in addition, avoid demixing 
during postgranulation processes. Moreover, the fi nal characteristics of tablets or 
capsules such as drug dissolution rate, disintegration time, porosity, friability and 
hardness are signifi cantly infl uenced by the properties of the powder blends used 
during their manufacture. 
During product manufacture large volumes of powder blends are fed through 
production equipment/processes, and it is essential to be able to accurately 
determine, defi ne, and control powder properties to ensure reproducible manufacture 
and product performance. Therefore the characterization of the physicochemical 
properties of powder blends is extremely important. It is well accepted 
that there are inherent diffi culties in characterizing the entire mass of a bulk 
powder blend or process stream, so it is essential to remove and analyze discrete 
samples. 
Sampling is a useful technique that allows an appropriate aliquot to be withdrawn 
from the bulk so as to collect a manageable amount of powder which is representative 
of the batch [3] , in other words, every particle should have an equal chance of 
being selected [4] . However, there are many circumstances that may result in the 
selection of nonrepresentative samples and hence the defi nition of powder characteristics 
that are not a true estimation of the entire bulk powder. Typically, powder 
masses with an extremely wide particle size distribution or diverse physical properties 
are highly likely to be heterogeneous, which may result in high levels of variability 
and samples that do not represent bulk mass. Moreover, powder characteristics 
may change because of the attrition and segregation during transfer that can make 
sampling extremely diffi cult. 
It is well accepted that two types of sampling errors are possible when removing 
small masses of powder from bulk [5] . 
1. Segregation errors, which are due to segregation within the bulk and can be 
minimized by suitable mixing and the use of a large number of incremental 
samples to form a larger test sample. 
2. Statistical errors, which arise because the quantitative distribution in 
samples of a given magnitude is not constant but is subject to random fl uctuations. 
Consequently, it is an example of a sampling error that cannot 
be prevented but can be estimated and indeed reduced by increasing the 
sample size. 
Therefore, sampling procedures are of the greatest importance in order to reduce 
the effect of nonuniform size segregation and nonrandom homogeneity of a system 
to achieve statistically meaningful sampling results. Careful attention and faithful 
observance must be demonstrated and it is extremely important that sampling 
occurs when the powders are in motion [6] and samples are withdrawn from the 
whole stream for equal periods of time, rather than part of the stream for all of the 
time [3] . 
POWDER CHARACTERIZATION TECHNIQUES 1171

1172 EFFECTS OF GRINDING IN PHARMACEUTICAL TABLET PRODUCTION 
TABLE 1 Stationary Bulk Sampling 
Sampling Devices Procedure of Sampling 
Application and 
Characteristics 
Low volume powder 
sampler (Figure 
5 a ) 
In operation the sampler is 
inserted into the product to be 
sampled. At a specifi c sampling 
depth the operator pushes 
down on the T bar, which 
opens the sampling chamber. 
When released the spring - 
loaded T bar will close the 
sampling chamber. 
Used for small quantity of 
sample powders. The 
sampler has a sampling 
chamber volume 
approximately equal to 
2 mL. 
Pneumatic lance 
sampler (Figure 
5 c ) 
A gentle fl ow of air out of the 
nozzle allows the probe to move 
through the powder bed. At the 
site, the air is slowly reversed to 
draw up a sample, which is 
collected against a porous plate 
at the end of the probe [7] . 
Minimizes powder 
disturbance and therefore 
is better than a sample 
thief, but bias still cannot 
be avoided [8] . 
Scoop sampler A single swipe of the scoop 
completely across the powder 
bulk collects the sample. Each 
collection should use opposite 
directions. 
Suitable only for materials 
that are homogeneous 
within the limits set by 
the quantity of material 
taken by the scoop. It 
may be used for non - 
free - fl owing or damp 
materials where 
instrumental methods 
are inappropriate [9] . 
Thief/spear probe 
sampler 
(Figure 5 b ) 
One or more cavities are stamped 
in a hollow cylinder enclosed by 
an outer rotating sleeve. The 
thief is inserted into sample 
with the cavities closed, once 
opened the sample fi lls the hole. 
The cavities are closed and the 
thief is withdrawn. It must be 
ensured that samples are 
withdrawn from different 
locations 
Thief samplers belong to 
two main classes, side 
sampler (has one or more 
cavities along the probe) 
and end sampler (has a 
single cavity at the end of 
the probe), which are the 
most common used for 
stored non - fl owing 
material [10] . 
There are a number of sampling techniques for particle sampling, which can be 
classifi ed in many different ways. Here, particle sampling techniques are divided 
into three parts: stationary bulk sampling (Table 1 and Figure 5 ), fl owing stream 
sampling (Table 2 and Figure 6 ), and subsampling (Table 3 and Figure 7 ). The 
sampling devices, procedures and application overview of the common used techniques 
in corresponding fi elds are shown as follow. 

FIGURE 5 Stationary bulk sampling: ( a ) low - volume powder sampler; ( b ) thief/spear 
probe sampler; ( c ) pneumatic lance sampler [7] . 
(a) 
(b) 
(c) 
Press 
Side sampler 
End sampler 
Airflow 
Porous plate Fluidizing jets 
6.7.3.3 Particle Density and Voidage 
Particle density may be defi ned as the total mass of the particle divided by its total 
volume; however, depending upon the different defi nitions of the total volume (or 
the different ways to measure the particle volume), there are various defi nitions of 
particle density in existence (see Table 4 ). 
In order to get clear understanding of the subtle differences between the defi nitions 
of various particle density types, an illustration can be formed as shown in 
Figure 8 . 
Particle Density Methods Density is defi ned as the ratio of mass to volume, so 
the density determination can be separated into two steps: measurement of mass 
and measurement of volume. Determining the mass of an object is rather straightforward; 
however, it is much more diffi cult to directly determine the volume of a 
solid. The volume of a solid object with a regular geometric shape may be calculated 
POWDER CHARACTERIZATION TECHNIQUES 1173

1174 EFFECTS OF GRINDING IN PHARMACEUTICAL TABLET PRODUCTION 
TABLE 2 Flowing Stream Sampling 
Sampling 
Devices Procedure of Sampling Application and Characteristics 
Auger sampler 
(Figure 6 a ) 
[Line sampler 
for stream] 
A pipe with a slot is placed 
inside the process stream, 
permitting easy capture of 
powder through the process 
stream cross section when 
rotated. Samples are 
subsequently then delivered 
into a separate container by 
gravitational forces. [6] 
While this is often used for 
stream sampling, it is diffi cult 
to collect a representative 
sample when stream is 
heterogeneous [10] . 
Constant - volume 
sampler 
(Figure 6 b ) 
[Point sampler 
for stream] 
Sampling occurs when the stream 
falls down through a pipe and a 
constant - volume container is 
inserted or withdrawn from the 
stream system. 
Designed to extract a constant 
volume of homogeneous 
granular material for 
subsequent chemical analyses 
and is not suitable for 
withdrawing samples for 
physical analyses [11] . 
Diverter sampler 
(Figure 6 c ) 
[Cross - 
sectional 
sampler for 
stream] 
The whole stream is diverted by 
opening a sliding cover or 
pivoting an external fl ap in the 
bottom of a gravity - fl ow chutes 
or pipes or screw conveyors 
[12] . The samples could be 
removed to a low - angle laser 
light-scattering instrument then 
returned to the process stream 
[6] . 
The process could be automated 
and highly suitable method 
for online particle size 
measurement. [7] 
Full stream 
sampler 
(Figure 6 d ) 
[Cross - 
sectional 
sampler for 
stream] 
Samples are withdrawn from 
conveyors, carried out only on 
the return stroke. 
Extremely useful for dusty 
materials provided the trough 
extends the whole length of 
the stream and does not 
overfi ll [6] . 
mathematically; however, in most conditions, the shape of a particle is often irregular, 
especially in powder technology, which makes it extremely diffi cult to measure 
geometrically. Therefore, various methods have been developed to determine the 
volume of particles and powders. The two most in use in both laboratory and industrial 
settings are liquid and gas displacement methods. The different values of particle 
density can also be expressed in a dimensionless form, as “ relative density ” (or 
specifi c gravity), which is the ratio of the density of the particle to the density of 
water. 
The discussion that follows will give an overview of the common methods used 
in particle density measurement. 

FIGURE 6 Flowing stream sampling: ( a ) auger sampler [6] ; ( b ) constant - volume sampler 
[6] ; ( c ) diverter sampler; ( d ) full - steam sampler. 
(a) (b) 
(c) (d) 
Powder flow 
Sample 
Process stream Process stream 
Discharge position Sampling position 
To analyzer 
Stream 
Step 1 
Normal position 
Step 2 
Sampling stroke 
Step 3 
Discharging sample 
Samples 
Stream 
Measurement of Particle Density 
1. Liquid Pycnometry Method There are several British standards that deal 
with liquid pycnometry applied to specifi c materials [18 – 23] . A pycnometer bottle 
is weighted empty (M1), and then full of liquid (M2). Following these two initial 
measurements, two subsequent measurements are made: a sample of powder 
approximately one - third of maximum container volume (M3) and the bottle fi lled 
to capacity containing the sample and water (M4). Great care is required in the fi nal 
step to ensure that the liquid is fully wetted and all the air removed. Variations in 
recorded weight also arise depending on how much liquid escapes when the ground 
glass stopper is inserted in the liquid - fi lled container. It is extremely important that 
the liquid used in this procedure does not solubilize or react with the solid particles. 
Moreover, the solid particles must not absorb the selected fl uid. 
2. Gas Pycnometry Method Principally this method is similar to liquid pycnometry 
in that volume determination is achieved by detecting the pressure or volume 
change associated with the displacement of a gas (rather than liquid) by a solid 
object. Given that this method is largely dependent upon the diffusivity of the gas, 
helium is often used since it has a low molecular weight and a small atomic radius, 
allowing high diffusivity into small pores. Sample volumes are often displayed on a 
POWDER CHARACTERIZATION TECHNIQUES 1175

1176 EFFECTS OF GRINDING IN PHARMACEUTICAL TABLET PRODUCTION 
TABLE 3 Subsampling 
Sampling Devices Procedure of Sampling Application and Characteristics 
Coning and 
quartering 
(Figure 7 a ) 
A cross - shaped cutter is used 
to separate the sample heap 
(which is fi rst fl attened at 
the top) into four equal 
parts. The segments are 
drawn apart and two 
opposite quadrants are 
combined together. This 
procedure is repeated at 
least 4 times until a small 
enough sample has been 
generated. 
The fi rst choice for non - free - 
fl owing powders and 
nonfl owing powders. Prone to 
operator bias as fi ne particles 
remain in the center of the 
cone and should never be used 
with free - fl owing powders [13] . 
Oscillating hopper 
sample divider 
(Figure 7 c ) 
Hopper (paddle) oscillates and 
powder falls into two 
collectors placed under the 
hopper (paddle). 
Used for small quantity of 
samples. Sample size can be 
controlled by monitoring time 
over each collector [7] . 
Revolving sample 
splitter (Figure 
7 f ) 
The revolving feeder 
distributes the sample 
material equally (in time) 
over a number of radial 
chutes, assuming constant 
rotational speed [14] . 
Very easy to perform and several 
versions are available that are 
suitable for free - fl owing 
powders, dusty powders, and 
cohesive powders. Handling 
quantities can vary from 40 L to 
a few grams. 
Riffl e/chute splitter 
(Figure 7 e ) 
The sample is introduced to a 
rectangular area, divided by 
parallel chutes leading to 
two separate receptacles 
[14] . 
Well - accepted method for sample 
reduction that is highly suitable 
for free - fl owing powders. Used 
to produce samples with a 
minimum volume of 5 mL. 
Spinning riffl er 
(Figure 7 d ) 
a steady stream of powder is 
run into a rotating basket of 
containers [8] . 
Useful in subsampling large 
samples [15] . Suitable for free - 
fl owing materials [13] . 
Table sampler 
(Figure 7 b ) 
In a sampling table, powder 
fl ows down from the top of 
an inclined plane, holes and 
prisms splitting the powder. 
The powder that reaches the 
bottom of the plane is the 
sample. 
Used for sample reduction with 
the advantages of low price and 
lack of moving parts. 
digital counter on the testing equipment [24] ; however, such volumes are easily 
calculated using the pressure change and the ideal gas law, PV = nRT . The true 
density of the particle can be measured using this method if the particles have no 
closed pores, while the apparent particle density can be measured if there are any 
closed pores. Additionally, if open pores are fi lled with wax, envelope volumes may 

FIGURE 7 Subsampling: ( a ) coning and quartering; ( b ) table sampler [6] ; ( c ) oscillating 
hopper (paddle) sample divider [6] ; ( d ) spinning riffl er (BSI); ( e ) riffl e/chute splitter (BSI); 
( f ) revolving sample splitter [14] . 
Sample
(b) (a) 
(c) (d) 
(e) (f) 
Repeat 
Discard 
AAA 
Container 1 
Container 1 
Container 2 
POWDER CHARACTERIZATION TECHNIQUES 1177

1178 EFFECTS OF GRINDING IN PHARMACEUTICAL TABLET PRODUCTION 
TABLE 4 Defi nitions of Density Terms 
Density 
Types Density Defi nitions 
Volumes in Defi nition 
Solid 
Material 
Volume 
Closed - 
Pore 
Volume 
Open - 
Pore 
Volume 
Interparticle 
Void 
Volume 
Absolute 
powder 
density 
Mass of powder per unit of 
absolute volume, which is 
defi ned as the solid matter 
after exclusion of all the 
spaces (pores and voids) 
(BSI) 
 	 	 	 
Apparent 
particle 
density 
Mass of particles divided by its 
apparent particle volume, 
which is defi ned as the total 
volume of the particle, 
excluding open pores but 
including closed pores (BSI) 
  	 	 
Apparent 
powder 
density 
Mass of powder divided by its 
apparent powder volume, 
which is defi ned as the total 
volume of solid matter, 
including open pores and 
closed pores and interstices 
(BSI) 
   	 
Bulk 
density 
Mass of the particles divided 
by the volume they occupy, 
which includes the space 
between the particles 
(ASTM) 
    
Effective 
particle 
density 
Mass of a particle divided by 
its volume, including open 
pores and closed pores (BSI) 
   	 
Envelope 
density 
Ratio of the mass of a particle 
to the sum of the volumes of 
the solid in each piece and 
voids within each piece, 
which is, within close - fi tting 
imaginary envelopes, 
completely surrounding each 
piece (ASTM) 
   	 
Skeletal 
density 
Ratio of the mass of discrete 
pieces of solid material to 
the sum of the volumes of 
the solid material in the 
pieces and closed pores 
within the pieces (ASTM) 
  	 	 
Tap 
density 
Apparent powder density 
obtained under stated 
conditions of tapping (BSI) 
    
True 
density 
Mass of a particle divided by 
its volume, excluding open 
pores and closed pores (BSI) 
 	 	 	 
Note: BSI = British Standards Institute [16] , ASTM = American Society for Testing and Material [17] . 
 , included; 	 , excluded. 

Eliminating 
interparticle 
void 
volume 
Bulk volume Envelope/effective 
volume 
Apparent/skeletal 
volume 
True/absolute 
volume 
Porous particles 
in container 
Bulk density Envelope/effective 
density 
Apparent/skeletal 
density 
True/absolute 
density 
Eliminating 
open pores, 
cracks 
volume 
Eliminating 
closed pores 
volume 
FIGURE 8 Various density types. The density value increases from bulk density to true 
density while the volume value decreases from bulk volume to true volume. 
be determined and the difference between envelope and apparent volume can yield 
the open - pore volume, which is sometimes used as a measure of porosity. 
3. Hydrostatic Weighing Method The volume of a solid sample is determined 
by comparing the mass of the sample in air with the mass of sample immersed in a 
liquid with a known density. The volume of sample may be calculated using the 
difference between the two measured mass values divided by the density of the 
liquid. This method can be used to determine the bulk or apparent volume. It is 
extremely important that the suspending liquid does not interact with the powder 
under investigation. 
4. Float – Sink or Suspension Method This method involves placing a solid 
sample into a liquid with known and adjustable density. The density of liquid is 
incrementally adjusted until the sample begins to sink – fl oat (ASTM C729 - 75 [25] ), 
or is suspended at neutral density in the liquid (ASTM C693 - 93 [26] ). At the point 
of equilibrium the density of the sample is equal to the density of the liquid. 
5. Bed Pressure Drop Method This technique is based on making measurements 
of bed pressure drop as a function of gas velocity at two voidages, when gas 
is passed through the bed of powder in the laminar fl ow regime [24] . During measurement 
pressure changes for at least four velocities must be measured. The effective 
particle density . p can be calculated using the equation 
s
s
1
2 
3 
= . 
b1 
b2 
p b2 
p b1 
where s is the gradient of pressure drop with gas velocity, . b is the bulk density, . p 
is the particle effective density. 
6. Sand Displacement Method The sand displacement method is another useful 
way of measuring the envelope density of a particle using fi ne sand as the displacement 
media. Sand is mixed with a known amount of particles, then the density of 
the sample particles can be determined from the difference of the bulk density 
between sand alone and that with samples. 
7. Mercury Porosimetry Method Mercury is a nonwetting liquid that must be 
forced to enter a pore by application of external pressure. Consequently it is an 
extremely useful and convenient liquid for measuring the density of powders and/or 
particles. This method can measure the apparent and true density of one sample by 
POWDER CHARACTERIZATION TECHNIQUES 1179

1180 EFFECTS OF GRINDING IN PHARMACEUTICAL TABLET PRODUCTION 
applying different pressures. At atmospheric pressure, mercury will resist entering 
pores smaller than about 6 . m in diameter, but at pressures of approximately 60,000 
psi (414 MPa) mercury will be forced to enter pores with diameters as small as 
0.003 . m [27] . 
Measurement of Bulk Density Bulk density is very important in determining the 
size of containers used for handling, shipping, and defi ning storage conditions for 
pharmaceutical powders and granules. It is a property that is also pertinent in defi ning 
the size of hoppers and receivers for milling equipment and for sizing blending 
equipment in the scale - up to pilot and to commercial production [28] . The concept 
of bulk density is the mass of particles divided by the bulk volume, which includes 
not only the envelope volume of particles but also the spaces between particles, so 
it should not be confused with particle density [24] . 
The most convenient method to measure bulk density is to fi ll the particles into 
a known volume container (usually cylindrical), level the surface, and weigh the 
particles in the container. The bulk density is calculated by the mass of the particles 
divided by the volume that can be read from the scale of the measuring cylinder. 
In order to minimize experimental errors, the container should be ideally at least 
1 L in volume, and the ratio of length and diameter should be about 2 : 1. Also it is 
recommended to leave the sample for approximately 10 min to achieve an equilibrium 
volume (density) value before making readings. 
Given that the bulk volume associated with the particle mass is a mixture of air 
and solid material, the bulk density value is highly dependent on sample history 
prior to measurement. Calculation of the tapped density can then be achieved by 
tapping the bulk powder a specifi ed number of times (to overcome cohesive forces 
and remove entrapped air) to determine the tapped volume of the powder. The 
tapped and bulk density values can be used to defi ne the fl owability and compressibility 
of a powder using Carr ’ s index and the Hausner ratio. 
6.7.3.4 Particle Surface Area 
Surface area is one of the most important characteristics in particle technology. 
Particles with a different surface area will express different physical properties 
that will subsequently affect many applications and ultimately fi nal dosage form 
properties. 
Similar to particle density, there are various defi nitions relating to particle surface 
area [16] : 
1. Adsorption surface area : the surface area calculated from an adsorption 
method. 
2. BET surface area : the surface area calculated from the Brunauer, Emmett, 
and Teller theory of multilayer adsorption of a gas on a solid surface. 
3. Calculated surface area : the surface area of a powder calculated from its particle 
size distribution. 
4. Effective permeability mass - specifi c surface : the effective volume - specifi c 
surface divided by the effective solid density, determined by permeametry. 

5. Effective permeability volume - specifi c surface : the effective surface area 
divided by the effective solid volume, determined by permeametry. 
6. Permeability surface area : the surface area of a powder calculated from the 
permeability of a powder bed under stated conditions. 
7. Specifi c surface area ( S w ): the surface area of a unit mass of material determined 
under stated conditions, where S w is usually expressed in centimeters 
squared per gram or meters squared per gram and can be used for quality 
control purposes [28] . 
Particle Surface Area Determination Methods From the standard defi nitions of 
particle surface area, it can be seen that various determination methods are used 
for surface area measurement, such as adsorption (including Langmuir ’ s equation 
for monolayer adsorption and the BET equation for multilayer adsorption), particle 
size distribution, and permeability methods. The different methods are rarely in 
agreement because the value obtained depends upon the procedures used and also 
on the assumptions made in the theory relating the surface area to the phenomena 
measured. The most common methods used for measuring particle surface area are 
described below. 
1. Gas Adsorption Method Gas adsorption methods measure the surface area 
of particles/powders through measurement of the amount of gas adsorbed onto the 
sample surface. The methods can measure both external and internal surfaces 
(including open pores in the particles) and can yield physically meaningful average 
particle sizes with nonporous materials [24] . The amount of gas adsorbed depends 
upon the nature of the solid (adsorbent) and the pressure at which adsorption takes 
place. The amount of gas (adsorbate) adsorbed can be found by determining the 
increase in weight of the solid (gravimetric method) or the amount of gas removed 
from the system due to adsorption by application of the gas laws (volumetric 
method [6] ). The adsorption used in this method is physical adsorption, which is a 
relatively weak interaction between samples and gases and therefore can be removed 
by evacuation. 
In this method, a graph of the number of moles of gas adsorbed per gram of 
solid, at constant temperature, against the equilibrium gas pressure is called an 
adsorption isotherm. A point must be chosen on this isotherm corresponding to the 
completion of the adsorbed monolayer in order to calculate S w [29] . 
2. Permeametry Method This method is based on the fact that the fl ow rate of 
a fl uid through a bed of particles depends on the pore space, the pressure drop 
across the bed, the fl uid viscosity, dimensional factors such as the area of the bed, 
and specifi c surface area ( S w ). The determination of permeability can be made either 
under continuous steady - state fl ow (constant fl ow rate) or under variable - fl ow 
(constant - volume) conditions. 
All of the permeability methods are based on the Kozeny – Carman equation, 
which is used to calculate a surface area of a packed powder bed from its permeability. 
The Kozeny – Carman equation is expressed as [16] 
S 
A p 
K Lq k = 
. 
.. . 
.. . 
.
. . 
3 
2 1 
. 
( ) 
POWDER CHARACTERIZATION TECHNIQUES 1181

1182 EFFECTS OF GRINDING IN PHARMACEUTICAL TABLET PRODUCTION 
where S k = effective permeability volume - specifi c surface of powder assuming only 
viscous fl ow occurs in determination (Kozeny – Carman term) 
A = cross - sectional area of bed of powder perpendicular to direction of fl ow 
of air 
. = porosity of bed of powder 
. p = pressure difference across bed of powder 
K = Kozeny constant 
L = linear dimension of bed of powder parallel to direction of fl ow of air 
(commonly known as height of powder bed) 
. = viscosity of air at its temperature at time of determination 
q = rate of fl ow of incompressible fl uid through bed of powder 
The specifi c surface area calculated here only involves the walls of the pores of 
the bed and excludes the pores within the particles. Therefore, the surface area 
measured in this method can be much smaller than the total surface area measured 
by gas adsorption methods [24] . 
3. Particle Size Distribution Method The surface area of particles can be determined 
using particle size and particle shape values. The “ equivalent spherical diameter 
” is used in this technique and many attempts to measure the surface area using 
this method have led to values that are signifi cantly less than the true value (large 
deviations arising from inability to defi ne particle shape due to surface irregularities 
and porosity). Surface area values calculated from particle size distribution methods 
will in effect establish the lower limit of surface area due to the implicit assumptions 
of sphericity or other regular geometric shapes and by ignoring the highly irregular 
nature of real surfaces [30] . 
Besides the three methods introduced above, there are many other methods of 
surface area determination: Any surface - dependent phenomenon can be used for 
such measurement [24] . Some available methods (mercury porosimetry, adsorption 
from solution, adsorption of dyes, chemisorption, density methods, and secondary 
ion mass spectroscopy) are explained in more detail elsewhere [6, 30, 31, 32] . 
6.7.3.5 Particle Shape 
Particle behavior is a function of particle size, density, surface area, and shape. 
These interact in a complex manner to give the total particle behavior pattern [28] . 
The shape of a particle is probably the most diffi cult characteristic to be determined 
because there is such diversity in relation to particle shape. However, particle shape 
is a fundamental factor in powder characterization that will infl uence important 
properties such as bulk density, permeability, fl owability, coatablility, particle 
packing arrangements, attrition, and cohesion [33 – 36] . Consequently it is pertinent 
to the successful manipulation of pharmaceutical powders that an accurate defi nition 
of particle shape is obtained prior to powder processing. 
A number of methods have been proposed for particle shape analysis, including 
shape coeffi cients, shape factors, verbal descriptions, curvature signatures, moment 
invariants, solid shape descriptors, and mathematical functions (Fourier series 

expansions or fractal dimensions); these are beyond the scope of this chapter but 
have been adequately described in other texts [37] . 
In the most simplistic means of defi ning particle shape, measurements may be 
classifi ed as either macroscopic or microscopic methods. Macroscopic methods 
typically determine particle shape using shape coeffi cients or shape factors, which 
are often calculated from characteristic properties of the particle such as volume, 
surface area, and mean particle diameter. Microscopic methods defi ne particle 
texture using fractals or Fourier transforms. Additionally electron microscopy 
and X - ray diffraction analysis have proved useful for shape analysis of fi ne 
particles. 
Particle Shape Measurement 
1. Shape Coeffi cients and Shape Factors There are various types of shape 
factors, the majority based on statistical considerations. In essence this translates to 
the use of shape factors that do refer not to the shape of an individual particle but 
rather to the average shape of all the particles in a mass of powder. However, a 
method developed by Hausner [38] that uses three factors — elongation factor, bulkiness 
factor, and surface factor — may be used to characterize the shape of individual 
particles (Table 5 ). 
2. Determining Particle Shape by Fourier Analysis Fourier transforms have 
been previously used to determine particle shape and the rollability of individual 
particles from the coeffi cients of the resulting series [39] . Moreover, fast Fourier 
transforms have been successfully used to determine coeffi cients and a particle 
“ signature ” by plotting ln An versus ln n , where An is the n th Fourier coeffi cient 
and n is the frequency [29, 40, 41] . In brief, Fourier method consists of fi nding the 
centre of gravity of a particle and its perimeter, from which a polar coordinate 
system is set up. Amplitude spectra of a fi nite Fourier series in closed form are used 
as shape descriptors of each particle [42] . Several research papers have focused on 
the characterization of individual particle shape using Fourier grain analysis or 
morphological analysis [43 – 44] . The method has also been extended to the measurement 
of particle shapes in a blend [45] and to relate particle attrition rate in a milling 
operation to particle shape [46] . 
3. Determining Particle Shape by Electron Microscopy Electron microscopy 
has been used for the examination of fi ne powder dispersions and will provide 
information on particle shape perpendicular to the viewing direction. Standard 
shadowing procedures may be useful in obtaining information on shape in the third 
dimension. Scanning electron microscopy can give direct and valuable information 
on the shape of large particles [47] . 
4. Determining Particle Shape by X - Ray Diffraction Broadening The broadening 
of X - ray diffraction lines is primarily a measure of the departure from single - 
crystal perfection and regularity in a material and can therefore be used to 
characterize particle shape. This is the only method that gives the size of the primary 
crystallites, irrespective of how they are aggregated or sintered, and is of great value 
for determining the properties of fi ne powders [48, 49] . 
5. Other Methods for Particle Shape Determination Gotoh and Finney [50] 
proposed a mathematical method for expressing a single, three - dimensional body 
POWDER CHARACTERIZATION TECHNIQUES 1183

1184 EFFECTS OF GRINDING IN PHARMACEUTICAL TABLET PRODUCTION 
TABLE 5 Shape Coeffi cients and Shape Factors 
Coeffi cients and Factors Symbols Defi nitions and Equations 
Volume shape coeffi cient . v 
.v = V
d3 
where V = average particle volume 
d = mean particle diameter 
Surface shape coeffi cient . s 
.s = S 
d2 
where S = average particle surface 
d = mean particle diameter 
Volume – surface shape 
coeffi cient 
. vs 
. .
. vs 
s
v 
= 
where . v = volume shape coeffi cient 
. s = surface shape coeffi cient 
Shape factor . 0 
. . o m n = v 
where . o = shape factor for equidimensional 
particle and thus represents part of . v 
which is due to geometric shape only 
. v = volume shape coeffi cient 
m = fl akiness ratio, or breadth/thickness 
n = elongation ratio, or length/breadth 
Sphericity shape factor . w Sphericity = (surface area of sphere having same 
volume as particle) / (surface area of 
actual particle) 
Circularity shape factor Circularity = (perimeter of particle outline) 2 / 
4 . (cross - sectional or projection 
area of particle outline) 
Source: From refs. 6 and 42 . 
by sectioning it as an equivalent ellipsoid with the same volume, surface area, and 
average projected area as the original body. Moreover, wedge - shaped photodetectors 
to measure forward light - scattering intensity have also been explored for determination 
of crystal shape [51] . More recently a technique referred to as time of 
transition (TOT) that was fi rst introduced in 1988 has also been used for the analysis 
of particle size and shape [52, 53] . 
6.7.4 EFFECT OF PARTICLE SIZE REDUCTION ON 
TABLETING PROCESSES 
Particle size plays a critical role in the effi cacy of a drug product. It can impact not 
only bioavailability but also the effi ciency and success of production process and 
ultimately the properties of the fi nal dosage form. 

6.7.4.1 Wet Granulation Processes 
The particle size of an active pharmaceutical ingredient can have signifi cant effect 
on the processing behavior of a formulation, such as granule growth during wet 
granulation and hence the resulting granule characteristics. The particle size of the 
starting material can affect the strength and deformability of moist granules and 
hence their behavior during the wet granulation process. 
The effect of particle size on granule growth is a function of several interacting 
factors, the balance of which largely depends on the nature of the material and the 
experimental conditions. Differences in granule structure and porosity, resulting 
from changes in starting material particle size, can also affect other characteristics 
(e.g., compressibility) of the granulation. 
Badawy et al. [57] studied the effect of DPC 963 (a nonnucleoside reverse transcriptase 
inhibitor) particle size on the granule growth, porosity, and compressibility 
of granules manufactured by a high - shear wet granulation process. It was found that 
DPC 963 granule growth in the high - shear granulator and the resulting granule 
compressibility and porosity were sensitive to relatively small changes in drug substance 
particle size. Decreasing the particle size resulted in more pronounced 
granule growth and enhanced the porosity and compressibility of the granulation. 
Higher pore volume for the granulation manufactured using the active ingredient 
with a smaller particle size may be the reason for its higher compressibility. The 
high granulation porosity resulted in an increased fragmentation propensity and 
volume reduction behavior of the granulation that led to increased compressibility. 
The more porous granulation has higher tendency to densify upon application of 
the compression force, resulting in closer packing of the particles. 
6.7.4.2 Mixing Processes 
Mixing may be defi ned as a unit operation that aims to treat two or more components, 
initially in an unmixed or partially mixed state, so that each unit of the components 
lies as nearly as possible in contact with a unit of each of the other 
components [2] . Whenever a product contains more than one component, mixing 
will be required in the manufacturing process in order to ensure an even distribution 
of the active component(s). 
It is well accepted that mixing solid ingredients is usually more effi cient and 
uniform if the active ingredient and excipients are approximately the same size, 
which ultimately provides a greater uniformity of dose [1] . Particle size and particle 
size distribution are important in the powder - mixing process since they largely 
determine the magnitude of forces, gravitational and inertial, that can cause interparticulate 
movement relative to surface forces, which resist such motion. As a 
consequence of high interparticulate forces, as compared with the gravitational 
forces, powders of less than 100 . m mean particle diameter sizes are not free 
fl owing. Powders that have high cohesive forces due to interaction of their surfaces 
can be expected to be more resistant to intimate mixing than those whose surfaces 
do not interact strongly [2] . 
In moving from one location to another, relative to neighboring particles, a particle 
must surmount a certain potential energy barrier that arises from forces resisting 
movement. This effect is a function of both particle size and shape and is most 
EFFECT OF PARTICLE SIZE REDUCTION ON TABLETING PROCESSES 1185

1186 EFFECTS OF GRINDING IN PHARMACEUTICAL TABLET PRODUCTION 
pronounced when high packing densities occur. Ideal mixing may be achieved when 
all the particles of the powder mix have similar size, shape, and density characteristics 
whereas segregation (demixing) may occur when powder blends are not composed 
of monosized near - spherical particles but contain particles that differ in size, 
shape, and density. Segregation is more likely to occur if the powder bed is subjected 
to vibration. 
The main reason for segregation in powder blends is the difference in the particle 
size of the components of the particles contained within the blend. Due to the high 
diffusivity of small particles, such materials move through the voids between larger 
particles and so migrate to the lower regions of the powder mix. Moreover, during 
mixing operations, extremely fi ne particles have a high tendency to be forced 
upward by turbulent air currents as the powder blend tumbles and subsequently 
become isolated from the mixing process through continuous suspension above the 
blend. When mixing is stopped, these particles will sediment and form a layer on 
top of the coarser particles. 
It is important to control the particle size distribution of pharmaceutical granules 
or powder blends because a wide size distribution can lead to a situation with a high 
probability of segregation. If this occurs within the hoppers of tablet machines, 
nonuniform products may be manufactured due principally to large weight variations. 
Tablet dies are fi lled by volume rather than weight, and consequently, the 
establishment of different regions within a hopper containing granules of different 
sizes (and hence bulk density) will contain a different mass of granules. This will 
lead to an unacceptable distribution of the active pharmaceutical content within the 
batch of fi nished product, even though the drug is evenly distributed by weight 
throughout the granules. 
6.7.4.3 Flowability of Pharmaceutical Powders 
Due to the relatively small particle size, irregular shape, and unique surface characteristics, 
many pharmaceutical powders have a high tendency to be extremely 
cohesive. This high level of cohesion results in “ sticky ” powders that have poor 
fl owability, commonly resulting in large mass variability within the fi nal product 
owing to unpredictable and variable fi lling of tablet dies. 
Powders with different particle sizes have different fl ow and packing properties, 
which signifi cantly alter the volume of powder expelled from manufacturing equipment 
during, for example, encapsulation or tablet compression. In order to avoid 
such problems, the particle sizes of the active pharmaceutical ingredient and other 
powder excipients should be defi ned and controlled during formulation so that 
problems during production are avoided. Most notably, powder fl owability is of 
critical importance in the successful production of acceptable pharmaceutical dosage 
forms. High levels of fl owability within pharmaceutical powders is not just important 
in the fi nal stages of manufacture but is essential for many industrial processes, 
particularly mass transport. 
Poor or uneven powder fl ow can result in excess entrapped air within powders, 
which may induce capping or lamination in specifi c high - speed tableting equipment. 
Moreover, uneven powder fl ow that is a direct result of the presence of excess fi nes 
within a powder blend will also promote increased particle – die wall friction, lubrica

tion problems, and very importantly increased dust contamination hazards to operating 
personnel. 
Although particle size is a signifi cant factor controlling the fl owability of pharmaceutical 
powders or granules, other factors must be considered. The presence of 
molecular forces between particle/granule surfaces increases the probability for 
cohesion and adhesion between solid particles. Cohesion may be defi ned as the 
attractive forces between like surfaces, such as component particles of a bulk solid, 
whereas adhesion may be defi ned as the attractive force between two unlike surfaces, 
for example, between a particle and a tablet punch. It is extremely important 
to appreciate that cohesive forces acting between particles in a powder bed are 
attributed mainly to short - range nonspecifi c van der Waals forces that are signifi - 
cantly altered as particle size and relative humidity change. 
Cohesion and adhesion are phenomena that occur at the surface of a solid and 
hence particles with an extremely large surface area will have greater attractive 
forces than those with a smaller surface area. Consequently particle surface area 
will have a dramatic effect on the fl owability of pharmaceutical powders. Typically, 
fi ne particles with very high surface - to - mass ratios will be more cohesive than larger 
particles, which are infl uenced more by gravitational forces. Particles larger than 
250 . m are usually relatively free fl owing, but as the size falls below 100 . m, powders 
become cohesive and fl ow problems are likely to occur. Powders having a particle 
size less than 10 . m are usually extremely cohesive and resist fl ow under gravity. 
Although it has been previously stated that particles with similar particle sizes 
are desirable for pharmaceutical processes, a bulk powder mass with a narrow particle 
size distribution accompanied with dissimilar particle shapes can produce a 
bulk mass with inherently different fl ow properties, owing principally to differences 
in interparticle contact area. 
6.7.4.4 Compression Processes 
In general, the strength of a compressed powder depends on the inherent ability of 
the powder to reduce in volume during compression and the amount of interparticulate 
attraction in the fi nal compact. The decrease in compact volume with 
increasing compression load is attributed normally to particle rearrangement, elastic 
deformation, plastic deformation, and particle fragmentation. Pharmaceutical materials 
normally consolidate by more than one of these mechanisms [58, 59] . Unmodi- 
fi ed paracetamol crystals exhibit poor compressibility during compaction, resulting 
in weak and unacceptable tablets with a high tendency to cap [60] . Moreover the 
incidence of capping and lamination during production, following ejection of tablets 
from the die, depended on the plastic and elastic behaviors of the excipients used 
[61] . It has been suggested that materials undergoing plastic deformation, in contrast 
to elastic deformation, display enhanced bond formation and produce strong 
tablets. 
The effect of particle size on the compression properties of paracetamol oral 
dosage forms has been previously reported [62] . Heckel analyses plots indicated 
that the predominant mechanism of compaction of paracetamol was fragmentation 
with larger particle fractions experiencing more fragmentation than the smaller 
particles. Furthermore, Heckel analysis also indicated that, for a given applied 
EFFECT OF PARTICLE SIZE REDUCTION ON TABLETING PROCESSES 1187

1188 EFFECTS OF GRINDING IN PHARMACEUTICAL TABLET PRODUCTION 
pressure, the larger particles of paracetamol produced denser compacts than the 
smaller particles. The results of elastic – plastic energy ratios indicated that the 
majority of energy involved during compaction of paracetamol was utilized as 
elastic energy. This suggested a massive elastic deformation of paracetamol particles 
under pressure, resulting in weak and capped tablets. It was found that larger particles 
exhibited less elastic recovery and elastic energy compared to smaller particles. 
This was attributed to increased fragmentation of larger particles, resulting in 
increased bonding between particles due to the formation of more new, fresh, and 
clean particle surfaces. 
REFERENCES 
1. Lachman , L. , Lieberman , H. A. , and Kanig , J. L. ( 1986 ), The Theory and Practice of 
Industrial Pharmacy Textbook , 3rd ed., Leo & Febiger , Philadelphia . 
2. Aulton , M. E. ( 2002 ), Pharmaceutics: The Science of Dosage Form Design Textbook , 
2nd ed., Churchill Livingstone , London . 
3. Svarovsky , L. ( 1990 ), Characterization of powders , in Rhodes , M. J. , Ed., Principles of 
Powder Technology , Wiley , Chichester , pp. 35 – 69 . 
4. Hawkins , A. E. ( 1990 ), Characterizing the single particle , in Rhodes , M. J. , Ed., Principles 
of Powder Technology , Wiley , Chichester , pp. 9 – 34 . 
5. Sommer , K. ( 1981 ), Sampling error on particle analysis , Aufbereit Tech. , 22 ( 2 ), 
96 – 105 . 
6. Allen , T. ( 1997 ), Particle Size Measurement , 5th ed., Vol. 1, Chapman & Hall , 
London , pp. 1 – 62 . 
7. Venable , H. J. , and Wells , J. I. ( 2002 ) Powder sampling , Drug Dev. Ind. Pharm ., 28 ( 2 ), 
107 – 117 . 
8. Kaye , B. H. ( 1997 ), Powder Mixing , Chapman & Hall , London , pp. 77 – 95 . 
9. British Standard, BS 3406 - 1 ( 1986 ), Methods for Determination of Particle Size Distribution 
— Part 1: Guide to Powder Sampling , British Standard Institution , London . 
10. Bicking , C. A. ( 1964 ), Sampling , in Standen , A. Ed. Kirk – Othmer Encyclopedia of 
Chemical Technology , 2nd ed., Interscience Publishers , New York , pp. 744 – 762 . 
11. Hulley , B. J. ( 1970 ), Sampling and Sample Conditioning in On-line Fertilizer Analysis , 
Chem. Eng ., 77 , 410 – 413 . 
12. Clarke , J. R. P. , and Carr – Brion , K. G. ( 1996 ), Sampling systems for process analysers 
2nd ed., The Bath Press , Avon, pp. 148 – 180. 
13. Allen , T. ( 1964 ), Sampling and size analysis , Silic. Ind . 29 ( 12 ), 509 – 515 . 
14. Petersen , L. ( 2004 ), Representative mass reduction in sampling — A critical survey of 
techniques and hardware , Chemometr. Intell. Lab. Syst ., 74 , 95 – 114 . 
15. Crosby , N. T. , and Patel , I. ( 1995 ), General Principles of Good Sampling Practice , Royal 
Society of Chemistry , Cambridge . 
16. British Standard BS 2955 ( 1993 ), Glossary of Terms Relating to Particle Technology , 
British Standards Institution , London . 
17. ASTM ( 1994 ), Compilation of ASTM Standard Defi nitions , 8th ed., American Society 
for Testing and Materials , Philadelphia, PA . 
18. British Standard BS 812 ( 1995 ), Testing Aggregates, Part 2: Methods of Determination 
of Density , British Standards Institution , London . 

19. British Standard 1016 ( 1980 ), Methods for the Analysis and Testing of Coal and Coke, 
Part 13: Test Special to Coke , British Standards Institution , London . 
20. British Standard BS 1377 ( 1975 ), Methods of Tests for Soils for Civil Engineering Purposes 
, British Standards Institution , London . 
21. British Standard BS 3483 ( 1974 ), Methods for Testing Pigments for Paints, Part B8: 
Determination of Density Relative to Water at 4 ° C , British Standards Institution , 
London . 
22. British Standard BS 4550 ( 1987 ), Methods for Testing Cement, Part 3.2: Density Test , 
British Standards Institution , London . 
23. British Standard BS 7755 ( 1998 ), Soil Quality, Part 5: Physical Method, Section 5.3: 
Determination of Particle Density , British Standards Institution , London . 
24. Svarovsky , L. ( 1987 ), Powder Testing Guide, Methods of Measuring the Physical Properties 
of Bulk Powders , Published on behalf of the British Materials Handing Board by 
Elsevier Applied Science , London , pp. 3 – 33 , 79 – 95 . 
25. ASTM C729 - 75 e1 ( 1995 ), Standard Test Method for Density of Glass by the Sink - Float 
Comparator , American Society for Testing and Materials , Philadelphia, PA . 
26. ASTM C693 - 93 ( 1998 ), Standard Test Method for Density of Plastics by the 
Density - Gradient Technique , American Society for Testing and Materials , Philadelphia, 
PA . 
27. Webb , P. ( 2001 ), Volume and Density Determinations for Particle Technologists , 
Micromeritics Instrument Corp , Georgia . 
28. Lieberman , H. , and Lachman , L. ( 1981 ), Pharmaceutical Dosage Forms Tablets , Vol. 2, 
Marcel Dekker , New York , pp. 112 – 150 , 202 – 222 . 
29. Beddow , J. K. , and Meloy , T. ( 1980 ), Testing and Characterization of Powders and Fine 
Particles , Heyden & Son , London , pp. 63 – 64 . 
30. Lowell , S. , and Shields , J. E. ( 1984 ), Powder Surface Area and Porosity , 2nd ed., 
Chapman & Hall , London . 
31. Beddow , J. K. ( 2000 ), Particle Characterization in Technology , Vol. I, CRC Press , Boca 
Raton, FL , 3 – 20 . 
32. Brunauer , S. , Deming , L. S. , Deming , W. S. , and Teller , E. ( 1940 ), Adsorption of gases 
in multimolecular layers , J. Am. Chem. Soc ., 62 , 1723 – 1732 . 
33. Fonner , D. E. , Banker , G. S. , and Swarbrick , J. ( 1966 ), Micromeritics of granular pharmaceutical 
solids. 1. Physical properties of particles prepared by 5 different granulation 
methods . J. Pharm. Sci ., 55 , 181 . 
34. Ridgway , K. , and Rupp , R. ( 1969 ), Effect of particle shape on particle properties , J. 
Pharm. Pharmacol ., 21 , 30 – 39 . 
35. Shotton , E. , and Obiorah , B. A. ( 1975 ), Effect of physical properties on compression 
characteristics , J. Pharm. Sci ., 64 ( 7 ), 1213 – 1216 . 
36. Heyd , A. , and Dhabbar , D. , Particle shape effect on caking of coarse granulated antacid 
suspensions , Drug Cosmet. Ind. 125 , 42 – 45 . 
37. Hawkins , A. E. ( 1993 ), The Shape of Powder - Particle Outline , Wiley , New York . 
38. Hausner , H. H. ( 1967 ), Characterization of the Powder Particle Shape in Particle Size 
Analysis , Society for Analytical Chemistry , London . 
39. Schwarcz , H. P. , and Shane , K. C. ( 1969 ), Measurement of particle shape by Fourier 
analysis , Sedimentology , 13 ( 3 – 4 ), 213 – 231 . 
40. Meloy , T. P. ( 1969 ), Screening , AIME , Washington, DC . 
41. Meloy , T. P. ( 1977 ), Fast Fourier transforms applied to shape analysis of particle silhouettes 
to obtain morphological data , Powder Technology , 17 , 27 – 35 . 
REFERENCES 1189

1190 EFFECTS OF GRINDING IN PHARMACEUTICAL TABLET PRODUCTION 
42. Ehrlich , R. , and Full , W. E. ( 1984 ), Fourier shape analysis — a multivariate pattern recognition 
approach , in Beddow , J. K. , Ed., Particle Characterization in Technology , Vol. 
II, Morphological Analysis, CRC Press , Boca Raton, FL . 
43. Meloy , T. P. , Clark , N. N. , Durney , T. E. , and Pitchumani , B. ( 1985 ), Measuring the 
particle shape mix in a powder with the cascadograph , Chemical Engineering Science , 
40 ( 7 ), pp. 1077 – 1084 . 
44. Alderliesten , M. ( 1991 ), Mean particle diameters. part II: standardization of nomenclature 
. Part. Part. Syst. Charact. , 8 , 237 – 241 . 
45. Fairbridge , C. , Ng , S. H. , and Palmer , A. D. ( 1986 ), Fractal analysis of gas adsorption 
on syncrude coke , Fuel , 65 , 1759 – 1762 . 
46. Shibata , T. , and Yamaguchi , K. ( 1990 ), paper presented at the Second World Congress 
Particle Technology, Sept., Part 1, Kyoto, Japan. 
47. Johari , O. , and Bhattacharyya , S. ( 1969 ), The application of scanning electron microscopy 
for the characterization of powders , Power Technol ., 2 , 335 . 
48. Hillard , J. E. , Cohen , J. B. , and Paulson , W. M. ( 1970 ), Optimum Procedures for determining 
ultra fi ne grain sizes , in Burke , J. J. , Reed , N. L. , and Weiss , V. , Eds., Ultrafi ne 
Grain Ceramics , Syracuse University Press , Syracuse, New York , pp. 73 . 
49. Oel , H. J. ( 1969 ), Crystal growth in ceramic powders , Gray , T. J. , and Frechette , V. D. , 
Eds., Kinetics of Reactions in Ionic Systems , Plenum , New York , p. 249 . 
50. Gotoh , K. , and Finney , J. L. ( 1975 ), Representation of the size and shape of a single 
particle , Powder Tech ., 12 , 125 – 130 . 
51. Heffel , C. , Heitzmann , D. , Kramer , H. , and Scarlett , B. ( 1995 ), paper presented at the 
6th European Symp. Particle Size Characterization, Partec 95, Nurenberg, Germany. 
52. Karasikov , N. , Krauss , M. , and Barazani , G. ( 1988 ), in Lloyd , P. J. , Ed., Particle Size 
Analysis , Wiley , New York . 
53. Manohar , B. , and Sridhar , B. S. ( 2001 ), Size and shape characterization of conventionally 
and cryogenically ground turmeric (Curcuma domestica) particles , Powder Technol ., 
120 , 292 – 297 . 
54. Abrahamsen , A. R. , and Geldart , D. ( 1980 ), Behaviour of gas - fl uidized beds of fi ne 
powders, Part I. Homogeneous expansion , Powder Technol ., 26 , 35 – 46 . 
55. Shibata , T. , Tsuji , T. , Uemaki , O. , and Yamaguchi , K. ( 1994 ), Am. Inst. Chem. Eng. Part 
1 , 59 , 95 – 100 . 
56. Realpe , A. , and Vel a zquez , C. ( 2005 ), Powder Technol ., 169 , 108 – 113 . 
57. Badawy , S. I. , Lee , T. J. , and Menning , M. M. ( 2000 ), Effect of drug substance particle 
size on the characteristics of granulation manufactured in a high - shear mixer , AAPS 
PharmSciTech , 1 ( 4 ). 
58. De Boer , A. H. , Bolhuis , G. K. , and Lerk , C. F. ( 1978 ), Bonding characteristics by scanning 
electron microscopy of powder mixed with magnesium stearate , Powder Technol ., 
20 , 75 – 82 . 
59. Duberg , M. , and Nystrom , C. ( 1986 ), Studies on direct compression of tablets. 17. Porosity 
pressure curves for the characterization of volume reduction — Mechanisms in powder 
compression , Powder Technol ., 46 , 67 – 75 . 
60. Krycer , I. , Pope , D. G. , and Hersey , J. A. (1982), The prediction of paracetamol capping 
tendencies , J. Pharm. Pharmacol , 34 , 802 – 804 . 
61. Malamataris , S. , Bin - Baie , S. , and Pilpel , N. ( 1984 ), Plasto - elasticity and tableting of 
paracetamol, Avicel and other powders , J. Pharm. Pharmacol . 36 , 616 – 617 . 
62. Garekani , H. A. , Ford , J. L. , Rubinstein , M. H. , and Rajabi - Siahboomi , A. R. ( 2001 ), 
Effect of compression force, compression speed, and particle size on the compression 
properties of paracetamol , Drug Dev. Ind. Pharm ., 27 ( 9 ), 935 – 942 . 

1191 
6.8 
ORAL EXTENDED - RELEASE 
FORMULATIONS 
Anette Larsson ,1 Susanna Abrahmsen-Alami, 2 and Anne Juppo 3 
1 Chalmers University of Technology, G o teborg, Sweden 
2 AstraZeneca R & D Lund, Lund, Sweden 
3 University of Helsinki, Helsinki, Finland 
Contents 
6.8.1 Introduction 
6.8.1.1 Background 
6.8.1.2 Biopharmaceutical Aspects on Oral ER Formulations 
6.8.1.3 Infl uence of Drug Properties 
6.8.1.4 Principles for Extended Drug Release 
6.8.2 Insoluble Matrix Tablets 
6.8.2.1 Principles of Formulation and Release Mechanisms 
6.8.2.2 Manufacturing of Insoluble Matrix Tablets 
6.8.3 Membrane - Coated Oral Extended Release 
6.8.3.1 Principles of Formulation and Release Mechanisms 
6.8.3.2 Manufacturing of Oral Membrane - Coated Systems 
6.8.4 Hydrophilic Matrix Tablets 
6.8.4.1 Principles of Formulation and Release Mechanisms 
6.8.4.2 Manufacturing of Hydrophilic Matrix Tablets 
6.8.5 Comparison and Summary of Different Technologies 
6.8.6 Other Oral ER Formulations 
References 
6.8.1 INTRODUCTION 
6.8.1.1 Background 
In order to achieve therapeutic effect, a drug needs to reach the right place in the 
body at the right time. For some drugs, this may be achieved by simple solutions or 
solid dosage forms with an instant drug release while, for others, one has to modify 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

1192 ORAL EXTENDED-RELEASE FORMULATIONS 
the drug release. To understand the literature within the area of modifi ed drug 
release, it is important to be aware of the standard terms used for dosage forms 
within this fi eld. Malinowski and Marroum have summarized these terms in 
the book Encyclopedia of Controlled Drug Delivery [1] . The authors state that 
modifi ed - release (MR) formulations refer to “ dosage forms for which the drug 
release characteristics of time course and/or location are chosen to accomplish 
therapeutic or convenience objectives not offered by conventional dosage forms. ” 
One group of MR formulations is the delayed - release dosage form, which does not 
release the drug immediately after administration. One example of delayed - release 
formulation is the enteric - coated formulations. Another subgroup of MR formulations 
is the extended - release (ER) dosage forms, which are the focus of the present 
chapter. According to a defi nition from the U.S. Pharmacopeia (USP), ER formulations 
can be referred to as dosage forms that allow at least a twofold reduction in 
the dosing frequency compared to conventional dosage forms [2, 3] . 
The interest in oral ER formulations has dramatically increased in recent years. 
This can be seen in Figure 1 , where the bars in the diagram correspond to the 
number of publications found in a search in the database SciFinder Scholar 2006 
[4] that include the words oral extended release . This increase in publications con- 
fi rms that there are many ongoing activities in this fi eld. The expression oral extended 
release occurs in the database for the fi rst time in 1954, when Yamanaka et al. utilized 
the slow dissolution rate of various salts of a drug (pyrimidine penicillin) to 
extend the period of time when the drug had a clinical effect [5] . In 1959 Robinson 
and Suedres made a formulation of sulfamethylthiadiazole together with hydrogenated 
castor oil, which was suspended in an aqueous vehicle, creating a formulation 
with extended drug release [6] . Later, in the late 1950s and early 1960s Sj o gren and 
Frykl o f compressed active substances (e.g., pentobarbitone sodium and theophyl- 
FIGURE 1 Number of publications containing the words “ oral extended release ” identifi ed 
in the database SciFinder Scholar 2006 for the fi ve - year period 1960 – 2005 [4] . 
0 
200 
400 
Number of publications 
600 
800 
1000 
1200 
1400 
1600 
1800 
2000 
2001– 
2005 
1996– 
2000 
1991– 
1995 
1986– 
1990 
1981– 
1985 
Five-year periods 
1976– 
1980 
1971– 
1975 
1966– 
1970 
1961– 
1965 
>1960

INTRODUCTION 1193 
line) together with polyvinyl chloride (PVC) and obtained extended drug release 
from these insoluble matrix tablets [7] . In an early publication they showed that 
increased dose loadings or addition of channeling agents increased the drug release 
rate. It is also interesting to note that the most popular ways to prepare oral 
ER formulations today were already mentioned in an early review from the early 
1970s [8] . 
6.8.1.2 Biopharmaceutical Aspects on Oral ER Formulations 
The clinical effect of low - molecular - weight substances is often related to the concentration 
of the drug in the blood plasma. Classical blood plasma profi les for both 
immediate - release (IR) and ER formulations are shown in Figure 2 . It is well known 
that a drug only has a clinical effect when the concentration in the blood plasma is 
above the minimum effective concentration (MEC). If the concentration of the 
active substance is above the maximum safe concentration (MSC), the side effects 
will be unacceptable. The interval between the MEC and MSC is called the therapeutic 
window or therapeutic range, and the time when the concentration is above 
the MEC is called the “ duration ” of the drug. One aim of ER formulations is to 
increase the time the substance is above its MEC by continuous release of the drug 
from the formulation. Under optimal conditions the rate - limiting step in the drug 
absorption process of an ER formulation is its release rate, which then can be 
directly related to the concentration of the drug in the blood plasma. When the drug 
release rate from an ER formulation is constant, the blood plasma concentration 
will be constant under ideal conditions, whereas ER formulations with time - 
FIGURE 2 ( a ) Schematic picture of blood plasma concentration profi le after administration 
of a drug to an individual, including the MSC, MEC, therapeutic range, and duration. 
( b ) Repeated administration of IR formulation (four times daily) of a drug with short pharmacokinetic 
half - time and administration once daily of an ideal ER formulation with constant 
drug release (broken line) or ER formulation with nonconstant drug release rate 
(dotted line). 
0
5 
10 
15 
20 
25 
30 
Conventional IR 
Ideal ER 
ER 0
5 
10 
15 
20 
25 
30 
n 
Time (h) 
Blood plasma concetration 
Time (h) 
Blood plasma concentration 
MEC 
MSC 
Duration 
Therapeutic range 
MEC 
MSC 
(a) (b) 
0 5 10 15 20 25 0 5 10 15 20 25

1194 ORAL EXTENDED-RELEASE FORMULATIONS 
dependent drug release rate may give rise to time variations in the concentration 
of the drug in the blood plasma (Figure 2 ). 
The ER concept might offer several advantages, such as reduction in administration 
frequency, reduction of side effects, less irritation in the gastrointestinal tract, 
and improved patient compliance. Speers and Bonnano have also mentioned some 
economic aspects of ER formulations, such as the possibility to patent line extensions 
and to reduce manufacturing costs since fewer units are required to obtain 
the same effect [9] . On the other hand, ER formulations may have several drawbacks, 
for example, large variations in effect between patients due to varying physiological 
factors within the patient group, limited transit time for the ER formulation, 
drug stability problems during the gastrointestinal passage, and more severe complications 
such as dose dumping. 
In order to obtain a clinical effect by an orally administered drug, it is, for 
example, required that the drug is (i) dissolved and released from its formulation, 
(ii) transported over the mucosal barrier, and (iii) has passed from the lumen to the 
systemic blood circulation without being metabolized by, for example, the lumen 
or the liver. The drug dissolution rate and the rate of absorption of the dissolved 
active substance as well as the relation between these processes are important, in 
particular the dissolution process since the absorption of undissolved drug particles 
can be disregarded. 
The ER formulations can be a single - unit, monolithic system or multiple - unit 
systems containing many individual units with extended release. Multiple - unit 
systems consist of many small pellets and are normally produced by extrusion and 
spheronization or coating on inert spheres [2, 10] . The composition and ER mechanism 
can vary for multiple - unit systems, and some examples are membrane - coated 
reservoir systems and polymer - or lipid - based matrix systems, where the matrices 
can be made of both soluble and insoluble carriers [11 – 16] . 
From a biopharmaceutical point of view, the multiple - unit systems have many 
advantages, for example, a more consistent gastrointestinal transit compared to 
larger monolithic systems [17] . The gastrointestinal transit times for monolithic and 
multiple - unit systems were compared in a study by Abrahamsson et al. [17] . It was 
found that the gastric emptying time for the small multiple units was considerably 
shorter than that of larger monolithic systems (on average 3.6 and 9.6 h, respectively). 
The transit times through the small intestine were approximately equal, 
whereas the transit time in the colon for the multiple units was longer compared to 
the monolithic system, which was explained by different infl uence of the motility 
on the different systems. Another advantage with multiple - unit compared to monolithic 
systems is that the effects of dose dumping become less severe [2] . A breakage 
and instant drug release from one pellet will have considerably lower effect than 
breakage of one monolithic system. 
There may also be development and manufacturing advantages and disadvantages 
with multiple - unit systems compared to monolithic systems. The dose for 
multiple - unit systems may be easier to adjust since one can readily increase the 
number of pellets in the formulation and thus increase the dose. One can adjust the 
drug release profi les for multiple - unit systems by mixing pellets with different 
release profi les. The multiple - unit systems offer the possibility to mix pellets containing 
different active substances, which can be an advantage for the patients who 
then only need to take one formulation at a time (containing more than one active 

INTRODUCTION 1195 
substance) instead of several formulations. Disadvantages with multiple - unit systems 
may also exist; for example, the time to develop the multiple - unit systems may well 
be longer than for monolithic systems. There are greater challenges in the scaleup 
procedure for the multiple - unit systems since several expensive and specialized 
types of equipment may be needed [18] . For fi lm - coated formulations, additional 
changes in the drug release rate may be obtained upon storage as a consequence 
of aging of the fi lm. However, this change can be suppressed by introducing a curing 
step in the production [10] . 
6.8.1.3 Infl uence of Drug Properties 
Drug properties that are important to consider during development of IR tablets 
are metabolism, stability, permeability, and solubility [18, 19] . In the development 
of ER formulations, these aspects are also important, but in addition to IR formulations, 
they must be considered in relation to the different environments that the ER 
formulations meet during their passage through the gastrointestinal tract, and some 
of these aspects will be discussed briefl y below. It can be mentioned that, based on 
these initial properties for drug candidates, Thrombre has constructed a feasibility 
assessment fl ow chart for ER formation development [18] . 
The pharmacokinetic half - life for a drug may give an indication of whether a 
conventional or ER formulation is to be chosen. For drugs with short biological t 50 
(less than 1 – 2 h), devices that continuously release the active substance are required 
[2, 18] . For drugs with lifetimes longer than about 10 h, ER formulations may not 
add any benefi ts compared to IR formulations. However, for drugs with half - lives 
between these limits, ER formulations may be a good alternative to IR 
formulations. 
The stability of a drug in the solid state or in aqueous solution is a critical parameter 
when selecting an appropriate manufacturing process. A drug in an oral ER 
formulation reaches aqueous environments with, for example, variations in pH (1 – 
8), ionic strength, and bile salt concentration, which requires high chemical stability 
of the drug [19] . Furthermore, the substance should be stable not only against 
chemical degradation such as hydrolysis but also against enzymatic degradation 
(metabolism) during the passage from the lumen to the systemic blood circulation. 
The drug is released from oral ER formulations along the whole gastrointestinal 
tract. This implies that, in contrast to IR formulations, drug permeability must be 
good along the whole gastrointestinal tract for drugs in ER formulations [19] . Furthermore, 
the solubility and the dissolution rate of the drug are extremely important 
to consider, since these factors will directly infl uence the release rate for the drugs 
from ER formulations. The dissolution rate can be described as the fl ux J of dissolved 
material from a drug particle and, according to the Noyes – Whitney equation, 
it is [20, 21] : 
J 
D
h 
C C = . ( ) s b (1a) 
where D is the drug diffusion coeffi cient, h is the thickness of the stagnant diffusion 
layer around the particles, and C s and C b are the concentrations of the drug at the 
particle surface and in the surrounding bulk media, respectively. For substances in 

1196 ORAL EXTENDED-RELEASE FORMULATIONS 
their most stable solid - state form, the concentration C s close to the particle surface 
is equal to the saturated concentration. However, when a substance is in a different 
polymorphic state or in an amorphous state, C s can be larger than the saturation 
concentration. 
Many drugs are weak acids or bases with one or several p K a values. According 
to the Henderson – Hasselbalch equation, the solubility of an acidic drug will depend 
on the p K a and pH as 
pH p a 
i = + K 
S
S 
log 
0 
(2) 
where S i and S 0 are the concentrations of the drug ’ s un - ionized and ionized forms, 
respectively. Since the pH varies along the gastrointestinal tract, the solubility and 
the dissolution rate [Equation (1a) ] of the drug depend on the position of the drug 
in the gastrointestinal tract. Furthermore, for some drugs, such as indomethacin, the 
dissolution of the acid drug may lead to a changed microenvironmental pH within 
the stagnant layer and thus also infl uence the dissolution rate [22, 23] . 
6.8.1.4 Principles for Extended Drug Release 
The main principles related to ER systems are as follows: 
(i) Insoluble matrix formulations 
(ii) Membrane - coated solid dosage forms including osmotic pump systems 
(iii) Soluble hydrophilic matrix formulations 
Below we will discuss each of these formulation principles in terms of basic 
release mechanisms and the advantages or drawbacks associated with the different 
formulations and manufacturing processes. However, drug release from all kinds of 
ER formulations starts with hydration of the formulation and water diffusion into 
the system. The presence of water in the formulation facilitates the start of the dissolution 
process of the drug, whereby the dissolved drug can be released from the 
formulations. 
The driving forces for transport of water and drug are the differences in chemical 
potentials between the formulation and its surrounding. Due to the similarities in 
the driving forces for the dissolution of a drug and the release from an ER formulation, 
one can modify Equation (1a) to 
dM
dt 
JA 
DA 
h 
C C t= = . ( ) s b (1b) 
where M t is the amount of active drug that is released at the time t , D the drug diffusion 
coeffi cient, and J the fl ux of the drug from the formulation. The other parameters 
in Equation (1a) have been adjusted to fi t the drug release from the formulation 
and therefore A becomes the surface area of the releasing system in Equation (1b) 
(e.g., the area of the membrane - coated tablet), h the diffusion pathlength, and C s 
and C b correspond to the concentration of the dissolved active drug at the surface 

of the drug particles/formulation and in the bulk solution surrounding the ER 
device, respectively. Depending on the exact type of ER system, some modifi cations 
in Equation (1b) may be needed to fully describe the drug release. One important 
factor in the equation is the difference in concentration ( Cs . Cb ), and Cb is often 
assumed to be zero due to release under so - called sink conditions. For active substances 
in their most stable solid state, the remaining concentration, Cs , is equal to 
the saturation concentration in that medium. However, as mentioned above, Cs can 
be oversaturated or depend on pH. This means that the drug release from formulations 
depends on the solid - state properties and p Ka of the drug as well as the pH 
to which the formulation is exposed [24] . Since the pH varies along the gastrointestinal 
tract, the drug release will be dependent on the position of the formulation in 
this tract. Several attempts to avoid pH - independent drug release has been made, 
for example, by including buffers [25 – 27] . 
6.8.2 INSOLUBLE MATRIX TABLETS 
6.8.2.1 Principles of Formulation and Release Mechanisms 
The history of insoluble matrix tablets goes back to the beginning of the 1960s, when 
H a ssle and Abbott developed the Duretter and the Gradumet, respectively [2] . 
Since then, many ER tablets based on this principle have been developed. Looking 
at the homepage of the U.S. Food and Drug Administration (FDA [28] ) and searching 
for “ insoluble matrix tablets ” produces more than 140 hits, which indicates that 
this research area is still active. 
The term insoluble matrix tablet refers to tablets in which the drug is embedded 
in an inert carrier that does not dissolve in the gastrointestinal fl uids. The carrier 
material in insoluble matrix tablets can be based on insoluble lipids or polymers, 
both matrix builders whose function it is to keep the matrix together during the 
passage through the gastrointestinal tract and thus prolong the diffusion path of the 
drug before it is released from the formulation. The drug can be dispersed or dissolved 
or both in a matrix carrier (see Figure 3 ) and, depending on the formulation, 
different mechanisms can be regarded to take place: 
• Dissolved drug in the matrix diffuses through the matrix. 
• Dissolved drug in the matrix diffuses through pores in the matrix. 
• Dispersed drug dissolves and diffuses through the matrix. 
• Dispersed drug dissolves and diffuses through pores in the matrix. 
FIGURE 3 Schematic pictures of insoluble matrix systems. Left: Drug (light gray) molecularly 
dissolved in carrier material (black); middle: drug particles dispersed in carrier material; 
right: drug particles dispersed in carrier material at higher drug loading, leading to 
continuous network of drug. 
INSOLUBLE MATRIX TABLETS 1197

1198 ORAL EXTENDED-RELEASE FORMULATIONS 
As early as 1963, Higuchi [29] derived an expression for drug release from 
insoluble matrix systems. In this historical paper, Higuchi derived two equations for 
two different geometries, the simple planar sheet matrix system with infi nite area 
and spherical pellets. Furthermore, two special cases were treated, one where the 
matrix is a homogeneous matrix without pores and another where the matrix contains 
pores. In the system without pores, the drug is assumed to diffuse through the 
homogeneous matrix. For matrix systems with pores the effi ciency in transport 
through liquid - fi lled pores is greater than through the solid matrix carrier. Therefore, 
the main contribution to the drug release is transport in the pores. This is 
gained by penetration of the surrounding medium into the pores, where it dissolves 
the drug. The dissolved drug can diffuse through the pores and be released at the 
surface of the matrix. The simplest theoretical treatment of the drug dissolved in 
the matrix carriers assumes the following [30] : 
• There is no breakage of the matrix. 
• There is no dissolution of the matrix. 
• There is no resistance to drug transport in the boundary layer surrounding the 
device. 
• There is no accumulation (e.g., adsorption) of the drug in the device. 
• The saturated concentration C s,m of the drug in the matrix is constant during 
the process. 
• The drug - loading concentration C 0 is larger than C s,m . 
• The drug concentration around the matrix is zero, C b = 0 (sink conditions). 
• The diffusion constant D m in the matrix is independent of the drug 
concentration. 
• The partition coeffi cient K between the matrix material and the surrounding 
release medium is independent of the drug concentration. 
A handy derivation of the equation describing the release from planar homogeneous 
matrix systems can be found in a book by Wu [30] . It is derived for a 
sheet with the area A and assumes that the concentration of dissolved drug 
inside the matrix is constant and equal to the saturation concentration C s,m . 
Under these assumptions, the amount of drug, M t , that is released at time t can be 
predicted as 
M ADKC t C C t= . ( ) [ ] m s,m s,m 2 0 
0 5 . (3) 
When the matrix contains a drug - fi lled network (Figure 3 , right image), water can 
diffuse and dissolve the drug, and this creates a pore structure. The equation 
describing the drug release from matrices with networks is modifi ed to include 
information about the created pore structure [Equation (4) ]. This can be described 
by the porosity . (the volume of the pores in proportion to the total volume of the 
device) and the tortuosity . of the pores (a measure of how much the diffusion 
path is lengthened due to lateral excursions). Also the diffusion coeffi cient D m and 
C s,m in Equation (3) are replaced with D and C s in Equation (4) , corresponding to 
the diffusion coeffi cient and the solubility of the drug in the solution inside the 
pores, respectively:

M A 
D C t 
C C t= . ( ) ... 
... 
.
. 
. s 
s 2 0 
0 5 . 
(4) 
The most common types of insoluble matrix tablets are those containing pores. 
From the equations above one can see that the drug - release depends on the solubility 
of the drug, the drug - loading concentration, and the diffusion coeffi cient, which 
is related to the molecular size of the drug. The area of the insoluble matrix tablet 
also affects the drug release and can be changed by altering the dimensions or the 
geometry of the tablet. The drug release from insoluble matrix tablets also depends 
on the porosity and pore structure of the tablet, and the drug release rate increases 
with increasing porosity. 
A comparison of Equations (3) and (4) shows that, in both equations, the amounts 
of released drug are directly dependent on the area of the device, the square root 
of the time t , the drug - loading concentration C 0 , the respective saturated drug concentrations, 
and the drug diffusion coeffi cients. In addition, the release rate (the 
time derivate of the amount of released material) depends on the square root of 
time and can be stated as 
dM
dt t 
t . 1 (5) 
As pointed out above, for ideal ER formulations, the rate - limiting step for drug 
absorption is the release rate from the ER formulation. Thus, since the release rate 
from an insoluble matrix system depends on time, the concentration of drug in the 
blood plasma will also be time dependent and not constant (Figure 2 ), which may 
be a therapeutic drawback. Another factor infl uencing the concentration of drug in 
the blood plasma is the gastrointestinal transit times. When the transit times of the 
formulations vary, the reproducibility between different administration occasions 
in one patient will be low, and furthermore, great variation in the patient group 
may be obtained. However, these conclusions are valid for all ER formulations 
based on matrix systems and not limited to insoluble matrix systems only. 
The equations above are valid when no depletion of drug occurs inside the 
device. The equations for release rate will be much more complex when depletion 
of the drug can occur [30] . However, it has been shown that, when the amount of 
released material is less than approximately 60%, the release rate will depend on 
time as t . 0.5 [29, 30] . 
6.8.2.2 Manufacturing of Insoluble Matrix Tablets 
Insoluble matrix tablets need a carrier, which can be a lipid - or polymer - based 
excipient [7, 31 – 36] . Some suggestions of carrier materials can be found in Table 1 . 
The table also presents the number of hits found upon searching the FDA ’ s homepage 
[37] for the number of times an excipient is registered as a component in oral 
extended, sustained, or controlled formulations. This list gives an indication of how 
often these excipients are commercially used in oral ER formulations but does not 
automatically tell us the exact formulation or exact mechanistic effect of the excipient. 
The choice of carrier material is important, and one should be aware of possible 
INSOLUBLE MATRIX TABLETS 1199

1200 ORAL EXTENDED-RELEASE FORMULATIONS 
TABLE 1 FDA Registered Oral ER Formulations Containing Commonly Used 
Excipients in Insoluble Matrix Formulations 
Excipient 
Number of Hits on 
FDA Homepage Content Interval (mg) 
Lipid based 
Carnauba wax 9 46 – 300 
Stearyl alcohol 4 25 – 244 
Glyceryl behenate 3 15 – 51 
Castor oil 2 23 
Cottonseed oil, hydrogenated 2 58 – 402 
Cetyl alcohol 2 44 – 59 
Paraffi n 2 50 – 150 
Stearic acid 2 26 – 180 
Castor oil, hydrogenated 1 295 – 410 
Vegetable oil, hydrogenated 1 228 
Mineral oil 1 
Microcrystalline wax 0 
Insoluble polymer 
Ethylcellulose 9 15 – 309 
Ammonia methacrylate copolymer 5 37 – 138 
Polyvinyl acetate 1 46 
Polyethylene 0 
Inorganic 
Calcium phosphate (dibasic) 6 33 – 335 
Source : http://www.accessdata.fda.gov/scripts/cder/iig/index.cfm . 
exposure of lipid - based formulations to erosion, which can be the result of enzymatic 
degradation of the lipids [38] . This will of course also infl uence the drug 
release rate. 
In the compositions of insoluble matrix systems, excipients other than the carrier 
material are needed to obtain products with processability and that meet requirements 
from the pharmacopedias. Examples of categories of excipients included in 
insoluble matrix tablets are binders, lubricants, glidants, colorant, taste maskers, 
and channeling agents. As mentioned above, the drug release rate can be regulated 
by the porosity in the insoluble matrix system. The properties and amounts of drugs 
and excipients that can create pores will have a large impact on the release rate. 
Examples of channeling agents are sugars, salts such as sodium chloride, and polyols 
[2] . The pore structure also depends on other factors such as the particle sizes of 
the excipients and the drug, the size and porosity of the granules, and the compaction 
pressure. 
The choice of process steps depends on the properties of the drug and the chosen 
excipients. For insoluble matrix tablets one often mixes the active substance with 
the excipients. Either this mixture can be directly compressed to matrix tablets or 
the powder mixture can be exposed to a granulation technology to enlarge the 
particle sizes. One such technique is dry granulation, that is, compaction of the 
mixture in, for example, a roller compactor, followed by milling to desirable granule 
sizes. Another granulation technique is melt granulation, where the melted granulation 
liquid agglomerates the particles to granules. The most common granulation 

method for insoluble matrix tablets is probably wet granulation [2, 7] with aqueous - 
based or organic granulation liquids. The wet granulated masses are dried in fl uid 
bed driers or ovens. In order to increase the drying speed, microwaves can be used. 
The powder mixtures or granules are compressed in ordinary tableting machines 
[7] . However, it is an advantage if the compaction pressures can be carefully monitored, 
since this pressure may infl uence the porosity and thus the drug release. The 
fi nal tablet can be coated to, for example, mask the taste. 
Alternative production methods to the traditional compaction of powder to 
insoluble matrix tablets are available, some of which are based on melting technologies, 
but of course these methods rely on the ability of the carrier materials 
or additives to melt. The drug is commonly dissolved or dispersed in the melt. 
This melt can be fi lled into hard gelatin capsules [31] or it can be spray chilled by 
pressing it through a nozzle into a vessel containing solid carbon dioxide [39] . Hot - 
melt extrusion of polymer - based systems to form multiple - unit systems has been 
investigated. The carrier material in these cases can be, for example, Eudragit 
[40 – 42] . 
6.8.3 MEMBRANE - COATED ORAL EXTENDED RELEASE 
6.8.3.1 Principles of Formulation and Release Mechanisms 
One way to protect the drug from being directly released is to coat the system with 
an insoluble fi lm. The drug is suspended or dissolved in a reservoir system which 
can consist of monolithic or multiple - unit systems. The MR fi lms surrounding the 
reservoir will be insoluble and thus give the system extended drug release properties 
or they can become soluble by external trigging. The latter case is defi ned as a 
delayed - release formulation, which means that the formulation does not release 
directly after administration. This can be achieved, for example, by enteric coatings, 
where the fi lm - forming materials are insoluble in aqueous solutions at low pH but 
soluble at high pH values. This results in delayed release from an enteric - coated 
formulation, since the pH is low in the stomach. When the units are transported 
into the intestine, the pH increases, the fi lm dissolves, and the drug can be released 
immediately. This type of formulation will not be further discussed here, but the 
work of Hogan is recommended for further information on these systems [43, 44] . 
In ER reservoir systems, a membrane surrounds a reservoir of the drug, also 
called the core of the system. The membrane controls the drug release and the 
driving force is the difference in chemical potential over the membrane, which can 
be correlated with a concentration gradient over the membrane. The transport of 
the drug through the ER membranes can be divided into three different mechanisms 
[45 – 47] : 
• Diffusion through the membrane 
• Diffusion through pores and cracks in the membrane 
• Osmotic transport through pores, cracks, or drilled holes 
The overall drug release process for common membrane - coated systems has been 
shown to pass through three different time periods (Figure 4 ) [48] . During the initial 
MEMBRANE-COATED ORAL EXTENDED RELEASE 1201

1202 ORAL EXTENDED-RELEASE FORMULATIONS 
period, the amount of released drug will be low. The water penetrates the membrane 
and at the same time starts to dissolve the material in existing pores, consisting 
of water - soluble sugars, salt, or polymers. The water that has penetrated the 
membrane and reached the core starts to dissolve the active substance in the reservoir 
[11] . The concentration of the active drug increases continuously until its saturation 
concentration has been reached and pseudoequilibrium between the solid 
material and saturated solution inside the membrane has developed. The osmotic 
pressure in the reservoir will depend on the concentrations of all dissolved species 
inside the membrane, and an increase of the osmotic pressure may lead to membrane 
rupture. After an initial lag period, the number of cracks and pores becomes 
constant, and the osmotic pressure and the concentration of dissolved species in the 
reservoir and in the membrane reach their steady - state levels. When these parameters, 
which are the driving forces for the drug release, are constant, the drug release 
rate will also be constant. Therefore, a second period with time - independent drug 
release rate will occur. 
When the solid drug material inside the membrane is completely dissolved and 
the concentration of the dissolved drug decreases, a fi nal period with declining 
FIGURE 4 Release from membrane - coated reservoir system, where the three different 
stages are depicted (initial lag period, “ steady - state ” period, and fi nal depletion period). A 
schematic picture of a formulation with the drug reservoir (dark grey) surrounded by the 
membrane (middle grey) is presented at the top. The dissolution medium penetrates the 
membrane and dissolves the drug (light gray), and pores/cracks are formed through which 
the drug can be released. 
0.0 
20.0 
40.0 
60.0 
80.0 
100.0 
0 5 10 15 20 25 
Time (h) 
Fraction released (%)

release rate will be entered. Ragnarsson et al. [49] have shown that the solid material 
disappears earlier as the drug solubility increases and that the third stage with 
time - dependent and decreasing release rate appears earlier compared to drugs with 
lower solubility. 
The contribution of diffusion to the release process can be modeled by using 
Fick ’ s fi rst law. For diffusion of a substance through the membrane, it will turn out 
as (assuming sink conditions) 
dM
dt 
JA 
D K A 
h 
C C 
D K AC 
h 
t= = . ( )= m m 
s b 
m m s (6) 
where K m is the partition coeffi cient for the drug between the membrane and solution, 
D m is the diffusion coeffi cient in the membrane, A is the area of the membrane, 
h is the thickness of the membrane, and C s and C b are the concentrations on the 
inside of the membrane surface and in the bulk, respectively. The equation for diffusion 
through pores or cracks resembles Equation (6) : 
dM
dt 
JA 
D KA 
h 
C C 
D KAC 
h 
t= = . ( )= 
. 
. 
. 
. s b 
s (7) 
where . and . are introduced to describe the porosity and tortuosity in the 
membrane, respectively. The parameters D m and K m in Equation (6) are replaced 
in Equation (7) by D and K , which are the diffusion coeffi cient in the solution 
inside the pores and the partition coeffi cient between the solution and materials 
surrounding the liquid - fi lled pores, respectively. Equations (6) and (7) depend on 
the concentration gradient over the membrane, and both are independent of the 
time. 
The osmotic contribution to the drug transport is described by the so - called 
Kedem – Katchalsky equations (based on nonequilibrium thermodynamics) [50, 51] . 
A simplifi ed version is 
dM
dt 
JA 
AC L
h 
t= = s p... (8) 
where . . is the osmotic pressure difference over the membrane and L p and . are 
the hydraulic permeability and the refl ection, respectively. 
A comparison of Equations (6) – (8) shows important similarities; they depend on 
the solubility of the drug, the area of the device, and the thickness of the membrane. 
This means that an increased solubility, larger area of the membrane, and thinner 
membranes will facilitate the drug release rate. This can be exemplifi ed by a study 
by Ragnarsson and Johansson [11] , who showed that, for different salt forms of 
metoprolol, an increased solubility also increased the drug release rate, which was 
predicted from the equations. Furthermore, Equations (6) – (8) show constant and 
time - independent release rates. This constant amount of released drug will be a 
biopharmaceutical benefi t since it theoretically makes it possible to achieve a constant 
concentration of the drug in the blood plasma. 
MEMBRANE-COATED ORAL EXTENDED RELEASE 1203

1204 ORAL EXTENDED-RELEASE FORMULATIONS 
One special type of ER formulation based on coated reservoir systems is the so - 
called osmotic - controlled oral drug delivery system or osmotic pump. Pure osmotic 
systems have semipermeable membranes; that is, water can permeate the membrane 
but not other substances. These semipermeable membranes can be made of, 
for example, cellulose acetate [44] , and for such formulations the dominating release 
mechanism is the osmotic pressure [Equation (8) ]. The oldest formulation based on 
osmotic release was OROS from Alza Corporation [47] . In order to achieve drug 
release through the semipermeable membrane, a laser hole was drilled, but today 
many osmotic formulations instead use pores fi lled with water - soluble materials. 
The aspects of the formulation and different types of commercially available formulations 
are summarized in the review by Verma et al. [52] . Some advantages with 
osmotic pumps compared to other ER formulations such as hydrophilic matrix 
systems are (i) the time - independent drug release (often mentioned as zero - order 
release), (ii) the superior in vivo – in vitro correlation which facilitates further formulation 
development, and (iii) less variation between fasted and fed states. Potential 
drawbacks may be high initial development costs and lack of in - house 
competence. Drawbacks from an economic point of view may be the necessity to 
pay royalties and the need for special equipment associated with laser drilling 
technology. 
6.8.3.2 Manufacturing of Oral Membrane - Coated Systems 
The fi rst step in the production of membrane - coated systems is to prepare the 
drug reservoir, the core, of the system. The process steps for producing the core 
depend on the size of the core. In monolithic membrane - coated systems, the core 
can be a fi lled capsule or a tablet which is produced in the traditional ways. This 
may include mixing of active substance and excipients, possibly granulation and 
drying, fi lling into capsules, or compaction into tablets. The production of cores for 
multiple - unit systems (often termed pellets) is more sophisticated and may be performed 
in different ways. One, and probably the most common way, is to produce 
cores for multiple - unit systems by extrusion and spheronizing [53 – 55] . An alternative 
methodology is to coat an inert core, e.g. glass or nonpareil beads, with the 
active substance and the excipients [11] . When the drug - containing core is manufactured, 
the process continues with the coating of the release - controlling 
membrane. 
The composition of the core depends on the properties of the drug and the 
excipients, the chosen production chain, and whether the systems should be a monolithic 
or multiple - unit system. The compositions and production steps are reviewed 
by Tang et al. [56] . In general, the core will include the active substance together 
with the fi ller materials and, if necessary, solubilizers and lubricants/glidants. Classical 
fi ller materials are lactose and microcrystalline cellulose, but also other materials 
such as dextrose, mannitol, sorbitol, and sucrose can be used. However, it should 
be remembered that the dissolution of the fi ller material might infl uence the osmotic 
pressure. The effect of fi ller solubility has been investigated by Sousa et al. [54] , 
who found a relation between the solubility of the fi ller materials and drug release 
rate. For fi ller materials with large water solubility, there is a great risk that the 
membrane will rupture due to the development of an excessive osmotic pressure, 
which will infl uence the drug release rate. 

The choice of fi lm - forming materials and fi lm - coating techniques is critical for 
the drug release rate [54] . The ER membrane should remain intact during the 
release, which implies that it should not dissolve or erode. As fi lm - forming material, 
water - insoluble substituted cellulose derivatives such as ethylcellulose have been 
suggested [53, 57] as well as synthetic polymers such as methylacrylates (e.g., 
Eudragit NE 30D, RS30D or RL30D, where NE stands for nonionic and RS/RL 
correspond to cationic polymers) [55] . A commercial technology platform is available 
under the name Eudramode, which is a platform for development of multiple - 
unit systems with extended drug release based on Eudragit [58, 59] . Other 
fi lm - coating materials such as shellac and zein have been used, but a drawback with 
these naturally occurring materials is the variation in quality. To obtain a fi lm with 
satisfying release properties, channeling agents such as the hydrophilic polymers 
hydroxypropyl methylcellulose (HPMC), hydroxypropyl cellulose (HPC), and polyethylene 
glycol (PEG) or other water - soluble materials such as sodium chloride or 
lactose may be used [2, 57] . To improve the mechanical properties of the fi lm and 
thus avoid ruptures and cracks in the fi lm, plasticizers may be added to the formulation. 
Examples of plasticizers are PEGs, diethyl phthalate, triacetin, mineral oils, 
glycerol, and chlorobutanol [60] . 
The fi lm coating may be performed in different types of equipment. For coating 
of larger units, such as tablets or capsules, a rotating drum is often used, for example 
an Accela Coater, but other similar equipment may also be used [10, 53, 57, 61] . 
This type of equipment contains a perforated pan that rotates and thus mixes the 
bed of units. At the same time, a coating liquid is applied to the moving units by 
means of a spray gun, where the mixing of the units ensures uniform coverage of 
the coating. The fi lm coating is dried by blowing a stream of hot air onto the surface 
of the tablets. For all types of coating processes, there are many parameters, such 
as the temperature and relative humidity of the inlet air, drum rotation speed, 
spraying rate, and droplet size of the coating liquid, that have to be adjusted in order 
to produce good coated fi lms. 
For coating smaller units, such as pellets, the fl uid bed coating technique is used 
[56] . This is an attractive technique in which the starting material is placed in the 
coater and heated air is blown through a base plate. This leads to vigorous mixing 
of the pellet units. By changing the pressure of the incoming air stream, the material 
becomes suspended in the air. This happens when the bed starts to fl uidize and the 
bed will then have fl uidized properties similar to the properties of ordinary liquids. 
There are different designs of fl uid bed: top spray coating, bottom spray coating (or 
Wurster coating), or tangential coating. They differ with regard to placement of the 
spray guns. In top spray coating, the liquid is sprayed from the top of the equipment 
and the droplets hit the particles moving in opposite direction. In the bottom spray 
coating, the sprayed liquid drops and particles fl ow in the same direction, which 
avoids the problem of blocking the spray guns that may occur in top spray equipment. 
In the more rarely tangential spray coating equipment the base plate rotates 
and the spray guns are spraying in a tangential direction to the spiral moving particles 
[10, 62] . 
A critical parameter for obtaining fi lms with desirable properties is the creation 
of coating droplets, a process often referred to as atomization, that is, when a bulk 
liquid is dispersed in air to form a spray or a mist [61] . Atomization is done by 
letting pressurized air and bulk solution pass simultaneously through a nozzle (spray 
MEMBRANE-COATED ORAL EXTENDED RELEASE 1205

1206 ORAL EXTENDED-RELEASE FORMULATIONS 
gun). The air divides the droplets into smaller units, but in spite of the name atomization, 
the droplets are dispersed not at atomic scale but rather in the nano - to 
micrometer - scale range [61] . 
The mechanism for fi lm formation depends on whether the polymer is dissolved 
or dispersed as small latex particles in the solution. For both technologies the fi lm 
formation process starts with wetting and spreading of the coating droplets on the 
surface of the reservoir system [10] . For the case with polymer dissolved in solution, 
the fi lm formation mechanism continues with evaporation of the solvent. This leads 
to an increase of the polymer concentration, and at a certain limit the polymers 
precipitate and a coated fi lm is formed. In the case of ER fi lms, the most commonly 
dissolved polymer is ethyl cellulose and the most commonly used solvent is ethanol 
but other organic solvents can also be used. The use of organic solvent is problematic 
from the SHE (safety, health, and environmental) point of view and therefore 
these aspects must be considered before starting to use organic - based coating 
process technologies. An alternative process methodology is to disperse particles 
of the fi lm - forming polymer in an aqueous solution [63] . Film formation of dispersed 
particles undergoes the following steps: evaporation of water, close packing of the 
particles, deformation of the particles, and annealing of the particles by migration 
of individual polymer chains between the particles to form a coherent, smooth fi lm 
coating. The fi rst steps of the process occur in the coating equipment but the last, 
the annealing step, may continue days after the coated product has left the coating 
equipment. 
6.8.4 HYDROPHILIC MATRIX TABLETS 
6.8.4.1 Principles of Formulation and Release Mechanisms 
Several recent informative review articles on hydrophilic matrix systems have been 
published [e.g., 24 , 64 ]. Hydrophilic matrix tablets are composed of an active substance, 
a hydrophilic polymer, release modifi ers, lubricants, and glidants. The technology 
goes back to the mid - 1960s when Lapidus and Lordi [65, 66] and Huber 
et al. [67] determined the drug release from hydrophilic matrix systems. The release 
mechanism for this type of formulation starts with dissolution of hydrophilic matrix 
polymers and the formation of a highly viscous polymer layer around the tablet 
(Figure 5 ) [64] . This layer is often referred to as a gel layer even though it normally 
contains only physical entanglements and not chemical cross - linkers, which is traditionally 
required for gels. However, the gel layer surrounds the inner (more or 
less dry) part of the tablet and this part is called the core. In traditional hydrophilic 
tablets, the active substance particles are embedded in the matrix carrier. The dissolution 
process of the active substance can start when the carrier material has dissolved 
in water and formed the aqueous gel layer, since without exposure to water 
the active substance cannot dissolve. Therefore, the “ dry ” core will shield the active 
drug from dissolution, which is one reason for the extended drug release from this 
type of formulation. 
The release process for hydrophilic matrix tablets can be schematically described 
as in Figure 5 . The left side of the fi gure shows a hydrophilic tablet undergoing dissolution, 
swelling, and release. An interface between the solution and gel layer, here 

called the erosion front, can be identifi ed, and the polymer chains and drug molecules 
are released at this front. In the gel layer, the polymer concentration will 
decrease (Figure 5 ) from a highly concentrated solution at the swelling front, the 
interface between the gel layer and more or less dry core, to a diluted polymer 
solution at the erosion front [68] . Some authors have suggested that the polymer 
concentration at the erosion front is related to the overlap concentration [68 – 70] . 
At the same time, the water content in the gel layer gradually increases from the 
center of the tablet toward the erosion front, and thus the dissolution of the active 
substance particles can start already in the gel layer. However, the volume fraction 
of dissolved drug depends on the amount of available water, which in turn is a function 
of the position in the gel layer [64, 71] . Assuming that the drug saturation 
concentration is equal in water solutions and polymer gels, the volume fraction 
dissolved drug will correlate with the volume fraction water available, and the 
volume fraction drug will gradually increase with increasing distance from the core. 
Far from the core, the variations in the concentration of polymer and water are less 
pronounced and, as long as solid particles coexcite with the saturated drug solution, 
the volume fraction of the drug will theoretically be almost constant. A third front 
has also been introduced, the diffusion front, which corresponds to the position in 
the gel layer where all of the active substance has dissolved. Between the swelling 
and dissolution front, dissolved and undissolved drug particles will coexist, but 
between the dissolution and erosion fronts, only dissolved drug molecules occur. In 
this region, the diffusion of drug out from the matrix will give rise to a decrease in 
the volume fraction of the drug. 
Achieving a mechanistic understanding of drug release from hydrophilic matrix 
tablets is not a trivial task since the release depends on the properties of both the 
polymer and the drug. The swelling process is directly related to the properties of 
the polymers, and this is an important factor for drug release, since the polymer 
swelling process can, for example, transport individual drug particles through the 
FIGURE 5 ( a ) Hydrophilic matrix system shown with core (dark gray) and drug parti cles 
(small dark gray particles). The swelling, diffusion, and erosion fronts are depicted. 
( b ) Dependencies of volume fraction polymer (solid line) and dissolved drug (broken line) 
as function of position in matrix system together with swelling, diffusion, and erosion fronts. 
Top shows how the solid drug particles diminish in size. ( c ) Examples of drug release as 
function of time for erosion - controlled ( n = 1, solid line) and diffusion - controlled systems 
(broken line, n = 0.5). 
0 
25 
50 
75 
100 
Time (h) 
Fraction released (%) 
Swelling front Diffusion front Erosion front 
(a) (b) 
0 6 12 18 24 
(c) 
HYDROPHILIC MATRIX TABLETS 1207

1208 ORAL EXTENDED-RELEASE FORMULATIONS 
gel layer, which has been shown by Adler et al. [72] . Macroscopically, polymer 
swelling can be observed as an increase in the size of the tablet. On a molecular 
level, the swelling depends on the dilution and transport of water into the gel layer. 
This transport is driven by changes in the chemical potential, and the main contribution 
is the increase of conformational entropy when the polymer chains are diluted 
[73] . The kinetics of the swelling process may vary. A faster polymer release rate 
compared to the swelling rate results in a movement of the erosion front toward 
the center of the tablet and the size of the tablet will diminish. Conversely, the tablet 
size increases when the polymer release is slower than the swelling process. When 
the swelling rate is on the same order as the polymer release rate, the position of 
the erosion front (i.e., tablet size) will remain constant. 
The swelling front between the gel layer and amorphous (or semicrystalline) core 
material has traditionally been described as corresponding to a transition of the 
solid states of the polymers. The polymers in the core, for example in HPMC tablets, 
are in a glassy state, and the polymer material is transformed to a rubbery state due 
to the fact that water acts as a plasticizer and decreases the glass transition temperature 
[74] . This rubbery state can be regarded as a polymer solution, and therefore 
the glassy - to - rubbery state transition can be regarded as a dissolution process 
of the polymer, where the dissolution rate will determine the position of the swelling 
front. A commonly described special case for hydrophilic tablets is the so - called 
front synchronization, which is when the movements of the swelling front and the 
erosion front occur equally fast. This special case corresponds to a constant gel layer 
thickness. 
Depending on the drug solubility and the dissolution, swelling, and release processes 
of the polymer, either of two different drug mechanisms can be observed: 
erosion - or diffusion - controlled drug release (Figure 5 ). One way to characterize 
these two mechanisms is to compare the drug and the polymer release. The erosion 
mechanism is characterized by equal release of the polymer and the drug, whereas 
when the release of the drug is faster than that of the polymer, this is called diffusion 
- controlled drug release [64] . The diffusion - controlled mechanism occurs when 
the diffusion front, the border between undissolved and dissolved drug, is displaced 
in the gel layer and the drug can effi ciently diffuse out from the gel layer. The 
erosion mechanism dominates when the diffusion and erosion fronts overlap. This 
means that drug particles may be released from the surface of the gel layer. When 
this occurs, the drug particles will dissolve faster in the free solution than in the gel 
layer due to the fact that stirring is more effi cient in the free solution, which results 
in a decreased thickness of the unstirred boundary layer around the particles and 
thus an increased dissolution rate [Equation (1a) ]. Whether the release mechanism 
will be diffusion or erosion controlled depends on (i) the polymer release rate, 
which governs the position of the rate erosion front, and (ii) the drug dissolution 
rate, which governs the position of the diffusion front. The position of the diffusion 
front and the dissolution rate depend on the solubility of the drug. Lower solubility 
of the drug gives slower dissolution rates and hence the diffusion front can overlap 
more easily with the erosion front, which yields erosion - controlled drug release. On 
the other hand, large solubility of the drug will give diffusion - controlled drug release 
[75 – 78] . 
Traditionally, the drug release rate from hydrophilic matrix systems has been 
modeled as [79, 80] 

Q 
dM 
dt 
dt kt t n = = .0 
(9) 
where Q is the accumulated amount of released drug and k and n are constants. 
The values of n have been suggested to describe the drug release mechanisms. 
Release from a planar surface with n = 1 has been shown to correspond to erosion - 
controlled drug release and n = 0.5 to pure diffusion - controlled drug release. This 
is strictly only valid when the polymer release has n = 1, but this is often the case. 
In practice, for hydrophilic matrix systems, one often fi nds n to be between 0.5 
and 1, indicating that both the diffusion of the drug and the polymer erosion infl uence 
drug release. For other geometric shapes, such as tablet shapes, the limits for 
n shift to 0.45 and 0.89 for diffusion - and erosion - controlled release, respectively 
[81 – 83] . 
Another popular way to describe the drug release is to characterize the infl uence 
of the relative contributions of erosion and diffusion to drug release as [84] 
Q at bt m m = + 2 (10) 
where a , b , and m are constants. The fi rst factor in Equation (10) should represent 
the Fickian diffusional contribution and the second term to the erosion contribution 
to the drug transport. One mechanistic drawback with this approach is that it treats 
the diffusion and erosion processes as independent of each other, which they are 
probably not in any practical case. 
The drug release from hydrophilic matrix tablets has been found to vary with the 
polymer parameters, the composition of the formulation, and the process parameters. 
Examples of important polymer - related parameters with signifi cant infl uence 
on drug release are the viscosity and the hydrophilicity of the polymer, where polymers 
of larger viscosity grades give lower release rates and longer durations of the 
release [85] and an increased hydrophilicity gives larger swelling and faster drug 
release, which was found by comparing different degrees of substitution of HPMC 
[86] . The drug release rate also depends on the composition of the formulation. 
When components with high water solubility, such as lactose, are included in the 
matrix, the drug release increases, which can be seen as a dilution of the gel - forming 
material [69, 87] . Similarly, an increased amount of a soluble active substance also 
increases the release rate of drugs, probably also due to the corresponding decrease 
of the relative amount of hydrophilic polymer [88, 89] . The size and shape of the 
tablet also infl uence drug release, and the release rate increases as the area - to - 
volume ratio increases [90 – 93] . The infl uence of particle size on the drug release 
rate has also been investigated [89, 94, 95] . The size of polymer particles seems to 
have low infl uence on drug release when there is enough gelling polymer available 
to quickly form a coherent gel layer. In contrast, at low amounts of hydrophilic 
matrix polymer, gel formation may be too slow, which makes polymer particle size 
important. In this case, the matrix may disintegrate before it develops a coherent 
gel layer [95 – 97] . The effect of drug particle size on the drug release rate depends 
on the solubility of the substance. Varying the particle size of drugs with high solubility 
seems to have little infl uence [89] , whereas the release rate may depend on 
the particle sizes of drugs with moderate solubility [24, 89] . 
HYDROPHILIC MATRIX TABLETS 1209

1210 ORAL EXTENDED-RELEASE FORMULATIONS 
The effects of process parameters on drug release have been discussed in the 
literature. Different granulation technologies, such as dry granulation [98 – 100] or 
wet granulation [101 – 103] (which includes fl uid bed granulation [104, 105] ), have 
been used. Also direct compression has been used for production of hydrophilic 
matrix tablets [95] . The effects on the choice of production steps may be critical 
when the relative fraction of the polymers is low in the formulations. The effect of 
the compaction pressure on drug release has also been studied. Several authors [89, 
88, 106, 107] have found that, while the compaction pressure has a signifi cant effect 
on the tensile strength of the tablets, it has a minimal infl uence on drug release. 
This can be due to the fact that, when a coherent gel layer is formed, only the 
parameters governing the performance of this gel layer are important, and since 
parameters such as porosity do not affect the gel layer, they are of low importance 
for the drug release rate. 
6.8.4.2 Manufacturing of Hydrophilic Matrix Tablets 
The traditional way of producing hydrophilic matrix tablets resembles the production 
of the core for membrane - coated tablets and insoluble matrix tablets. It includes 
a mixing step, possibly a granulation step, a compaction step, and sometimes a 
coating step. However, one large difference between the production of insoluble 
and soluble matrix systems is notable; the latter matrix type has strong interactions 
with water, which complicates the production steps when water is present [108] . 
Therefore, for hydrophilic matrix systems with large fractions of hydrophilic polymers, 
traditional wet granulation with water as granulation liquid may cause problems 
with formation of hard lumps [108] . To avoid this problem, a new technique 
using foam granulation has been suggested [109, 110] . During granulation, the 
foams will fl ow on the top of already foam - wetted particles, which may lead to 
superior distribution of the granulation liquid. An alternative wet granulation 
method is to use organic solvents such as ethanol as granulation liquid [111] . 
However, even if the production of granules with good compaction properties can 
be maintained in this way, it may, as already mentioned, be an advantage to consider 
the SHE aspects before choosing organic solvents as granulation liquid. Another 
alternative to wet granulation is to use dry granulation technologies such as roller 
compaction [98, 99, 100] . Although this technique has several advantages compared 
to traditional wet granulation with water, it may result in lower tensile strengths of 
the tablets [112] . An alternative to granulation technologies is direct compression. 
This can be done by purchasing special direct - compression qualities of the hydrophilic 
polymers. A drawback that always arises in relation to direct compression is 
the diffi culty to achieve content uniformity of the tablets, a problem that increases 
with decreasing doses of the active drug. Therefore, special care should be taken 
with regard to formulations with low doses of active substance when direct compaction 
is used [2] . 
Water - soluble hydrophilic matrix systems may also be extruded, both to 
monolithic systems and to multiple - unit systems [12, 14 – 16] . Polyethylene oxide 
(PEO) and chloropheniramine maleate have, for example, been extruded to a 
monolithic unit [113] . This manufacturing method proved more feasible for mixtures 
between low - and high - molecular - weight PEO, since systems containing only 
high - molecular - weight PEO proved too viscous and diffi cult to extrude. It was also 

shown that degradation of PEO to lower molecular weight might occur due to the 
high temperature used in the extruder. Extrusion to small pellets based on hydrophilic 
polymers hydroxyethyl cellulose (HEC) and HPMC has been performed, and 
isopropyl alcohol instead of water was used to avoid lump formation [12] . Also 
Carbopol [14 – 16] , pectin [114] , and xanthan [115, 116] have been extruded and used 
as matrix carrier materials for extended release. 
The choice of hydrophilic polymer is one determining factor for the drug release 
rate, as discussed above. The most common type used for hydrophilic matrix systems 
is HPMC (hypromellose, hydroxypropyl methyl cellulose) [24, 83] but some alternatives 
are HEC [117 – 119] , HPC [120, 121] , methyl cellulose [121] , xanthan [122 – 127] , 
PEO [68, 128] , Carbopol [129 – 131] , pectin [131 – 133] , and alginate [134 – 136] ; see 
Table 2 . HPMCs are available in several approved degrees of substitution [60] . To 
achieve different release durations, different viscosity grades of the polymers may 
be used (the higher the viscosity grade, the slower the release rate). If the desired 
viscosity is not available, one can mix two polymer grades with different viscosity 
grades [68] . 
6.8.5 COMPARISON AND SUMMARY OF DIFFERENT TECHNOLOGIES 
The main principles for oral extended drug release reviewed here are the 
membrane - coated reservoir systems and hydrophilic or hydrophobic matrix systems. 
They all have advantages and drawbacks and Table 3 summarizes some aspects of 
the different formulation principles. 
There are a number of commercial variants of the above - mentioned principles. 
Examples of commercial matrix - based formulations can be found in a review by 
Varma et al. [137] . One of the oldest commercial hydrophilic matrix systems is 
TABLE 2 FDA Registered Oral ER Formulations Containing Commonly Used 
Excipients in Hydrophilic Matrix Tablets 
Hydrophilic Polymer a 
Fraction of 
Excipient (%) 
Number of Hits on 
FDA Homepage 
Other Names 
of Excipients 
Hydroxypropyl methyl 
cellulose [24] 
10 – 80 12 Hypromellose, 
HPMC, 
MHPC 
Hydroxypropylcellulose 
[120, 121] 
15 – 35 4 HPC 
Polyethylene oxide [68, 128] 5 – 85 4 PEO 
Sodium alginate [134] 5 – 50 3 
Xanthan gum [125, 137 – 140] 10 – 99 3 
Hydroxyethyl cellulose 
[117 – 119] 
40 – 97.5 3 
Carbomer [129] 20 – 75 1 Carbopol, 
polyacrylic 
acid 
Methyl cellulose [121] 5 – 75 1 MC 
Source : http://www.accessdata.fda.gov/scripts/cder/iig/index.cfm . 
a Examples of references using them are given. 
COMPARISON AND SUMMARY OF DIFFERENT TECHNOLOGIES 1211

1212 ORAL EXTENDED-RELEASE FORMULATIONS 
TABLE 3 Comparison between Three Drug Release Principles 
Aspects 
Insoluble Matrix 
Systems 
Membrane - Coated 
Systems 
Hydrophilic Matrix 
Systems 
Drug solubility Dose dependent 
[18] 
Dose dependent 
[18] 
Whole range 
Release 
mechanism 
Diffusion controlled Diffusion and 
osmotic 
Diffusion to erosion 
controlled 
Release profi le Q . t0.5 Q . t1 Q . tn , 0.4 < n < 1 
Main release 
dependence 
(except drug 
properties) 
Channeling 
components and 
processes 
parameters 
Properties of 
membrane 
Properties of 
polymer carrier 
Composition 
alternatives 
Many approved and 
functional 
excipients 
available 
Pore - channeling 
excipients may 
determine drug 
release 
Small changes in 
formulation may 
change release 
rate 
Many approved 
and functional 
excipients 
available 
Release can be 
changed by fi lm 
and core 
compositions 
Many polymers are 
based on natural 
material which 
may give large 
batch - to - batch 
variation 
Many grades of 
available polymers 
exist 
Easy to regulate drug 
release by means 
of composition and 
polymer properties 
Many polymers are 
based on natural 
material, which 
may result in large 
batch - to - batch 
variation 
Manufacturing 
aspects 
Uses traditional 
production 
technologies and 
is inexpensive 
Release depends on 
process 
parameters 
Coating process can 
be used to 
control drug 
release 
Film coating 
process 
dominates 
release, which 
may be sensitive 
in relation to 
many different 
process 
parameters 
Release is mainly 
robust against 
process parameters 
Granulation with 
water is diffi cult 
In vivo – in 
vitro 
correlation 
Release depends on 
fasted and fed 
state 
Good Release depends on 
fasted and fed 
state 
TIMERx, which is based on xanthan gum mixed with, for example, locus bean gum 
[138] . A mixture of these polymers has a special property; the different polymers 
interact strongly with each other and this interaction produces gels with large viscosity. 
Variants of osmotically controlled ER formulations, some of which are commercially 
available, are reviewed by Verma et al. [52] . 

6.8.6 OTHER ORAL ER FORMULATIONS 
Here some new oral ER technologies based on principles other than those mentioned 
above will be presented. It is beyond the scope of this chapter to cover all 
systems and details thereof, and the interested reader is recommended to make use 
of the literature included in the references. 
One promising technology is to let the active drug interact with the excipients, 
for example, by covalent binding between the drug and excipient. The azoaromatic 
cross - linkers between the drug and excipient can exemplify this. These special cross - 
linkers break due to bacterial degradation in the colon, but not until they are 
exposed to this bacterial fl ora [139 – 141] . This means that the drug is hindered from 
release before it reaches the colon. This is an example of site - specifi c delivery to 
the colon, an area that is reviewed by a special issue of the journal Advanced Drug 
Delivery Reviews 2005 (volume 57, number 2). 
Another example of modifi cations of ER systems is when electrostatic interaction 
between charged drugs and excipients (with opposite charge) is used. This 
concept can be valuable for drugs with pH - dependent solubility. If, for example, 
the drug is a weak base, it will have a large solubility at low pH, and a major part 
of the drug may be rapidly released already in the stomach, which is not desirable. 
An introduction of polyions with opposite charge can result in electrostatic interaction 
between the drugs and the polyions, which can change the solubility or decrease 
the diffusion rate of the drug through the ER formulation. This concept is, for 
example, used when propranolol hydrochloride interacts with sodium carboxymethylcellulose 
(NaCMC) in HPMC matrices [142] . In addition, buffering of the ER 
systems by organic or inorganic buffers has been used to obtain pH - independent 
drug release [27, 143, 144] . A recent publication by Riis et al. showed that insoluble 
inorganic buffers such as magnesium hydroxide and magnesium oxide provided 
stable drug release over longer periods of time than when more soluble buffer 
systems were used [26] . 
Sophisticated systems based on responsive gels are promising alternatives in 
terms of oral extended release. The principle behind many of these systems is that 
the formulation should react and undergo some kind of transition due to a trigger 
signal [145] . In general, the triggers can be various factors such as temperature, light, 
pressure, electrosensitivity, or interactions with specifi c molecules such as glucose 
or antigens. For oral drug delivery systems, the triggers can also be a biological 
change in the gastrointestinal environment, such as a change in the bacteria fl ora, 
as mentioned earlier [139] . It can also be a physiochemical change, such as a pH 
change in the gastrointestinal tract. Hydrogels composed of copolymers of 
poly(acrylic acid) and covalently attached Pluronic surfactants is one such example 
that reacts on pH changes. The cross - linked microgels can be loaded with drug and 
tailored to collapse in low pH but swell and release the drug at high pH [146] . 
A novel method for producing ER formulations is a technology called three - 
dimensional printing (TheriForm technology) [147] , which is similar to the one used 
in ordinary printers. The ink is here replaced with an active substance and carrier 
material. The layer - by - layer “ printing ” provides controlled placement of the active 
drug and thus of the release from the device [148] . 
Even if there have been advances in oral drug delivery technologies during the 
last 50 years, many highly sophisticated drug delivery systems have failed and have 
OTHER ORAL ER FORMULATIONS 1213

1214 ORAL EXTENDED-RELEASE FORMULATIONS 
not reached the market [149] . This can be due to several reasons and in many cases 
the formulation is probably not the cause. However, on occasion one may be 
tempted to agree with Rocca and Park in their review of prospects and challenges 
in the oral drug delivery: “ breakthrough technologies are required to generate novel 
dosage forms raising drug delivery to higher level ” (p. 52) [149] . 
ACKNOWLEDGMENT 
Sven Engstr o m, Chalmers University of Technology, is acknowledged for his useful 
suggestions and comments. 
REFERENCES 
1. Malinowski , H. J. , and Marroum , P. J. ( 1999 ), Encyclopedia of Controlled Drug Delivery 
, vols. 1 and 2, Food and Drug Administration Requirements for Controlled Release 
Products , John Wiley & Sons , New York , vol. 1 , pp. 381 – 395 . 
2. Aulton , M. E. ( 2002 ), Pharmaceutics: The Science of Dosage Form Design , Churchill 
Livingstone , Hartcourt Publisher , Edinburgh . 
3. Marroum , P. J. ( 1997 ), Bioavialability/Bioequivalence for Oral Release Producs, Controlled 
Release Drug Delivery Systems. Paper presented at the 5th Int. Symp. Drug Del., 
East Brunswick, NJ, May 15 – 17. 
4. SCHOLAR, SciFinder ( 2006 ), American Chemical Society, Washington, DC. 
5. Yamanaka , K. , Yamamoto , H. , Kawamura , Y. , and Ito , S. ( 1954 ), Pyrimidine pencillin, 
U.S. Patent 2,681,339 . 
6. Robinson , M. J. , and Svedres , E. V. ( 1957 ), Sustained - release pharmaceutical preparations, 
U.S. Patent 2805977 . 
7. Sj o gren , J. , and Frykl o f , L - E. ( 1960 ), Duretter — A new type of oral sustained action 
preparation , Farmacevtisk Revy , 59 , 171 – 179 . 
8. Sj o gren , J. ( 1971 ), Studies on a sustained release principle based on inert plastic matrix , 
Acta Pharm. Suecica , 8 , 153 – 168 . 
9. Speers , M. , and Bonnano , C. ( 1999 ), Economic aspects of controlled drug delivery , in 
( Mathiowitz , E. , Ed.), Encyclopedia of Controlled Drug Delivery , Wiley , New York , 
pp. 341 – 347 . 
10. Cole , G. , Hogan , J. , and Aulton , M. , Eds. ( 1995 ), Pharmaceutical Coating Technology , 
Taylor & Francis , London , 1995. 
11. Ragnarsson , G. , and Johansson , M. O. ( 1988 ), Coated drug cores in multiple unit preparations. 
Infl uence of particle size , Drug Dev. Ind. Pharm. , 14 , 2285 – 2297 . 
12. Chatlapalli , R. , and Rohera , B. D. ( 1998 ), Physical characterization of HPMC and HEC 
and investigation of their use as pelletization aids . Int. J. Pharm. , 161 , 179 – 193 . 
13. Kojima , M. , and Nakagami , H. ( 2002 ), Development of controlled release matrix pellets 
by annealing with micronized water - insoluble or enteric polymers , J. Controlled Release , 
82 , 335 – 343 . 
14. Young , C. R. , Dietzsch , C. , Cerea , M. , Farrell , T. , Fegely , K. A. , Rajabi - Siahboomi , A. , 
and McGinity , J. W. ( 2005 ), Physicochemical characterization and mechanisms of 
release of theophylline from melt - extruded dosage forms based on a methacrylic acid 
copolymer , Int. J. Pharm. , 301 , 112 – 120 . 

15. Neau , S. H. , Chow , M. Y. , and Durrani , M. J. ( 1996 ), Fabrication and characterization 
of extruded and spheronized beads containing Carbopol 974P, NF resin , Int. J. Pharm. , 
131 , 47 – 55 . 
16. Bommareddy , G. S. , Paker - Leggs , S. , Saripella , K. K. , and Neau , S. H. ( 2006 ), Extruded 
and spheronized beads containing Carbopol 974P to deliver nonelectrolytes and salts 
of weakly basic drugs , Int. J. Pharm. , 321 , 62 – 71 . 
17. Abrahamsson , B. , Alpsten , M. , Jonsson , U. E. , Lundberg , P. J. , Sandberg , A. , Sundgren , 
M. , Svenheden , A. , and Toelli , J. ( 1996 ), Gastro - intestinal transit of a multiple - unit 
formulation (metoprolol CR/ZOK) and a non - disintegrating tablet with the emphasis 
on colon , Int. J. Pharm. , 140 , 229 – 235 . 
18. Thrombre , A. G. ( 2005 ), Assessment of feasibility of oral controlled release in an 
exploratory development setting , Drug Discov. Today , 10 , 1159 – 1166 . 
19. Li , S. , He , H. , Parthiban , L. J. , Yin , H. , and Serajuddin , A. T. M. ( 2005 ), IV - IVC considerations 
in the development of immediate - release oral dosage form , J. Pharm. Sci. , 
94 , 1396 – 1417 . 
20. Noyes , A. A. , and Whitney , W. R. ( 1897 ), The rate of solution of solid substances in 
their own solutions , J. Am. Chem. Soc. , 19 , 930 – 934 . 
21. Dokoumetzidis , A. , and Macheras , P. ( 2006 ), A century of dissolution research: From 
Noyes and Whitney to the Biopharmaceutics Classifi cation System , Int. J. Pharm. , 321 , 
1 – 11 . 
22. Mooney , K. G. , Mintun , M. A. , Himmelstein , K. J. , and Stella , V. J. ( 1981 ), Dissolution 
kinetics of carboxylic acids. I: Effect of pH under unbuffered conditions , J. Pharm. Sci. , 
70 , 13 – 22 . 
23. Mooney , K. G. , Mintun , M. A. , Himmelstein , K. J. , and Stella , V. J. ( 1981 ), Dissolution 
kinetics of carboxylic acids. II: Effects of buffers , J. Pharm. Sci. , 70 , 22 – 32 . 
24. Li , C. L. , Martini , L. G. , Ford , J. L. , and Roberts , M. ( 2005 ), The use of hypromellose 
in oral drug delivery , J. Pharm. Pharmacol. , 57 , 533 – 546 . 
25. Tatavarti , A. S. , and Hoag , S. W. ( 2006 ), Microenvironmental pH modulation based 
release enhancement of a weakly basic drug from hydrophilic matrices , J. Pharm. Sci. , 
95 , 1459 – 1468 . 
26. Riis , T. , Bauer - Brandl , A. , Wagner , T. , and Krantz , H. ( 2007 ), pH - independent drug 
release of an extremely poorly soluble weakly basic acidic drug from multiparticulate 
extended release formulations, Eur. J. Pharm. Biopharm. , 65 , 78 – 84 . 
27. Streubel , A. , Siepmann , J. , Dashevsky , A. , and Bodmeier , R. ( 2000 ), pH - independent 
release of a weakly basic drug from water - insoluble and - soluble matrix tablets , J. 
Controlled Release , 67 , 101 – 110 . 
28. U.S. Food and Drug Administration (FDA) ( 2006 ), available: http://www.fda.gov.search. 
html . 
29. Higuchi , T. ( 1963 ), Mechanism of sustained - action medication. Theoretical analysis of 
rate of release of solid drugs dispersed in solid matrices , J. Pharm. Sci. , 52 , 1145 – 
1149 . 
30. Wu , X. S. ( 1996 ), Controlled Drug Delivery Systems , Technology Publishing , Lancaster, 
PA . 
31. Jannin , V. , Pochard , E. , and Chambin , O. ( 2006 ), Infl uence of poloxamers on the dissolution 
performance and stability of controlled - release formulations containing Precirol 
ATO 5 , Int. J. Pharm. , 309 , 6 – 15 . 
32. Sinchaipanid , N. , Junyaprasert , V. , and Mitrevej , A. ( 2004 ), Application of hot - melt 
coating for controlled release of propranolol hydrochloride pellets , Powder Technol. , 
141 , 203 – 209 . 
REFERENCES 1215

1216 ORAL EXTENDED-RELEASE FORMULATIONS 
33. Rodriguez , L. , Caputo , O. , Cini , M. , Cavallari , C. , and Greechi , R. ( 1993 ), In vitro release 
of theophylline from directly - compressed matrixes containing methacrylic acid copolymers 
and/or dicalcium phosphate dihydrate , Farmaco , 48 , 1597 – 1604 . 
34. Ceballos , A. , Cirri , M. , Maestrelli , F. , Corti , G. , and Mura , P. ( 2005 ), Infl uence of 
formulation and process variables on in vitro release of theophylline from directly - 
compressed Eudragit matrix tablets , Farmaco , 60 , 913 – 918 . 
35. Oezyazici , M. , Goekce , E. H. , and Ertan , G. ( 2006 ), Release and diffusional modeling of 
metronidazole lipid matrices , Eur. J. Pharm. Biopharm. , 63 , 331 – 339 . 
36. Hamdani , J. , Moes , A. J. , and Amighi , K. ( 2003 ), Physical and thermal characterization 
of Precirol and Compritol as lipophilic glycerides used for the preparation of controlled - 
release matrix pellets , Int. J. Pharm. , 260 , 47 – 57 . 
37. U.S. Food and Drug Administration (FDA) (2006), available: http://www.accessdata.fda. 
gov/scripts/cder/iig/index.cfm . 
38. Mehnert , W. , and Mader , K. ( 2001 ), Solid lipid nanoparticles. Production, characterization 
and applications , Adv. Drug Deliv. Rev. , 47 , 165 – 196 . 
39. Savolainen , M. , Herder , J. , Khoo , C. , Lovqvist , K. , Dahlqvist , C. , Glad , H. , and Juppo , A. 
M. ( 2003 ), Evaluation of polar lipid - hydrophilic polymer microparticles , Int. J. Pharm. , 
262 , 47 – 62 . 
40. Zhu , Y. , Shah , N. H. , Malick , A. W. , Infeld , M. H. , and McGinity , J. W. ( 2006 ), Controlled 
release of a poorly water - soluble drug from hot - melt extrudates containing acrylic polymers 
, Drug Dev. Ind. Pharm. , 32 , 569 – 583 . 
41. Young , C. R. , Dietzsch , C. , and McGinity , J. W. ( 2005 ), Compression of controlled - release 
pellets produced by a hot - melt extrusion and spheronization process , Pharm. Dev. 
Technol. , 10 , 133 – 139 . 
42. Young , C. R. , Koleng , J. J. , and McGinity , J. W. ( 2002 ), Production of spherical pellets by 
a hot - melt extrusion and spheronization process , Int. J. Pharm. , 242 , 87 – 92 . 
43. Hogan , J. E. ( 1995 ), Modifi ed release coatings for pharmaceutics , in Cole , G. , Hogan , 
J. , and Aulton , M. , Eds., Pharmaceutical Coating Technology , pp. 409 – 438 . 
44. Edgar , K. J. , Buchanan , C. M. , Debenham , J. S. , Rundquist , P. A. , Seiler , B. D. , Shelton , 
M. C. , and Tindall , D. ( 2001 ), Advances in cellulose ester performance and application , 
Prog. Polym. Sci. , 26 , 1605 – 1688 . 
45. Hj a rtstam , J. ( 1998 ), Ethyl Cellulose Membranes Used in Modifi ed Release Formulations , 
Chalmers University of Technology , G o teborg, Sweden . 
46. Savastano , L. , Leuenberger , H. , and Merkle , H. P. (1995), Membrane modulated dissolution 
of oral drug delivery systems , Pharm. Acta Helv. , 70 , 117 – 124 . 
47. Theeuwes , F. ( 1975 ), Elementary osmotic pumps , J. Pharm. Sci. , 64 , 147 – 157 . 
48. Ragnarsson , G. , Sandberg , A. , Johansson , M. O. , Lindstedt , B. , and Sjoegren , J. ( 1992 ), 
In vitro release characteristics of a membrane - coated pellet formulation. Infl uence of 
drug solubility and particle size , Int. J. Ph+arm. , 79 , 223 – 232 . 
49. Ragnarsson , G. , Sandberg , A. , Jonsson , U. E. , and Sjoegren , J. ( 1987 ), Development of a 
new controlled release metoprolol product , Drug Dev. Ind. Pharm. , 13 , 1495 – 1509 . 
50. Kedem , O. , and Katchalsky , A. ( 1961 ), J. Gen. Phys , 45 , 143 . 
51. Kedem , O. , and Katchalsky , A. ( 1958 ), Biochem. Biophys. Acta , 27 , 229 . 
52. Verma , R. K. , Krishna , D. M. , and Garg , S. ( 2002 ), Formulation aspects in the development 
of osmotically controlled oral drug delivery systems , J. Controlled Release , 79 , 
7 – 27 . 
53. Elchidana , P. A. , and Deshpande , S. G. ( 1999 ), Microporous membrane drug delivery 
system for indomethacin , J. Controlled Release , 59 , 279 – 285 . 

54. Sousa, J. J. , Sousa, A. , Moura, M. J. , Podczeck, F. , and Newton, J. M. (2002), The infl uence 
of core materials and fi lm coating on the drug release from coated pellets , Int. J. Pharm. , 
233 , 111 – 122 . 
55. Husson , I. , Leclerc , B. , Spenlehauer , G. , Veillard , M. , and Couarraze , G. ( 1991 ), Modeling 
of drug release from pellets coated with an insoluble polymeric membrane , J. 
Controlled Release , 17 , 163 – 173 . 
56. Tang , E. S. K. , Chan , L. W. , and Heng , P. W. S. ( 2005 ), Coating of multiparticulates for 
sustained release , Am. J. Drug Deliv. , 3 , 17 – 28 . 
57. Verma , R. K. , Kaushal , A. M. , and Garg , S. ( 2003 ), Development and evaluation of 
extended release formulations of isosorbide mononitrate based on osmotic technology , 
Int. J. Pharm. , 263 , 9 – 24 . 
58. Ravishankar , H. , Patil , P. , Petereit , H - U. , and Renner , G. ( 2005 ), EUDRAMODE: A 
novel approach to sustained oral drug delivery systems , Drug Deliv. Technol. , 5 , 48 – 50 , 
52 – 55 . 
59. Ravishankar , H. , Iyer - Chavan , J. , Patil , P. , Samel , A. , and Renner , G. ( 2006 ), Clinical 
studies of terbutaline controlled - release formulation prepared using EUDRAMODE , 
Drug Deliv. Technol. , 6 , 50 – 56 . 
60. Kibbe , A. H. ( 2000 ), Handbook of Pharmaceutical Excipients , Pharmaceutical , 
London . 
61. Aulton , M. E. , and Twitchell , A. M. ( 1995 ), Solution properties and atomization in fi lm 
coating [of pharmaceuticals] , Pharm. Coat. Technol. , 64 – 117 . 
62. Glatt ( 2006 ), available: www.glatt.com . 
63. Harris , M. R. , and Ghebre - Sellassie , I. ( 1989 ), Aqueous polymeric coating for modifi ed - 
release pellets , Drugs Pharm. Sci. , 36 , 63 – 79 . 
64. Colombo , P. , Bettini , R. , Santi , P. , and Peppas , N. A. ( 2000 ), Swellable matrixes for controlled 
drug delivery: Gel - layer behavior, mechanisms and optimal performance , Pharm. 
Sci. Technol. Today , 3 , 198 – 204 . 
65. Lapidus , H. , and Lordi , N. G. ( 1966 ), Some factors affecting the release of a 
water - soluble drug from a compressed hydrophilic matrix , J. Pharm. Sci. , 55 , 840 – 
843 . 
66. Lapidus , H. , and Lordi , N. G. ( 1968 ), Drug release from compressed hydrophilic matrixes , 
J. Pharm. Sci. , 57 , 1292 – 1301 . 
67. Huber , H. E. , Dale , L. B. , and Christenson , G. L. ( 1966 ), Utilization of hydrophilic gums 
for the control of drug release from tablet formulations. I. Disintegration and dissolution 
behavior , J. Pharm. Sci. , 55 , 974 – 976 . 
68. Koerner , A. , Larsson , A. , Piculell , L. , and Wittgren , B. ( 2005 ), Tuning the polymer release 
from hydrophilic matrix tablets by mixing short and long matrix polymers , 
J. Pharm. Sci. , 94 , 759 – 769 . 
69. Ju , R. T. C. , Nixon , P. R. , and Patel , M. V. ( 1995 ), Drug release from hydrophilic 
matrixes 1. New scaling laws for predicting polymer and drug release based on the 
polymer disentanglement concentration and the diffusion layer , J. Pharm. Sci. , 84 , 
1455 – 1463 . 
70. Ju , R. T. C. , Nixon , P. R. , Patel , M. V. , and Tong , D. M. ( 1995 ), Drug release from hydrophilic 
matrixes. 2. A mathematical model based on the polymer disentanglement concentration 
and the diffusion layer , J. Pharm. Sci. , 84 , 1464 – 1477 . 
71. Bettini , R. , Colombo , P. , Massimo , G. , Catellani , P. L. , and Vitali , T. ( 1994 ), Swelling and 
drug release in hydrogel matrixes: Polymer viscosity and matrix porosity effects , Eur. J. 
Pharm. Sci. , 2 , 213 – 219 . 
REFERENCES 1217

1218 ORAL EXTENDED-RELEASE FORMULATIONS 
72. Adler , J. , Jayan , A. , and Melia , C. D. ( 1999 ), A method for quantifying differential expansion 
within hydrating hydrophilic matrixes by tracking embedded fl uorescent microspheres 
, J. Pharm. Sci. , 88 , 371 – 377 . 
73. Treloar , L. R. G. ( 1975 ), The Physics of Rubber Elasticity , Clarendon , Oxford . 
74. Siepmann , J. , Kranz , H. , Bodmeier , R. , and Peppas , N. A. ( 1999 ), HPMC - matrices for 
controlled drug delivery: A new model combining diffusion, swelling, and dissolution 
mechanisms and predicting the release kinetics , Pharm. Res. , 16 , 1748 – 1756 . 
75. Colombo , P. , Bettini , R. , Massimo , G. , Catellani , P. L. , Santi , P. , and Peppas , N. A. ( 1995 ), 
Drug diffusion front movement is important in drug release control from swellable 
matrix tablets , J. Pharm. Sci. , 84 , 991 – 997 . 
76. Bettini , R. , Catellani , P. L. , Santi , P. , Massimo , G. , Peppas , N. A. , and Colombo , P. ( 2001 ), 
Translocation of drug particles in HPMC matrix gel layer: Effect of drug solubility and 
infl uence on release rate , J. Controlled Release , 70 , 383 – 391 . 
77. Fu , X. C. , Liang , W. Q. , and Ma , X. W. ( 2003 ), Relationships between the release of 
soluble drugs from HPMC matrices and the physicochemical properties of drugs , 
Pharmazie , 58 , 221 – 222 . 
78. Tahara , K. , Yamamoto , K. , and Nishihata , T. ( 1996 ), Application of model - independent 
and model analysis for the investigation of effect of drug solubility on its release rate 
from hydroxypropyl methyl cellulose sustained - release tablets , Int. J. Pharm. , 133 , 
17 – 27 . 
79. Korsmeyer , R. W. , Gurny , R. , Doelker , E. , Buri , P. , and Peppas , N. A. ( 1983 ), 
Mechanisms of solute release from porous hydrophilic polymers , Int. J. Pharm. , 15 , 
25 – 35 . 
80. Rinaki , E. , Valsami , G. , and Macheras , P. ( 2003 ), The power law can describe the “ entire ” 
drug release curve from HPMC - based matrix tablets: A hypothesis , Int. J. Pharm. , 255 , 
199 – 207 . 
81. Ritger , P. L. , and Peppas , N. A. ( 1987 ), A simple equation for description of solute release. 
II. Fickian and anomalous release from swellable devices , J. Controlled Release , 5 , 
37 – 42 . 
82. Ritger , P. L. , and Peppas , N. A. ( 1987 ), A simple equation for description of solute release. 
I. Fickian and non - Fickian release from non - swellable devices in the form of slabs, 
spheres, cylinders or disks , J. Controlled Release , 5 , 23 – 36 . 
83. Siepmann , J. , and Peppas , N. A. ( 2001 ), Modeling of drug release from delivery systems 
based on hydroxypropyl methylcellulose (HPMC) , Adv. Drug Deliv. Rev. , 48 , 139 – 
157 . 
84. Peppas , N. A. , and Sahlin , J. J. ( 1989 ), A simple equation for the description of solute 
release. III. Coupling of diffusion and relaxation , Int. J. Pharm. , 57 , 169 – 72 . 
85. Gao , P. , and Meury , R. H. ( 1996 ), Swelling of hydroxypropyl methylcellulose matrix 
tablets. 1. Characterization of swelling using a novel optical imaging method , J. Pharm. 
Sci. , 85 , 725 – 731 . 
86. Mitchell , K. , Ford , J. L. , Armstrong , D. J. , Elliott , P. N. C. , Hogan , J. E. , and Rostron , C. 
( 1993 ), The infl uence of substitution type on the performance of methyl cellulose and 
hydroxypropyl methyl cellulose in gels and matrixes , Int. J. Pharm. , 100 , 143 – 
154 . 
87. Levina , M. , and Rajabi - Siahboomi , A. R. ( 2004 ), The infl uence of excipients on drug 
release from hydroxypropyl methylcellulose matrices , J. Pharm. Sci. , 93 , 2746 – 2754 . 
88. Ford , J. L. , Rubinstein , M. H. , and Hogan , J. E. ( 1985 ), Propranolol hydrochloride and 
aminophylline release from matrix tablets containing hydroxypropyl methyl cellulose , 
Int. J. Pharm. , 24 , 339 – 350 . 

89. Ford , J. L. , Rubinstein , M. H. , and Hogan , J. E. ( 1985 ), Formulation of sustained - release 
promethazine hydrochloride tablets using hydroxypropyl methyl cellulose matrixes , Int. 
J. Pharm. , 24 , 327 – 338 . 
90. Reynolds , T. D. , Mitchell , S. A. , and Balwinski , K. M. ( 2002 ), Investigation of the effect 
of tablet surface area/volume on drug release from hydroxypropyl methylcellulose 
controlled - release matrix tablets , Drug Dev. Ind. Pharm. , 28 , 457 – 466 . 
91. Skoug , J. W. , Borin, M. T. , Fleishaker , J. C. , and Cooper , A. M. (1991), In vitro and in vivo 
evaluation of whole and half tablets of sustained - release adinazolam mesylate , Pharm. 
Res. , 8 , 1482 – 1488 . 
92. Sung , K. C. , Nixon , P. R. , Skoug , J. W. , Ju , T. R. , Gao , P. , Topp , E. M. , and Patel , M. V. 
( 1996 ), Effect of formulation variables on drug and polymer release from HPMC - based 
matrix tablets , Int. J. Pharm. , 142 , 53 – 60 . 
93. Ford , J. L. , Rubinstein , M. H. , McCaul , F. , Hogan , J. E. , and Edgar , P. J. ( 1987 ), Importance 
of drug type, tablet shape and added diluents on drug release kinetics from hydroxypropyl 
methyl cellulose matrix tablets , Int. J. Pharm. , 40 , 223 – 234 . 
94. Heng , P. W. , Chan , L. W. , Easterbrook , M. G. , and Li , X. ( 2001 ), Investigation of the 
infl uence of mean HPMC particle size and number of polymer particles on the release 
of aspirin from swellable hydrophilic matrix tablets , J. Controlled Release , 76 , 39 – 49 . 
95. Velasco , M. V. , and Ford , J. L. , Rowe , P. , and Rajabi - Siahhoomi , A. R. ( 1999 ), Infl uence 
of drug: Hydroxypropyl methyl cellulose ratio, drug and polymer particle size and compression 
force on the release of diclofenac sodium from HPMC tablets , J. Controlled 
Release , 57 , 75 – 85 . 
96. Dabbagh , M. A. , Ford , J. L. , Rubinstein , M. H. , and Hogan , J. E. ( 1996 ), Effects of 
polymer particle size, compaction pressure and hydrophilic polymers on drug release 
from matrixes containing ethyl cellulose , Int. J. Pharm. , 140 , 85 – 95 . 
97. Mitchell , K. , Ford , J. L. , Armstrong , D. J. , Elliott , P. N. C. , Hogan , J. E. , and 
Rostron , C. ( 1993 ), The infl uence of the particle size of hydroxypropyl methyl 
cellulose K15M on its hydration and performance in matrix tablets , Int. J. Pharm. , 100 , 
175 – 179 . 
98. Sheskey , P. J. , Cabelka , T. D. , Robb , R. T. , and Boyce , B. M. ( 1994 ), Use of roller compaction 
in the preparation of controlled - release hydrophilic matrix tablets containing 
methyl cellulose and hydroxypropyl methyl cellulose polymers , Pharm. Technol. , 18 , 132 , 
134 , 136 , 138 , 140 , 142 , 144 , 146 , 148 – 150 . 
99. Sheskey , P. J. , and Hendren , J. ( 1999 ), The effects of roll compaction equipment variables, 
granulation technique, and HPMC polymer level on a controlled - release matrix model 
drug formulation , Pharm. Technol. , 23 , 90 , 92 , 94 , 96 , 98 , 100 , 102 , 104 , 106 . 
100. Sheskey , P. , Pacholke , K. , Sackett , G. , Maher , L. , and Polli , J. ( 2000 ), Effect of process 
scale - up on robustness of tablets, tablet stability, and predicted in vivo performance , 
Pharm. Technol. , 24 , 30 , 32 , 34 , 36 , 38 , 40 , 42 , 44 , 46 , 48 , 50 , 52 . 
101. Timmins , P. , Delargy , A. M. , Howard , J. R. , and Rowlands , E. A. ( 1991 ), Evaluation of 
the granulation of a hydrophilic matrix sustained - release tablet , Drug Dev. Ind. Pharm. , 
17 , 531 – 550 . 
102. Liu , C. H. , Chen , S. C. , Kao , Y. H. , Kao , C. C. , Sokoloski , T. D. , and Sheu , M. T. ( 1993 ), 
Properties of hydroxypropyl methyl cellulose granules produced by water spraying , Int. 
J. Pharm. , 100 , 241 – 248 . 
103. Herder , J. , Adolfsson , A. , and Larsson , A. ( 2006 ), Initial studies of water granulation of 
eight grades of hypromellose (HPMC) , Int. J. Pharm. , 313 , 57 – 65 . 
104. Dahl , T. C. , and Bormeth , A. P. ( 1990 ), Naproxen controlled release matrix tablets: Fluid 
bed granulation feasibility , Drug Dev. Ind. Pharm. , 16 , 581 – 590 . 
REFERENCES 1219

1220 ORAL EXTENDED-RELEASE FORMULATIONS 
105. Nellore , R. V. , Rekhi , G. S. , Hussain , A. S. , Tillman , L. G. , and Augsburger , L. L. ( 1998 ), 
Development of metoprolol tartrate extended - release matrix tablet formulations for 
regulatory policy consideration , J. Controlled Release , 50 , 247 – 256 . 
106. Liu , C. H. , Kao , Y. H. , Chen , S. C. , Sokoloski , T. D. , and Sheu , M. T. ( 1995 ), In - vitro and 
in - vivo studies of the diclofenac sodium controlled - release matrix tablets , J. Pharm. 
Pharmacol. , 47 , 360 – 364 . 
107. Dahl , T. C. , Calderwood , T. , Bormeth , A. , Trimble , K. , and Piepmeier , E. ( 1990 ), Infl uence 
of physicochemical properties of hydroxypropyl methyl cellulose on naproxen release 
from sustained release matrix tablets , J. Controlled Release , 14 , 1 – 10 . 
108. Shah , N. H. , Railkar , A. S. , Phuapradit , W. , Zeng , F. W. , Chen , A. , Infeld , M. H. , and 
Malick , A. W. ( 1996 ), Effect of processing techniques in controlling the release rate and 
mechanical strength of hydroxypropyl methyl cellulose based hydrogel matrixes , Eur. J. 
Pharm. Biopharm. , 42 , 183 – 187 . 
109. Sheskey , P. J. , and Keary , C. M. ( 2002 ), Aqueous air foams , U.S. Patent 7011702. 
110. Keary , C. M. , and Sheskey , P. J. ( 2004 ), Preliminary report of the discovery of a new 
pharmaceutical granulation process using foamed aqueous binders , Drug Dev. Ind. 
Pharm. , 30 , 831 – 845 . 
111. Cao , Q - R. , Choi , Y - W. , Cui , J - H. , and Lee , B - J. ( 2005 ), Effect of solvents on physical 
properties and release characteristics of monolithic hydroxypropylmethylcellulose 
matrix granules and tablets , Arch. Pharm. Res. , 28 , 493 – 501 . 
112. Sheskey , P. J. , and Williams , D. M. ( 1996 ), Comparison of low - shear and high - shear wet 
granulation techniques and the infl uence of percent water addition in the preparation 
of a controlled - release matrix tablet containing HPMC and a high - dose, highly water - 
soluble drug , Pharm. Technol. , 20 , 80 , 82 , 84 , 86 , 88 , 90 , 92 . 
113. Zhang , F. , and McGinity , J. W. ( 1999 ), Properties of sustained - release tablets prepared 
by hot - melt extrusion , Pharm. Dev. Technol. , 4 , 241 – 250 . 
114. Urbano , A. P. A. , Ribeiro , A. J. , and Veiga , F. ( 2006 ), Design of pectin beads for oral 
protein delivery , Chem. Ind. Chem. Eng. Q. , 12 , 24 – 30 . 
115. Fukuda , M. , Peppas , N. A. , and McGinity , J. W. ( 2006 ), Properties of sustained release 
hot - melt extruded tablets containing chitosan and xanthan gum , Int. J. Pharm. , 310 , 
90 – 100 . 
116. De Brabander , C. , Vervaet , C. , and Remon , J. P. ( 2003 ), Development and evaluation of 
sustained release mini - matrices prepared via hot melt extrusion , J. Controlled Release , 
89 , 235 – 247 . 
117. Baumgartner , S. , Slamersek , V. , and Kristl , J. ( 2003 ), Controlled drug delivery of 
hydrophilic drugs from cellulose ether matrix tablets: The infl uence of the drug molecule 
size on its release mechanism and kinetics , Farm. Vestnik (Ljubljana, Slov.) , 54 , 
359 – 360 . 
118. Genc , L. , Bilac , H. , and Guler , E. ( 1999 ), Studies on controlled release dimenhydrinate 
from matrix tablet formulations , Pharm. Acta Helv. , 74 , 43 – 49 . 
119. Sinha Roy , D. , and Rohera Bhagwan , D. ( 2002 ), Comparative evaluation of rate of 
hydration and matrix erosion of HEC and HPC and study of drug release from their 
matrices , Eur. J. Pharm. Sci. , 16 , 193 – 199 . 
120. Vueba , M. L. , Batista de Carvalho , L. A. E. , Veiga , F. , Sousa , J. J. , and Pina , M. E. ( 2006 ), 
Infl uence of cellulose ether mixtures on ibuprofen release: MC25, HPC and HPMC 
K100M , Pharm. Dev. Technol. , 11 , 213 – 228 . 
121. Vueba , M. L. , Batista De Carvalho , L. A. E. , Veiga , F. , Sousa , J. J. , and Pina , M. E. ( 2004 ), 
Infl uence of cellulose ether polymers on ketoprofen release from hydrophilic matrix 
tablets , Eur. J. Pharm. Biopharm. , 58 , 51 – 59 . 

122. Talukdar , M. M. , Michoel , A. , Rombaut , P. , and Kinget , R. ( 1996 ), Comparative study 
on xanthan gum and hydroxypropyl methyl cellulose as matrixes for controlled - release 
drug delivery I. Compaction and in vitro drug release behavior , Int. J. Pharm. , 129 , 
233 – 241 . 
123. Talukdar , M. M. , and Kinget , R. ( 1997 ), Comparative study on xanthan gum and 
hydroxypropyl methyl cellulose as matrixes for controlled - release drug delivery. II. 
Drug diffusion in hydrated matrixes , Int. J. Pharm. , 151 , 99 – 107 . 
124. Andreopoulos , A. G. , and Tarantili , P. A. ( 2002 ), Study of biopolymers as carriers for 
controlled release , J. Macromol. Sci. Phys. , B41 , 559 – 578 . 
125. Andreopoulos , A. G. , and Tarantili , P. A. ( 2001 ), Xanthan gum as a carrier for controlled 
release of drugs , J. Biomater. Appl. , 16 , 34 – 46 . 
126. Talukdar , M. M. , Rombaut , P. , and Kinget , R. ( 1998 ), The release mechanism of an oral 
controlled - release delivery system for indomethacin , Pharm. Dev. Technol. , 3 , 1 – 6 . 
127. Talukdar , M. M. , and Kinget , R. ( 1995 ), Swelling and drug release behavior of xanthan 
gum matrix tablets , Int. J. Pharm. , 120 , 63 – 72 . 
128. Maggi , L. , Segale , L. , Torre , M. L. , Ochoa Machiste , E. , and Conte , U. ( 2002 ), Dissolution 
behaviour of hydrophilic matrix tablets containing two different polyethylene 
oxides (PEOs) for the controlled release of a water - soluble drug. Dimensionality study , 
Biomaterials , 23 , 1113 – 1119 . 
129. Choulis , N. , and Papadopoulos , H. ( 1975 ), Timed - release tablets containing quinine 
sulfate , J. Pharm. Sci. , 64 , 1033 – 1035 . 
130. Huang , L - L. , and Schwartz , J. B. ( 1995 ), Studies on drug release from a carbomer tablet 
matrix , Drug Dev. Ind. Pharm. , 21 , 1487 – 1501 . 
131. El - Sayed , G. M. , El - Said , Y. , Meshali , M. M. , and Schwartz , J. B. ( 1996 ), Kinetics of 
theophylline release from different tablet matrixes , STP Pharma Sci. , 6 , 390 – 397 . 
132. Patel , G. N. , Patel , G. C. , Patel , R. B. , Patel , S. S. , Patel , J. K. , Bharadia , P. D. , and 
Patel , M. M. ( 2006 ), Oral colon - specifi c drug delivery: An overview , Drug Deliv. 
Technol. , 6 , 62 – 71 . 
133. Liu , L. S. , Fishman , M. L. , Kost , J. , and Hicks , K. B. ( 2003 ), Pectin - based systems for 
colon - specifi c drug delivery via oral route , Biomaterials , 24 , 3333 – 3343 . 
134. Liew , C. V. , Chan , L. W. , Ching , A. L. , and Heng , P. W. S. ( 2006 ), Evaluation of sodium 
alginate as drug release modifi er in matrix tablets , Int. J. Pharm. , 309 , 25 – 37 . 
135. Shilpa , A. , Agrawal , S. S. , and Ray , A. R. ( 2003 ), Controlled delivery of drugs from 
alginate matrix , J. Macromol. Sci. Polym. Rev. , C43 , 187 – 221 . 
136. Ostberg , T. , and Graffner , C. ( 1994 ), Calcium alginate matrixes for oral multiple unit 
administration: III. Infl uence of calcium concentration, amount of drug added and 
alginate characteristics on drug release , Int. J. Pharm. , 111 , 271 – 282 . 
137. Varma , M. V. S. , Kaushal , A. M. , Garg , A. , and Garg , S. ( 2004 ), Factors affecting 
mechanism and kinetics of drug release from matrix - based oral controlled drug delivery 
systems , Am. J. Drug Deliv. , 2 , 43 – 57 . 
138. McCall , T. W. , Baichwal , A. R. , and Staniforth , J. N. ( 2003 ), TIMERx oral controlled - 
release drug delivery system , Drugs Pharm. Sci. , 126 , 11 – 19 . 
139. Friend , D. R. ( 2005 ), New oral delivery systems for treatment of infl ammatory bowel 
disease , Adv. Drug Deliv. Rev. , 57 , 247 – 265 . 
140. Ghandehari , H. , Kopeckova , P. , and Kopecek , J. ( 1997 ), In vitro degradation of pH - 
sensitive hydrogels containing aromatic azo bonds , Biomaterials , 18 , 861 – 872 . 
141. Akala , E. O. , Kopeckova , P. , and Kopecek , J. ( 1998 ), Novel pH - sensitive hydrogels with 
adjustable swelling kinetics , Biomaterials , 19 , 1037 – 1047 . 
REFERENCES 1221

1222 ORAL EXTENDED-RELEASE FORMULATIONS 
142. Takka , S. , Rajbhandari , S. , and Sakr , A. ( 2001 ), Effect of anionic polymers on the 
release of propranolol hydrochloride from matrix tablets , Eur. J. Pharm. Biopharm. , 52 , 
75 – 82 . 
143. Nie , S. , Pan , W. , Li , X. , and Wu , X. ( 2004 ), The effect of citric acid added to hydroxypropyl 
methylcellulose (HPMC) matrix tablets on the release profi le of vinpocetine , 
Drug Dev. Ind. Pharm. , 30 , 627 – 635 . 
144. Varma, M. V. S. , Kaushal, A. M. , and Garg , S. (2005), Infl uence of micro - environmental 
pH on the gel layer behavior and release of a basic drug from various hydrophilic 
matrices , J. Controlled Release , 103 , 499 – 510 . 
145. Qiu , Y. , and Park , K. ( 2001 ), Environment - sensitive hydrogels for drug delivery , Adv. 
Drug Deliv. Rev. , 53 , 321 – 339 . 
146. Bromberg , L. ( 2005 ), Intelligent hydrogels for the oral delivery of chemotherapeutics , 
Exp. Opin. Drug Deliv. , 2 , 1003 – 1013 . 
147. Rowe , C. W. , Wang , C - C. , and Monkhouse , D. C. ( 2003 ), TheriForm technology , Drugs 
Pharm. Sci. , 126 , 77 – 87 . 
148. Lee , K - J. , Kang , A. , Delfi no , J. J. , West , T. G. , Chetty , D. , Monkhouse , D. C. , and Yoo , 
J. ( 2003 ), Evaluation of critical formulation factors in the development of a rapidly dispersing 
captopril oral dosage form , Drug Dev. Ind. Pharm. , 29 , 967 – 979 . 
149. Rocca , J. G. , and Park , K. ( 2004 ), Oral drug delivery: Prospects & challenges , Drug 
Deliv. Technol. , 4 , 52 – 54 , 57 . 

ROLE OF NANOTECHNOLOGY 
SECTION 7


1225 
7.1 
CYCLODEXTRIN - BASED 
NANOMATERIALS IN 
PHARMACEUTICAL FIELD 
Erem Bilensoy and A. Atilla Hincal 
Hacettepe University Faculty of Pharmacy, Ankara, Turkey 
Contents 
7.1.1 Introduction 
7.1.1.1 Cyclodextrins in Pharmaceutical Field 
7.1.2 Application of Cyclodextrins in Nanoparticles 
7.1.2.1 Incorporation of Drug – Cyclodextrin Complexes in Nanoparticulate Delivery 
Systems 
7.1.2.2 Preparation of Nanoparticles from Cyclodextrins 
7.1.2.3 Effi cacy and Safety of Amphiphilic Cyclodextrin Nanoparticles 
7.1.3 Conclusion 
References 
7.1.1 INTRODUCTION 
Cyclodextrins (CDs) have a wide range of application in the pharmaceutical fi eld 
due to their unique structure, which allows them to include hydrophobic molecules 
in their apolar cavity and to mask the physicochemical properties of the included 
molecule. This results in the enhancement of drug bioavailability by improving 
aqueous solubility and the physical and chemical stability of the drug, masking 
undesired side effects such as irritation, taste, or odor, and overcoming compatibility 
problems or interactions between drugs and excipients. 
Parallel to the increasing interest and successful licensing and commercialization 
of nanoparticulate pharmaceutical products, CDs have also been incorporated 
into nanoparticulate drug delivery systems for several purposes. This can be 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

1226 CYCLODEXTRIN-BASED NANOMATERIALS IN PHARMACEUTICAL FIELD 
achieved by two approaches: (1) complexation of active ingredient with an appropriate 
CD and entrapment into polymeric nanoparticles to solve problems arising 
from the drug ’ s physicochemical properties or (2) modifi cation of CDs to render 
an amphiphilic character to these molecules, which allows CDs to self - align into 
nanoparticles in the form of nanospheres, nanocapsules, solid lipid nanoparticles, 
nanosize liposomes, and nanosize vesicles with or without the presence of surface - 
active agents. 
In light of current research, this chapter will deal with the following issues 
concerning the use of CDs and derivatives as nanomaterials for drug delivery: 
use of CDs (natural and synthetic) derivatives in the pharmaceutical fi eld and 
application of CDs in nanoparticulate drug delivery systems. A major part of the 
chapter will be focused on new CD derivatives, amphiphilic CDs, and the characterization, 
effi cacy, and safety of nanoparticles prepared from amphiphilic 
CDs. 
7.1.1.1 Cyclodextrins in Pharmaceutical Field 
Natural Cyclodextrins Cyclodextrins are cyclic oligosaccharides obtained by the 
enzymatic degradation of starch. Major natural CDs are crystalline, homogeneous, 
nonhygroscopic substances which have a toruslike macroring shape built up from 
glucopyranose units, as seen in Figure 1 [1 – 3] . Cyclodextrins are named depending 
on the number of glucopyranose units. Major industrially produced CDs are named 
as follows; . - CD, possessing six units, . - CD, possessing seven units; and . - CD, possessing 
eight units. 
Natural CDs have been demonstrated to have a special structure; that is, glucose 
residues in the CD ring possess the thermodynamically favored 4 C 1 chair conformation 
because all substituent groups are in equatorial position. Cyclodextrins behave 
like rigid compounds with two degrees of freedom: rotation at the glucosidic links 
C(4) – O(4) and C(1) – O(4) and rotations at the O(6) primary hydroxyl groups at the 
C(5) – C(6) band. As a consequence of this chair conformation, all secondary hydroxyl 
groups at C(2) and C(3) are located at the broader side of the CD torus in the 
equatorial position. Hydroxyl groups on C(2) point toward the cavity and hydroxyl 
groups on C(3) point outward. The primary hydroxyl groups at the C6 position are 
located at the narrower side of the torus. These hydroxyl groups ensure good water 
solubility for the natural CDs. The cavity of the torus is lined with a ring of C . H 
groups (C3), a ring of glucosidic oxygen atoms, and another ring of C . H groups 
(C5). Thus, the cavity of CDs exhibits an apolar character. This is accompanied by 
a high electron density and Lewis base property. The physicochemical characteristics 
and inclusion behavior of CDs are a direct consequence of these special binding 
conditions [4, 5] . Certain physicochemical characteristics of natural CDs are given 
in Table 1 . 
Cyclodextrin Derivatives Natural CDs were reported to form total or partial 
inclusion complexes with several drugs to improve the aqueous solubility and stability 
under physiological or ambient conditions, reduce or mask completely the side 
effects associated with the included drug, and increase compatibility of the drug 
with other drugs in the formulation or excipients while improving patient compliance 
by masking the taste or odor of the active ingredient [6 – 8] . 

FIGURE 1 Schematic representation of natural CD structure and modifi cation sites. 
HOCH2 
HOCH2 
CH2OH 
CH2OH 
CH2OH 
CH2OH CH2OH 
O 
O 
O 
O 
O 
O O 
O 
O 
O 
O
O 
O 
O 
HO 
HO 
HO 
HO 
HO 
HO 
OH 
HO 
OH 
OH 
OH 
OH 
OH
OH 
6 
2 
3 
OH (6) 
OH (3) OH (2) 
Primary face 
Secondary face 
Apolar cavity 
Modification 
sites 
INTRODUCTION 1227 
TABLE 1 Some Physicochemical Characteristics of Natural Cyclodextrins 
Characteristics . - CD . - CD . - CD 
Number of glucose units 6 7 8 
Molecular weight, g/mol 972 1135 1297 
Internal diameter, A 4.7 – 5.2 6.0 – 6.4 7.5 – 8.3 
External diameter, A 14.2 – 15.0 15.0 – 15.8 17.1 – 17.9 
Depth, A 7.9 – 8.0 7.9 – 8.0 7.9 – 8.0 
Solubility in water (25 ° C), g/L 145 18.5 232 
Crystal water, w % 10.2 13.2 – 14.5 8.13 – 17.7 
Approximative cavity volume in 1 mol 
CD, A 3 
174 262 472 
Melting point, ° C 250 – 260 255 – 265 240 – 245 
Half - life in 1 M HCl at 60 ° C, h 6.2 5.4 3.0 
Crystal forms (from water) Hexagonal 
plate 
Monoclinic 
parallelogram 
Quadratic 
prism 
Partial molar volumes in solution, mL/mol 611.4 703.8 801.2 

1228 CYCLODEXTRIN-BASED NANOMATERIALS IN PHARMACEUTICAL FIELD 
In contrast to the advantageous nature of CDs for molecular inclusion, their 
surface makes it more diffi cult for the highly hydrophilic CD molecule to interact 
with lipophilic biological membranes. For this reason, natural CDs have been chemically 
modifi ed to alter their water solubility, interaction with biological membranes, 
and drug release properties. 
Two of the natural CDs are known to be parenterally unsafe due to nephrotoxic 
effects [9] . The etiology of the nephrotoxicity of . - and . - CD is unknown but is 
believed to be related to either CD uptake by kidney tubule cells resulting in disruption 
of intracellular function or the extraction of lipid membrane components 
by the CDs. The latter is suggested to be of validity since there seems to be a linear 
correlation between the ability of some CDs to disrupt cellular membranes and 
kidney nephrotoxicity [2, 6] . The ability of CDs to cause red blood cell hemolysis 
and membrane irritation seems also to correlate with their ability to extract lipid 
membrane components: cholesterol and phospholipids [10, 11] . 
Modifi cation of natural CDs has been the aim of many research groups to 
improve safety while maintaining the ability to form inclusion complexes with 
various substrates. Some groups have also focused on improving the interaction 
between the pharmaceuticals and the CDs while others have attempted to prepare 
materials that can be chemically defi ned more precisely. 
Methylated CDs are obtained by methylation of CDs on either all C2 secondary 
and C6 primary hydroxyl groups [dimethyl cyclodextrins (DIMEB)] or all the 
hydroxyl groups C2, C3, and C6 [trimethyl cyclodextrins (TRIMEB)]. Major disadvantage 
of methylated CDs is that their solubility decreases with increasing temperature 
and they are reported to be hemolytic [12] . This is a result of partial 
methylation of the hydroxyls of . - CD which leads to stronger drug binding but also 
to stronger hemolysis [12] . 
Hydroxypropylated CDs are statistically substituted derivatives because hydroxypropylation 
does not result in selective substitution as with methylation. While the 
reaction proceeds, the reactivity of the hydroxyl group changes, and this results in 
a mixture of products with various degrees of substitution. Their dissolution is 
endothermic so there is no decrease in solubility with increasing temperature 
[6, 13] . It is necessary to note that degree of substitution in hydroxypropylated CDs 
is inversely correlated with their inclusion capability [14, 15] . Hydroxyalkylated CDs 
are commercially available as tablets, ocular collyrs [16] , and excipients under the 
trademarks Encapsin and Molecusol. 
Sulfobutylether - . - cyclodextrins (SBE - . - CDs) are water soluble and parenterally 
safe. An advantage over hydroxypropylated - . - CDs (HP - . - CDs) is that higher sulfobutyl 
group substitution often results in higher drug complexation ability [17] . 
Inability of the SBE - . - CDs, especially the commercially available product (SBE) 7M - 
. - CD (Captisol), to form strong complexes with cholesterol and other membrane 
lipids, arising from their polyanionic nature causing Coulombic repulsions, results 
in a little or no membrane disruption [6, 13] and reduced hemolysis [18] . Captisol 
is used in parenteral and ocular systems as well as osmotic tablets and also as a 
freeze - drying excipient [19] . 
Branched CDs (mono - or di - glucosyl, maltosyl and glucopyranosyl . - and . - 
CDs) are more resistant to . - amylase than natural CDs. Natural . - CD and monoglucosyl 
-. - CD are stable in rat blood because they have no linear glycosidic bond. 
Branched CDs exert a lower hemolytic activity on human erythrocytes and are 

APPLICATIONS OF CYCLODEXTRINS IN NANOPARTICLES 1229 
weaker than natural CDs. Their inclusion capability is more or less drug dependent. 
Steroids were reported to show a slightly higher affi nity to branched CDs than to 
natural CDs [19] . 
The water solubility of acylated CDs decreases proportionally to their degree of 
substitution, whereas their solubility in less polar solvents such as ethanol increases. 
Acylated CDs of hydrophobic nature are useful for controlling the release rates of 
water - soluble drugs [20] . 
Ionizable . - CDs possess interesting solubility properties, too. Solubility in water 
for these derivatives depends on the pH of the solution. Carboxymethylethyl - . - 
cyclodextrin (CME - . - CD) is prepared as an enteric - type carrier system. The presence 
of a carboxymethyl group causes a pH - dependent solubility range in water, 
meaning that it is only slightly soluble in the low - pH region, but freely soluble in 
neutral and alkaline regions due to the ionization of the carboxyl group (p Ka about 
3 – 4) [21] . Inclusion - forming capability of CME - . - CD is dependent on drug properties, 
including size, shape, and charge of the molecule. 
Sulfated CDs are of anionic nature and are interesting from chemical and biological 
points of view because of their angiogenic and antiviral properties [22, 23] . Sulfated 
derivatives are also reported to have a heparin - like activity, resulting in 
increase in blood - clotting times, which limits their injectable dose [7] . 
Low - molecular - weight CD polymers (MW 3000 – 6000 Da) are soluble in water 
whereas high - molecular - weight CD polymers (MW > 10,000 Da) can only swell in 
water and form insoluble gels [24, 25] . Insoluble cross - linked bead polymers seem 
to be applicable as wound - healing agents for the treatment of wounds like burns or 
ulcers. Iodine has been complexed with such a CD polymer as an antiseptic wound 
healing agent [24] . 
7.1.2 APPLICATIONS OF CYCLODEXTRINS IN NANOPARTICLES 
7.1.2.1 Incorporation of Drug – Cyclodextrin Complexes in 
Nanoparticulate Delivery Systems 
Nanoparticles are of pharmaceutical interest due to their active and passive targeting 
properties and their ability to deliver poorly solube drugs and drugs with stability 
problems. Nanoparticles are considered more stable than liposomal delivery 
systems. However, a major drawback is associated with the drug - loading capacity 
of polymeric nanoparticles. Classical emulsion polymerization procedures result in 
considerably low drug - loading capacities. This results in the administration of excessive 
quantities of polymeric material which may impair the safety of the drug delivery 
system [26, 27] . 
For this reason, different techniques are used to improve the drug - loading 
properties of polymeric nanoparticles. Cyclodextrins are used for this reason to 
improve water solubility and sometimes the hydrolytic or photolytic stability of 
drugs for better loading properties [8] . Drug – CD complexes act to solubilize or stabilize 
active ingredients within the nanoparticles, resulting in increased drug concentration 
in the polymerization medium and increased hydrophobic sites in the 
nanosphere structure when large amounts of CDs are associated to the nanoparticles 
[27, 28] . 

1230 CYCLODEXTRIN-BASED NANOMATERIALS IN PHARMACEUTICAL FIELD 
The antiviral agent saguinavir was complexed to HP - . - CD to increase saquinavir 
loading into polyalkylcyanoacrylate nanoparticles by providing a soluble drug reservoir 
in the polymerization medium that is the basis of nanoparticle formation [29] . 
A dynamic equilibrium was observed between the complex, dissociated species of 
the complex, and the forming polymeric nanoparticle. During nanoparticle formation, 
free drug was believed to be progressively incorporated into a polymer network, 
driven by the drug partition coeffi cient between the polymer and polymerization 
medium. Simultaneous direct entrapment of some CD – drug complex was also suspected 
[28 – 30] . 
Incorporation of the steroidal drugs hydrocortisone and progesterone in complex 
with . - CD and HP - . - CD reduced the particle size for solid lipid nanoparticles 
(SLNs) below 100 nm. Steroids were demonstrated to be dispersed in the amorphous 
state. Compexation to CDs resulted in higher drug - loading properties for the 
more hydrophobic drug hydrocortisone and lower in vitro release for both drugs 
when they are complexed to CDs rather than their free form [31] . 
The in vivo behavior of nanoparticles obtained from drug – CD complexes was 
also evaluated. HP - . - CD addition in the polymerization medium of polyethylcyanoacrylate 
(PECA) nanospheres improved the subcutaneous absorption of metoclopramide 
in rats. PECA nanospheres with HP - . - CD provided the highest drug 
concentration and enhanced drug absorption compared with those with dextran or 
with drug solution. However, in addition to drug absorption from subcutaneous 
sites, HP - . - CD also enhanced the drug elimination by enhancing the drug absorption 
to reticuloendothelial tissues [32] . 
Progesterone complexed to HP - . - CD or DM - . - CD was loaded into bovine 
serum albumin (BSA) nanospheres. Dissolution rates of progesterone were signifi - 
cantly enhanced by complexation to CDs with respect to free drug. Nanospheres of 
100 nm loaded with drug – CD complexes provided a pH - dependent release profi le 
and good stability in an aqueous neutral environment [33] . 
In another approach, CD properties of complexation were combined with those 
of chitosan. Complexation with CD was believed to permit solubilization as well as 
protection for labile drugs while entrapment in the chitosan network was expected 
to facilitate absorption. Chitosan nanoparticles, including complexes of HP - . - CD 
with the hydrophobic model drugs triclosan and furosemide, were prepared by ionic 
cross - linking of chitosan with sodium tripolyphosphate (TPP) in the presence of 
CDs. Nanoparticles were then prepared by ionotropic gelation using the obtained 
drug – HP -. - CD inclusion complexes and chitosan. Cyclodextrin and TPP concentration 
largely affected particle size but the zeta potential remained unchanged with 
different parameters. On the other hand, drug entrapment increased up to 4 and 10 
times by triclosan and furosemide, respectively. The release profi le of nanoparticles 
indicated an initial burst release followed by a delayed release profi le lasting up to 
4 h [34] . 
Recently a CD – insulin complex was encapsulated in polymethacrylic acid – chitosan 
– polyether[polyethylene glycol (PEG) – propylene glycol] copolymer PMCP 
nanoparticles from the free - radical polymerization of methacrylic acid in the presence 
of chitosan and polyether in a medium free of solvents or surfactants. Particles 
had a size distribution of 500 – 800 nm. The HP - . - CD inclusion complex with insulin 
was encapsulated into the nanoparticles, resulting in a pH - dependent release profi le 
as seen in Figure 2 . The biological activity of insulin was demonstrated with enzyme -

APPLICATIONS OF CYCLODEXTRINS IN NANOPARTICLES 1231 
linked immunosorbent assay (ELISA). Cyclodextrin complexed to insulin encapsulated 
into mucoadhesive nanoparticles was believed to be a promising candidate for 
oral insulin delivery [35] . 
7.1.2.2 Preparation of Nanoparticles from Cyclodextrins 
Cyclodextrins are used as excipients in the preparation of nanoparticles by three 
approaches: 
1. Preparing nanoparticles from polymers under the presence of CDs in the 
medium 
2. Preparing nanoparticles from polymers incorporating or conjugated to CDs 
3. Preparing nanoparticles directly from amphiphilic CDs 
Nanoparticles consisting copolymers of aminoethylcarbamoyl - . - cyclodextrin 
(AEC - . - CD) and ethylene glycol diglycidyl ether (EGDGE) were prepared by an 
interfacial polyaddition reaction in a mini – emulsion system. By combining these 
two technologies, namely, cross - linking and modifi cation of hydroxyl groups, a novel 
functional nanoparticle based on . - CD was introduced as a novel material of nanobiotechnology 
[36] . 
Nanoparticles prepared fromn CDs are promising targeted delivery systems. 
Transferrin, an iron - binding glycoprotein ligand for tumor targeting, was conjugated 
to . - CD polymers and adamantane – PEG5000 through carbohydrate groups self - 
assembled into sub - 100 - nm particles in a recent study [37] . A CD - containing polyca- 
FIGURE 2 pH - dependent release profi le for insulin complexed to HP - . - CD and encapsulated 
in nanoparticles. ( Reprinted from S. Sajeesh and C. P. Sharma, International Journal of 
Pharmaceutics , 325, 147 – 154, 2006, Copyright 2006, with permission from Elsevier. ) 
100 
90 
80 
70 
60 
50 
40 
30 
20 
10
0 
0 50 100 150 200 250 300 350 
Time (min) 
Percentage of release 
HPCDI pH7.4 
HPCDI pH1.2 
I pH7.4 
I pH1.2

1232 CYCLODEXTRIN-BASED NANOMATERIALS IN PHARMACEUTICAL FIELD 
tion was used for nucleic acid condensation into nanoparticles [38] and the second 
component, adamantane - terminated modifi er for stabilizing nanoparticles to minimize 
interactions with plasma and to target cell surface receptors, was incorporated 
in this system. The particles were demonstrated to mediate transferrin - mediated 
delivery of nucleic acids to cultured cells [37] . 
Transferrin - containing CD polymer - based nanoparticles were studied as nucleic 
acid delivery system that can be modifi ed for targeted delivery of small interfering 
ribonucleic acid (siRNA) to cancer cells. Molecular studies showed that the siRNA 
CD nanoparticles reduced levels of Ewing ’ s transcript by 80% and inhibited growth 
of cultured Ewing ’ s tumor cell line. It was also reported that this delivery system 
indicated a lack of toxicity [39] . 
A new tadpole - shaped polymer was synthesized via a coupling reaction of PLA 
onto mono[6 - (2 - aminoethyl)amino - 6 - deoxy] - . - cyclodextrin (CDenPLA). A hydrophilic 
head consisting of the CD group was believed to bind proteins and the PLA 
tail gave the amphiphilic property [40] . BSA was incorporated into nanoparticles 
of CDenPLA using both nanoprecipitation and double - emulsion techniques, as can 
be seen in Figure 3 [40] . A similar process was used to couple PLGA onto amino - 
. - CD and ethylenediamino - bridged bis( . - CD) to afford amphiphilic conjugates 
forming nanoparticles with the nanoprecipitation technique. This approach was 
believed to be promising for protein delivery since BSA structure was unchanged 
FIGURE 3 TEM photographs of BSA - loaded . CDen47PLA nanoparticles prepared 
with different techniques and their magnifi ed images. ( Reprinted from H. Gao, Y. W. Yang, 
Y. G. Fan, and J. B., Ma, Journal of Controlled Release , 112, 301 – 311, 2006. Copyright 2006, 
with permission from Elsevier. ) 
0.5 mm 100 nm 
0.5 mm 100 nm 
DE1 DE2 
NP1 NP2

APPLICATIONS OF CYCLODEXTRINS IN NANOPARTICLES 1233 
after encapsulation into nanoparticles and during its release. Nanoparticles were 
reported to be stable after freeze drying [40, 41] . 
Nanoassemblies were formed by mixing solutions of . - CD polymer and dextran 
hydrophopbically modifi ed with alkyl chains (C12) and loaded with the model 
hydrophobic drug benzophenone. Nanoassemblies were characterized as 110 – 190 nm 
with relatively low drug - loading values ranging between 0.3 to 1.01% w/w. Authors 
suggested that hydrophobic model drug and hydrophobically modifi ed dextran 
compete for the apolar CD cavity; however, benzophenone does not impede the 
hydrophobic dextran to interact with . - CD polymer to form supramolecular assemblies 
at the nanoscale [42] . 
Another group has worked on the oligo(ethylenediamino) - . - cyclodextrin modi- 
fi ed gold nanoparticles (OEA - CD - NP) as a vector for DNA binding. Possessing 
many hydrophobic cavities at the outer space, OEACD - NP was believed to have a 
capability of carrying biological and/or medicinal substrates into cells. Presence of 
the CD moieties was suggested to be the key parameter in the high affi nity to DNA 
for the gold nanoparticles. In addition, CD moieties were demonstrated to reduce 
the cytotoxicity of gold nanoparticles arising from the gold clusters that impair 
plasma membrane functions and lead to cell death [43] . 
Nucleic acid delivery was also studies by Park et al. using CD - based nanoparticles 
prepared from . - CD - modifi ed poly(ethylenimine) (CD - PEI). The inclusion - forming 
capability of . - CD was used in order to immobilize the nanoparticles on solid surfaces 
(adamantine - modifi ed self - assembled monolayers). CD - PEI nanoparticles 
were proposed as delivery systems onto solid surfaces to attain specifi c and high 
affi nity loading. The interaction is schematized in Figure 4 [44] . 
FIGURE 4 Schematic representation of . - CD – adamantane inclusion complex formation 
and immobilization of CD nanoparticles on adamantine - modifi ed surface. ( Reprinted with 
permission from ref. 44 . Copyright 2006 by the American Chemical Society. ) 
+ 
= 
O O O 
O 
O
O
O O 
O 
O 
O
O 
O 
O 
OH 
OH 
OH 
OH 
OH 
HO 
OH 
HO 
HO 
HO
HO 
HO 
HO 
HO 
HO HO 
OH 
OH
OH OH
OH 
Scheme 1A 
Adamantane .-cyclodextrin Inclusion complex 
CD-PEI/DNA 
complex CD-PEI 
nanoparticle 
Specific 
binding with 
high affinity AD-modified 
surface 
50-.m gold layer 
Glass slide

1234 CYCLODEXTRIN-BASED NANOMATERIALS IN PHARMACEUTICAL FIELD 
Amphiphilic Cyclodextrins Nanoparticles have been obtained spontaneously 
from modifi ed CDs of amphiphilic structure since the last decade. This approach 
differs from the previously discussed approaches in that amphiphilic CDs do not 
require the presence of another polymer or macromolecule or even surfactants. 
Amphiphilic CDs have been synthesized to solve problems of natural CDs that 
limit their pharmaceutical applications. The main reasons for the synthesis of amphiphilic 
CDs are as follows: 
1. Enhancement of interaction of CDs with biological membranes through a 
relative external hydrophobicity 
2. Modifi cation or enhancement of interaction of CDs with hydrophobic drugs 
arising from the high number of long aliphatic chains and by increasing 
the number of hydrophobic sites for possible interactions with hydrophobic 
molecules 
3. Allowing self - assembly of CDs resulting in the spontaneous formation of 
nanosize carriers in the form of nanospheres and nanocapsules 
The unique advantage of amphiphilic CDs is that they possess self - assembling properties 
that are suffi cient to form nanoparticles spontaneously without the presence 
of a surfactant as well as the capability of including hydrophobic molecules in their 
cavity and within the long aliphatic chains [45 – 47] . Amphiphilic CDs can be classi- 
fi ed according to their surface charge. 
Nonionic Amphiphilic Cyclodextrins Nonionic amphiphilic CDs are obtained by 
grafting aliphatic chains of different length on the primary and/or secondary face 
of the CD glucopyranose unit. Different derivatives depicted in Figure 5 are named 
after their structure: 
1. Lollipop CDs [48] are obtained by grafting only one aliphatic chain to 
6 - amino - . - CD. 
2. Cup - and - ball CDs were synthesized by the introduction of a voluminous group 
such as the tert - butyl group which is linked to the end of the aliphatic chain 
in order to prevent self - inclusion of the pendant group [49, 50] . 
FIGURE 5 Schematic representation of some nonionic amphiphilic CDs. 
Lollipop 
CD 
Cup-and-ball 
CD 
Medusa-like CD 
Skirt-shaped CD 
Bouquet-shaped 
CD

APPLICATIONS OF CYCLODEXTRINS IN NANOPARTICLES 1235 
3. Medusa - like CDs are obtained by grafting aliphatic chains with length between 
C10 and C16 to all the primary hydroxyls of the CD molecule [51 – 53] . 
4. Skirt - shaped CDs consist of . - and . - CDs per - modifi ed with aliphatic esters 
(C2 – C14) on the secondary face [54 – 57] . 
5. Bouquet - shaped CDs result from the grafting of 14 polymethylene chains to 
3 - monomethylated . - cyclodextrin, meaning seven chains on each side of the 
CD ring molecule [58] . Per(2,6 - di - O - alkyl) CDs where the alkyl chain may be 
propyl, butyl, pentyl, 3 - methylbutyl, or dodecyl also take part in the bouquet 
family [4] . 
6. Cholesteryl CDs were recently introduced as more complicated derivatives 
[59] . They have been designed assuming that CD is the hydrophilic head group 
and cholesterol is the hydrophobic part. 
Interfacial properties of nonionic amphiphilic CDs have been demonstrated by 
different groups [60 – 62] . It was found that length, structure, and bond type of the 
aliphatic chain play important roles upon the surface - active characteristics of amphiphilic 
CDs. Alignment of the amphiphilic CD molecule at the air – water interface 
was demonstrated to be aliphatic chains perpendicular to and a CD ring parallel 
to the fi lm [62] . Inclusion - forming capability of nonionic amphiphilic CDs also has 
been reported with various model molecules of a different nature. It was suggested 
that leaving the wider side of the cavity, that is, the secondary face, unsubstituted 
may facilitate entrance of the drug in the cavity of the amphiphilic CD [63 – 65] . 
Cationic Amphiphilic Cyclodextrins Recently cationic amphiphilic CDs were 
obtained and characterized carrying an amino group as an ionic group. Heptakis(2 - 
. - amino - O - oligo(ethyleneoxide) - 6 - hexylthio) - . - CD, a “ stealth ” cationic amphiphilic 
CD because of the oligoethylene glycol group it carries, was synthesized [66] . The 
structural properties of cationic amphiphilic CDs were believed to be due to the 
balance between hydrophobic tails such as thioalkyl chains and hydrophilic components 
such as ethylene glycol oligomers. The presence of ethylene glycol chains in 
particular was believed to increase the colloidal stability of the supramolecular 
aggregates formed by cationic amphiphilic CDs. Amphiphilic alkylamino - . - and 
. - CDs were also reported regarding their synthesis and characterization [67] . 
A series of polyamino - . - CDs have been synthesized by Cryan et al. [68] and 
complete substitution by amine groups at the 6 - position. Neutral CDs have been 
shown to interact with nucleic acids and nucleotides and to enhance their transfection 
effi ciency in vivo. Cationic CDs have shown even greater ability to bind nucleotides 
and enhance delivery by viral vectors. The major advantage of polycationic 
CDs and their nanoparticles is their enhanced ability to interact with nucleic acids 
combined with their self - organizational properties [68] . 
Anionic Amphiphilic Cyclodextrins Anionic amphiphilic CDs possess a sulfate 
group in their structure to render an anionic nature to the molecule. An effi cient 
regiospecifi c synthetic route to obtain acyl - sulfated . - CDs was introduced in which 
the upper rim is functionalized with sulfates and the lower rim with fatty acid esters 
[69] . These derivatives were able to form aggregates in aqueous medium. 
Sulfated amphiphilic . - , . - , and . - CDs were demonstrated to form 1 : 1 inclusion 
complexes with the antiviral drug acyclovir. Noncovalent interactions between 

1236 CYCLODEXTRIN-BASED NANOMATERIALS IN PHARMACEUTICAL FIELD 
acyclovir and nonsulfated amphiphilic CDs (nonionic amphiphilic CDs) appeared 
to take place both in the cavity of the CD and inside the hydrophobic zone generated 
by alkanoyl chains. However, in the case of sulfated anionic amphiphilic CDs, 
the interactions appear to take place only in the hydrophobic region of the alkanoyl 
chains [70] . 
Fluorine containing anionic . - CDs were fi rst introduced by Granger et al. [71] 
functionalized at the 6 - position by trifl uoromethylthio groups. They exhibit an 
amphiphilic behavior at the air – water interface and are good candidates for a new 
class of amphiphilic carriers. P e roche et al. [72] described the synthesis of new 
amphiphilic perfl uorohexyl - and perfl uorooctyl - thio - . - CDs and their alkyl analogue, 
nonanethio - . - CD. The ability of these products to form nanoparticles was 
also investigated by photon correlation spectroscopy and imaging techniques such 
as scanning electron microscopy (SEM) and cryo – transmission electron microscopy 
(TEM). 
Fluorophilic CD derivatives have been obtained as a result of combinations 
of CDs and a linear perfl uorocarbon [73] . 2,3 - Di - O - decafl uorooctanoyl - . - CD was 
obtained with a protection – deprotection synthetic method and characterized further 
by thin - layer chromatography (TLC), Fourier transform infrared (FTIR) spectroscopy, 
differential scanning calorimetry (DSC), elemental analysis, and time - of - fl ight 
mass spectrometry (OF - MS). 
7.1.2.3 Safety and Effi cacy of Amphiphilic Cyclodextrin Nanoparticles 
Amphiphilic CDs yield nanoparticles spontaneously in the form of nanospheres 
or nanocapsules depending on the preparation technique. Nanoparticles have been 
manufactured using three different techniques. However, the nanoprecipitation 
technique is generally preferred since it is a simple technique resulting in unimodal 
distribution. The general preparation techniques for amphiphilic CD nanoparticles 
are as follows: 
1. Nanoprecipitation [74 – 76] 
2. Emulsion/solvent evaporation [77] 
3. Detergent removal [78] 
Nanocapsules are also prepared according to the same techniques. Amphiphilic CD 
and the oil Miglyol or benzyl benzoate are dissolved in suitable organic solvent 
(acetone, ethanol). The solution is poured into aqueous phase under constant stirring 
and the nanocapsules form spontaneously. Organic solvent is then evaporated. 
Resulting nanocapsules vary in size between 100 and 900 nm according to the preparation 
process and technological parameters [79] . 
Particle sizes of nanocapsules are mostly affected by the size of the oil droplet 
formed during the preparation along with the molar concentration and nature of 
amphiphilic CD. Nanospheres, on the other hand, are not signifi cantly affected 
by amphiphilic CD concentration and can be formed with very high concentrations 
of amphiphilic CDs. The modifi cation site of the CD (primary or secondary face) is 
infl uential for nanosphere size since modifi cations on the secondary face result in a 
larger surface area. The presence and concentration of a surfactant such as Pluronic 
F68 do not affect the particle size of nanospheres and nanocapsules [80] . Nano

APPLICATIONS OF CYCLODEXTRINS IN NANOPARTICLES 1237 
spheres and nanocapsules of amphiphilic CDs were imaged with different microscopic 
techniques such as cryo - TEM, atomic force microscopy (AFM), and scanning 
transmission microscopy (STM). SEM imaging, on the other hand, results in shrinkage 
or disruption of the nanoparticles due to electron bombardment. Figures 6 a 
and 6 b present TEM photomicrographs after freeze fracture for 6 - O - CAPRO - . - CD 
nanospheres and nanocapsules, respectively [62, 66] . 
Drug loading into amphiphilic nanospheres and nanocapsules is governed by the 
loading technique used. Amphiphilic CD nanoparticles can be loaded with the following 
techniques: 
1. Conventional Loading Drug solution is added to the organic phase during 
preparation. 
2. Preloading Nanoparticles are prepared directly from preformed drug – 
amphiphilic CD complexes. 
3. High Loading Nanoparticles are prepared directly from preformed drug – 
amphiphilic CD complexes and further loaded by the addition of drug solution 
in the organic phase. 
A high - loading technique results in two - to threefold increase in drug entrapment. 
Other factors infl uencing drug loading to amphiphilic CD nanospheres are 
related to drug physicochemical properties such as drug – CD association constant 
k 1:1 , representing the affi nity of the drug to the CD cavity, oil/water partition coeffi - 
cient, and aqueous solubility. The affi nity of the drug to the CD cavity is correlated 
with drug - loading capacity. Lipophilic drugs interact both with the CD cavity and 
the long aliphatic chains situated on either the primary or the secondary face. 
Drug release properties of amphiphilic CD nanospheres are affected by various 
parameters, including drug lipophilicity, drug – CD association constant, and loading 
technique with release profi les varying from 2 to 96 h depending on the above 
parameters. Nanocapsules, on the other hand, exert somewhat different drug release 
profi les that are mostly dependent on lipophilicity and aqueous solubility of the 
drug. Lipophilicity of the drug is inversely correlated with the rate of release, as 
seen in Figure 7 [81] . Nevertheless, preparing nanoparticles directly from preformed 
FIGURE 6 Cryo - TEM images of 6 - O - CAPRO - . - CD nanospheres ( a ) and nanocapsules 
( b ). [ ( a ) Reprinted from E. Memisoglu, A. Bochot, M. en, D. Duchene, and A. A. Hincal, 
International Journal of Pharmaceutics , 252, 143 – 153, 2003. Copyright 2003 with permission 
from Elsevier .) 
(a) (b) 
S

1238 CYCLODEXTRIN-BASED NANOMATERIALS IN PHARMACEUTICAL FIELD 
inclusion complexes helped reduce the initial burst effect observed in general for 
nanospheres due to their very large surface area. 
Cancer Therapy Nanoparticles were fi rst prepared with the concept of targeting 
colloidal carriers of nanosize to tumor tissues via the leaky vasculature in tumor 
regions. Since then nanoparticulate drug carriers have been associated with cancer 
therapy through passive and active targeting to cancer cells. Thus, amphiphilic CD 
nanoparticles were mainly focused on cancer therapy and its different aspects. 
Tamoxifen, an antiestrogen drug used for the fi rst - line and adjuvant therapy for 
metastatic breast cancer as long - term chemotherapy, has been incorporated into 
amphiphilic CD nanoparticles prepared using the amphiphilic CD, . - CDC6 seen in 
Figure 8 in order to reduce the severe side effects associated with the nonselective 
cytotoxicity of this drug. Tamoxifen citrate – loaded nanospheres and nanocapsules 
with approximately 65% entrapment effi ciency liberated the drug with a controlled - 
release profi le up to 6 h when the high - loading technique is used [82] . Anticancer 
effi cacy of tamoxifen citrate – loaded nanospheres and nanocapsules was demonstrated 
to be equivalent to tamoxifen citrate solution in ethanol against MCF - 7 
human breast cancer cells. Transcription effi ciency of the tamoxifen citrate nanocapsules 
and nanospheres was evaluated against MELN cells in the presence of 17 - 
. - estradiol (E2) for the inhibition of E2 - mediated luciferase gene expression. It was 
found that transcription effi ciency of tamoxifen citrate – loaded nanospheres and 
nanocapsules were concentration dependent [83] . 
Paclitaxel, an anticancer drug with bioavailability problems arising from its very 
low aqueous solubility, its tendency to recrystallize when diluted in aqueous media, 
FIGURE 7 In vitro release profi les of steroids with different physicochemical properties 
from . - CDC6 nanocapsules (HCR HL, hydrocortisone high loaded, HCR CL, hydrocortisone 
conventionally loaded; TST HL, testosterone high loaded; TST CL, testosterone conventionally 
loaded; PRO HL, progesterone high loaded; PRO CL, progesterone conventionally 
loaded). 
Cumulative drug (% released from 
Time (h) 
0 
20 
40 
60 
80 
100 
120 
0 2 4 6 8 12 14 16 18 20 22 24 10 
nanocapsules) 
HCR CL HCR HL TST CL TST HL PRO CL PRO HL

APPLICATIONS OF CYCLODEXTRINS IN NANOPARTICLES 1239 
and solubilizers used in its commercially available injectable formulations, has been 
loaded into nanoparticles prepared from amphiphilic . - CD modifi ed on the primary 
face with 6C aliphatic esters, 6 - O - CAPRO - . - CD, seen in Figure 8 . Paclitaxel - loaded 
6 - O - CAPRO - . - CD nanospheres and nanocapsules were characterized with a diameter 
of 150 nm for nanospheres and 500 nm for nanocapsules with high entrapment 
effi ciencies. Blank nanoparticles were proven to be physically stable in aqueous 
dispersion for 12 months. The in vitro release of paclitaxel from nanoparticles was 
completed in 24 h [84] . Amphiphilic . - CD nanoparticles were compared to the commercial 
vehicle Cremophor EL in terms of hemolysis and cytotoxicity. 6 - O - CAPRO - 
. - CD nanospheres in particular were found to be signifi cantly less hemolytic than 
paclitaxel solution in the Cremophor vehicle on human erythrocytes. Cytotoxic 
effects of blank nanoparticles were assessed against L929 mouse fi broblast cells and 
a vast difference in cytotoxicity of up to 100 - fold reduction was observed for amphiphilic 
CD nanoparticles. 
Drug - loaded nanoparticles were also evaluated for their safety and effi cacy. 
Paclitaxel - encapsulated 6 - O - CAPRO - . - CD nanospheres and nanocapsules were 
evaluated for their physical stability in a one - month period in aqueous dispersion 
form with repeated particle size and zeta potential measurements and AFM imaging 
to evaluate recrystallization in aqueous medium. Paclitaxel - loaded amphiphilic CD 
nanoparticles were found to be physically stable for a period of one month whereas 
recrystallization occurs within minutes when diluted for intravenous (IV) infusion 
[85] . Finally, paclitaxel - loaded amphiphilic nanoparticles were demonstrated to 
show similar anticancer effi cacy against MCF - 7 cells when compared to paclitaxel 
solution in a cremophor vehicle [85] . 
Our group is currently working on the formulation of another potent anticancer 
drug, camptothecin, that is clinically inactive due to its very low water solubility and 
poor stability under physiological pH, which causes the drug to be converted from 
its active lactone form to its inactive carboxylate form. Two different amphiphilic 
FIGURE 8 Amphiphilic . - CD derivatives modifi ed with 6C aliphatic esters on ( a ) secondary 
face, . - CDC6 and ( b ) primary face, 6 - O - CAPRO - . -CD. 
O 
6 
2 
3 
OH 
O 
O 
OH 
O 
C 
C O 
O 
7 
O 
6 
2 
3 
OH 
OH 
OH 
O 
O 
C O 
7 
(a) (b)

1240 CYCLODEXTRIN-BASED NANOMATERIALS IN PHARMACEUTICAL FIELD 
. - CD nanospheres, . - CDC6 and 6 - O - CAPRO - . - CD, have succeeded in maintaining 
camptothecin in its active lactone form with considerable loading values and 
release profi les prolonged up to 96 h [86, 87] . 
Cationic amphiphilic CDs, heptakis[2 - . - amino - O - oligo(ethylene oxide] hexylthio 
-. - CD nanoparticles, have encapsulated anionic porphyrins (TPPS) by entangling 
these molecules within the aliphatic chains aligning both faces of the cationic 
amphiphilic CD. These nanoparticles were demonstrated to preserve the photodynamic 
properties of the entrapped photoactive agent. The photodynamic effi cacy 
of the carrier/sensitizer nanoparticles was proven by in vitro studies on tumor HeLa 
cells showing signifi cant cell death upon illumination with visible light [88] . 
Oxygen Delivery Amphiphilic and fl uorophilic . - CD derivatives perfl uoro - . - CDs 
were used to prepare nanocapsules with a single - step nanoprecipitation technique. 
Highly fl uorinated materials have multiple properties, such as repellance to water 
and oil, unique dielectric, rheological, and optical properties, as well as exceptional 
chemical and biological inertness. The fl uorinated chains, due to their strong hydrophobic 
and fl urorophilic character, impart unique properties to the vesicles, 
including enhanced particle size stability, prolonged intravascular persistence, and 
increased drug encapsulation capability. Thus, 2,3 - di - O - decafl uorooctanoyl - . - CD 
nanoparticles were believed to be a suitable carrier for oxygen solubilization and 
delivery. Oxygen delivery of perfl uorinated amphiphilic CD nanocapsules was compared 
to water and showed a prolonged delivery of oxygen. Fluorophilic nanocapsules 
were believed to overcome fl uorocarbon emulsions as oxygen carriers due to 
their higher number of particles in the colloidal solution which will permit a greater 
rate of dissolved oxygen [73] . 
Oral Delivery Amphiphilic . - CD nanocapsules loaded with indomethacin have 
been evaluated in vivo. The nanocapsules have been applied to the rat model. It was 
reported that the gastrointestinal mucosa of the rat was signifi cantly protected from 
the ulcerogenic effects of the active ingredient indomethacin in free form. Drug 
encapsulation yield in the nanocapsules were > 98% and the drug content per CD 
unit was 7.5% w/w [89] . 
Cytotoxicity The cytotoxicity of nanocapsules was investigated against L929 
mouse fi broblast cells and human polymorphonuclear PMNC cells with MTT assay 
[90] . Cell viability values of different nanocapsule and nanosphere formulations on 
L929 and PMNC cells indicated that nonsurfactant . - CDC6 nanocapsules were less 
cytotoxic than nanocapsules containing surfactants. The cytotoxicity of the nanoparticles 
mostly arises from surfactant presence and was concentration dependent 
[90] . 
Nanospheres of . - CDC6 prepared without surfactant and with Pluronic F68 of 
varying concentrations between 0.1 and 1% were found to be slightly less cytotoxic 
than nanocapsules to both L929 and human PMNC cells. It was concluded that 
cytotoxicity increased with increasing concentration of surfactant and the most suitable 
percentage for surfactant if required was found to be 0.1% [80] . 
Sterilizability Three different sterilization techniques — autoclaving, fi ltration, 
and gamma sterilization — were evaluated for amphiphilic CD nanoparticles of 

. - CDC6 loaded with the model drug tamoxifen [90] . It was found that fi ltration was 
not suitable for injectable amphiphilic CD nanoparticles since nanoparticle sizes 
were too close to fi lter pore sizes of 0.22 . m. Autoclaving did not affect the nanoparticle 
yield but caused a signifi cant increase in particle size and aggregates. Gamma 
irradiation realized with a dose of 25 kGy was demonstrated to be a suitable sterilization 
technique since no signifi cant change was observed for mean diameter, zeta 
potential, drug entrapment effi ciency, and in vitro release profi les for nimodipine - 
loaded . - CDC6 nanospheres and nanocapsules . The in vitro release profi le of sterile 
and nonsterile nanospheres and nanocapsules of . - CDC6 loaded with nimodipine 
is seen in Figure 9 [90] . 
7.1.3 CONCLUSION 
Cyclodextrins have been involved in nanoparticulate drug delivery systems by 
increasing the solubility of the drug via complex formation, forming nanoparticles 
in the presence of another polymer/macromolecule, forming nanoparticles by conjugation 
to polymers, or modifi cation of natural CDs to render this molecule an 
amphiphilic character. This chapter mainly focused on the potential of amphiphilic 
CDs as promising carriers for anticancer drugs with bioavailability problems, oxygen 
delivery for the treatment of ischemia, or the safe oral administration of drugs with 
gastrointestinal side effects. 
FIGURE 9 In vitro release profi le of tamoxifen from . - CDC6 nanospheres and nanocapsules 
before and after gamma sterilization. ( Reprinted from E. Memisoglu - Bilensoy and 
A. A. Hincal, International Journal of Pharmaceutics , 311, 203 – 208, 2006. Copyright 2006 with 
permission from Elsevier. ) 
0 
10 
20 
30 
40 
50 
60 
70 
80 
90 
100
0 1 2 3 4 5 6 
Time (h) 
Cumulative TMX citrate (% released) 
Nonsterile NC Sterile NC Nonsterile NS Sterile NS 
CONCLUSION 1241

1242 CYCLODEXTRIN-BASED NANOMATERIALS IN PHARMACEUTICAL FIELD 
Many new studies are available to modify amphiphilic CDs further by giving 
them “ stealth ” properties or targeting moieties such as transferring to enable the 
active targeting of CD - based nanoparticles to tumor tissues. Amphiphilic CD nanocarriers 
now emerge as promising delivery systems for poorly soluble anticancer 
drugs, DNA and oligonucleotide delivery, and photodynamic and targeted tumor 
therapy. These systems are proven to be nonhemolytic and noncytotoxic and are 
capable of prolonging the release of drugs with different properties. 
ACKNOWLEDGMENTS 
The authors wish to thank the TUBITAK Turkish Council of Scientifi c and Technical 
Research, projects SBAG - CNRS - 3 and SBAG - CD - 66, and the Hacettepe University 
Research Fund, project 0202301005, for fi nancial support of the amphiphilic 
cyclodextrin research carried out by our group at Hacettepe University, Faculty of 
Pharmacy, Department of Pharmaceutical Technology. 
REFERENCES 
1. Albers , E. , and M u ller , B. W. ( 1995 ), Cyclodextrin derivatives in pharmaceuticals , CRC 
Crit. Rev. Ther. Drug Carrier Syst. , 12 ( 4 ), 311 . 
2. Loftsson , T. , and Duchene , D. ( 2007 ), Cyclodextrins and their pharmaceutical applications , 
Int. J. Pharm. , 329 ( 1 – 2 ), 1 – 11 . 
3. Uekama , K. ( 2004 ), Design and evaluation of cyclodextrin - based drug formulation , Chem. 
Pharm. Bull. , 52 ( 8 ), 900 – 915 . 
4. Wenz , G. ( 1991 ), Synthesis and characterization of some lipophilic per(2,6 - di - O - alkyl) 
cyclomaltooligosaccharides , Carbohydr. Res. , 214 , 257 – 265 . 
5. Duchene , D. , and Wouessidjewe , D. ( 1992 ), Industrial uses of cyclodextrins and their 
derivatives , J. Coord. Chem. , 27 , 223 – 236 . 
6. Thompson , D. O. ( 1997 ), Cyclodextrins as enabling excipients: Their present and future 
use in pharmaceuticals , CRC Crit. Rev. Ther. Drug Carrier Syst. , 14 ( 1 ), 1 – 110 . 
7. Rajewski , R. A. , and Stella , V. J. ( 1996 ), Pharmaceutical applications of cyclodextrins 2. 
In vivo drug delivery , J. Pharm. Sci. , 85 ( 11 ), 1142 – 1169 . 
8. Loftsson , T. , and Brewster , M. E. ( 1996 ), Pharmaceutical applications of cyclodextrins I. 
Drug solubilization and stabilization , J. Pharm. Sci. , 85 ( 10 ), 1017 – 1025 . 
9. Frank , D. W. , Gray , J. E. , and Weaver , R. N. ( 1976 ), Cyclodextrin nephrosis in the rat , 
Am. J. Pathol. , 83 , 367 – 382 . 
10. Szejtli , J. , Cserhati , T. , and Sz o gyi , M. ( 1986 ), Interactions between cyclodextrins and cell - 
membrane phospholipids , Carbohydr. Polym. , 6 , 35 – 49 . 
11. Debouzy , J. C. , Fauvelle , F. , Crouzy , S. , Girault , L. , Chapron , Y. , G o schl , M. , and Gadelle , 
A. ( 1997 ), Mechanism of . - cyclodextrin induced hemolysis 2. A study of the factors controlling 
the association with serine - , ethanolamine - and choline - phospholipids , J. Pharm. 
Sci. , 87 ( 1 ), 59 – 66 . 
12. Uekama , K. , Hirayama , F. , and Irie , T. ( 1991 ), Modifi cation of drug release by cyclodextrin 
derivatives , in Duchene , D. , Ed., New Trends in Cyclodextrins and Derivatives , Editions 
de Sant e , Paris , pp. 409 – 446 . 

13. Stella , V. J. , and Rajewski , R. A. ( 1997 ), Cyclodextrins: Their future in drug formulation 
and delivery , Pharm. Res. , 14 ( 5 ), 556 – 567 . 
14. M u ller , B. W. , and Brauns , U. ( 1986 ), Hydroxypropyl - . - cyclodextrin derivatives: Infl uence 
of average degree of substitution on complexing ability and surface activity , J. Pharm. 
Sci. , 75 , 571 – 572 . 
15. Yoshida , A. , Yamamoto , M. , Irie , T. , Hirayama , F. , and Uekama , K. ( 1989 ), Some pharmaceutical 
properties of 3 - hydroxypropyl/ and 2,3 - dihydroxypropyl . - cyclodextrins and 
their solubilizing and stabilizing ability , Chem. Pharm. Bull. , 37 , 1059 – 1063 . 
16. M u ller , B. W. ( 2000 ), Hydroxypropyl . - cyclodextrin in drug formulation, paper presented 
at the CRS Workshop What ’ s New in Cyclodextrin Delivery? Paris, July 7 – 8, 
Article 3. 
17. Zia , V. , Rajewski , R. , Bornancini , E. R. , Luna , E. A. , and Stella , V. J. ( 1997 ), Effect of alkyl 
chain length and substitution degree on the complexation of sulfoalkyl ether . - cyclodextrins 
with steroids , J. Pharm. Sci. , 86 , 220 – 224 . 
18. Stella , V. J. ( 2000 ), SBE 7M - . - CD or Captisol ® - possible utilizations, paper presented at the 
CRS Workshop What ’ s New in Cyclodextrin Delivery? Paris, July 7 – 8, Article 4. 
19. Ma , D. Q. , Rajewski , R. A. , and Stella , V. J. ( 1999 ), Thermal properties and processing of 
(sulfobutylether) - 7M -. - cyclodextrin as a freeze - drying excipient in pharmaceutical formulations 
, STP Pharma Sci. , 9 ( 3 ), 261 – 266 . 
20. Komiyama , M. , Yamamoto , Y. , and Hirai , H. ( 1984 ), Complex formation of modifi ed 
cyclodextrins with organic salts in organic solvents , Chem. Lett. , 1081 – 1084 . 
21. Uekama , K. , Horiuchi , Y. , rie , T. , and Hirayama , F. ( 1989 ), O - Carboxymethyl - O - ethyl - 
cyclomaltoheptaose as a delayed - release type drug carrier. Improvement of the oral 
availability of diltiazem , Carbohydr. Res. , 192 , 323 – 330 . 
22. Uekama , K. , Hirayama , F. , and Irie , T. ( 1998 ), Cyclodextrin drug carrier systems , Chem. 
Rev. , 98 , 2045 – 2076 . 
23. Anand , R. , Nayyar , S. , Pitha , J. , and Merril , C. R. ( 1990 ), Sulphated sugar alpha - 
cyclodextrin sulphate, a uniquely potent anti - HIV agent, also exhibits marked synergism 
with AZT, and lymphoproliferative activity, Antiviral Chem . Chemother. , 1 , 41 – 46 . 
24. Friedman , R. ( 1991 ), Cyclodextrin - containing polymers , in Duchene , D. , Ed., New Trends 
in Cyclodextrins and Polymers , Editions de Sant e , Paris , pp. 157 – 178 . 
25. Sebill e , B. ( 1987 ), Cyclodextrin derivatives , in Duchene , D. , Ed., Cyclodextrins and Their 
Industrial Uses , Editions de Sant e , Paris , pp. 351 – 393 . 
26. Memi o lu , E. , Bochot , A. , en , M. , Duchene , D. , and Hincal , A. A. ( 2003 ), Non - surfactant 
nanospheres of progesterone inclusion complexes with amphiphilic . - cyclodextrins , Int. 
J. Pharm. , 251 , 143 – 153 . 
27. Monza de Silveira , A. , Ponchel , G. , Puisieux , F. , and Duchene , D. ( 1998 ), Combined poly 
(isobutylcyanoacrylate) and cyclodextrin nanoparticles for enhancing the encapsulation 
of lipophilic drug , Pharm. Res. , 15 ( 7 ), 1051 – 1055 . 
28. Duchene , D. , Ponchel , G. , and Wouessidjewe , D. ( 1999 ), Cyclodextrins in targeting. Application 
to nanoparticles , Adv. Drug Deliv. Rev. , 36 , 29 – 40 . 
29. Boudad , H. , Legrand , P. , LeBas , G. , Cheron , M. , Duchene D. , and Ponchel , G. ( 2001 ), 
Combined hydroxypropyl - beta - cyclodextrin and poly(alkylcyanoacrylate) nanoparticles 
intended for oral administration of saquinavir , Int. J. Pharm. , 218 , 113 – 124 . 
30. Challa , R. , Ahuja , A. , Ali , J. , and Khar , R. K. ( 2005 ), Cyclodextrins in drug delivery. An 
updated review , AAPS PharmSciTech , 6 ( 2 ), E329 – E357 . 
31. Cavalli , R. , Peira , E. , Caputo , O. , and Gasco , M. R. ( 1999 ), Solid lipid nanoparticles as 
carriers of hydrocortisone and progesterone complexes with . - cyclodextrins , Int. J. 
Pharm. , 182 , 59 – 69 . 
REFERENCES 1243 
I 
s g S

1244 CYCLODEXTRIN-BASED NANOMATERIALS IN PHARMACEUTICAL FIELD 
32. Radwan , M. A. ( 2001 ), Preparation and in vivo evaluation of parenteral metoclopramide - 
loaded poly(alkylcyanoacrylate) nanospheres in rats , J. Microencapsul. , 18 , 467 – 477 . 
33. Luppi , B. , Cerchiara , T. , Bigucci , F. , Caponio , D. , and Zecchi , V. ( 2005 ), Bovine serum 
albumin nanospheres carrying progesterone inclusion complexes , Drug Deliv. , 12 , 
281 – 287 . 
34. Maestrelli , F. , Garcia - Fuentes , M. , Mura , P. , and Alonso , M. J. ( 2006 ), A new drug nanocarrier 
consisting of chitosan and hydoxypropylcyclodextrin , Eur. J. Pharm. Biopharm. , 
69 ( 2 ), 79 – 86 . 
35. Sajeesh , S. , and Sharma , C. P. ( 2006 ), Cyclodextrin - insulin complex encapsulated polymethacrylic 
acid based nanoparticles for oral insulin delivery , Int. J. Pharm. , 325 , 
147 – 154 . 
36. Eguchi , M. , Da , Y. Z. , Ogawa , Y , Okada , T. , Yumoto , N. , and Kodaka , M. ( 2006 ), Effects 
of conditions for preparing nanoparticles composed of aminoethylcarbamoyl - . - 
cyclodextrin and ethylene glycol diglycidyl ether on trap effi ciency of a guest molecule , 
Int. J. Pharm. , 311 , 215 – 222 . 
37. Bellocq , N. C. , Pun , S. H. , Jensen , G. S. , and Davis , M. E. ( 2003 ), Transferrin - containing, 
cyclodextrin polymer - based particles for tumor - targeted gene delivery , Bioconjug. Chem. , 
14 , 1122 – 1132 . 
38. Hwang , S. , Bellocq , N. , and Davis , M. ( 2001 ), Effects of structure of beta - cyclodextrin - 
containing polymers on gene delivery, Bioconjug. Chem. , 12, 280 – 290 . 
39. Hede , K. ( 2005 ), Blocking cancer with RNA interference moves toward the clinic , J. 
Natl. Cancer Inst. , 97 ( 4 ), 626 – 628 . 
40. Gao , H. , Wang , Y. N. Fan , Y. G. , and Ma , J. B. ( 2006 ), Conjugates of poly( dl - lactic acid) 
with ethylenediamino or diethylenetriamino bridged bis( . - cyclodextrin)s and their 
nanoparticles as protein delivery systems , J. Controlled Release , 112 , 301 – 311 . 
41. Gao , H. , Wang , Y. N. , Fen , Y. G. , and Ma , J. B. ( 2007 ), Conjugation of poly( dl - lactide - co - 
glycolide) on amino cyclodextrins and their properties as protein delivery system , 
J. Biomed. Mater. Rest. A , 80A , 111 – 122 . 
42. Daoud - Mahammed , S. , Ringard - Lefebvre , C. , Razzouq , N. , Rosilio , V. , Gillet , B. , Couvreur 
, P. , Amiel , C. , and Gref , R. ( 2007 ), Spontaneous association of hydrophobized dextran 
and poly - . - cyclodextrin into nanoassemblies , J. Colloid Interf. Sci. , 307 ( 1 ), 83 – 93 . 
43. Wang , H. , Chen , Y. , Liu , X. Y. , and Liu , Y. ( 2007 ), Synthesis of oligo(ethylenediamino) -. - 
cyclodextrin modifi ed gold nanoparticles as a DNA concentrator , Mol. Pharm. , 4 ( 2 ), 
189 – 198 
44. Park , I. K. , van Recum , H. A. , Jiang , S. , and Pun , S. H. ( 2006 ), Supramolecular assembly 
of cyclodextrin - based nanoparticles on solid surfaces for gene delivery , Langmuir , 22 , 
8478 – 8484 . 
45. Munoz , M. , Deschenaux , R. , and Coleman , A. W. ( 1999 ), Observation of microscopic 
patterning at the air/water interface by mixtures of amphiphilic cyclodextrins — A 
comparison isotherm and Brewster angle microscopy study , J. Phys. Org. Chem. , 12 , 
364 – 369 . 
46. Duchene , D. , Wouessidjewe , D. , and Ponchel , G. ( 1999 ), Cyclodextrins and carrier systems , 
J. Controlled Release , 62 , 263 – 268 . 
47. Memisoglu - Bilensoy , E. , Bochot , A. , Trichard , L. , Duchene , D. , and Hincal , A. A. ( 2005 ), 
Amphiphilic cyclodextrins and microencapsulation , in Benita , S. , Ed, Microencapsulation , 
2nd rev. ed. , Taylor & Francis , New York , pp. 269 – 295 . 
48. Bellanger , N. , and Perly , B. ( 1992 ), NMR investigations of the conformation of new cyclodextrins 
- based amphiphilic transporters for hydrophobic drugs: Molecular lollipops , 
J. Mol. Struct. , 15 , 215 – 226 . 

49. Dodziuk , H. , Chmurski , K. , Jurczak , J. , Kozminski , W. , Lukin , O. , Sitkowski , J. , and Stefaniak 
, J. ( 2000 ), A dynamic NMR study of self - inclusion of a pendant group in amphiphilic 
6 - thiophenyl - 6 - deoxycyclodextrins , J Mol. Struct. , 519 , 33 – 36 . 
50. Lin , J. ( 1995 ), Synth e se des cyclodextrins amphiphiles et e tude de leur incorporation dans 
des phases phospholipidiques, Ph.D. dissertation, University of Paris, Paris. 
51. Kawabata , Y. , Matsumoto , M. , Tanaka , M. , Takahashi , H. , Irinatsu , Y. , Tagaki , W. , Nakahara 
, H. , and Fukuda , K. ( 1983 ), Formation and deposition of monolayers of amphiphilic 
. - cyclodextrin derivatives , Chem. Lett. , 1933 – 1934 . 
52. Djedaini , F. , Coleman , A. W. , and Perly , B. ( 1990 ), New cyclodextrins based media for 
vectorization of hydrophilic drug, mixed vesicles composed of phospholipids and lipophilic 
cyclodextrins , in Duchene , D. , Ed., Minutes of the 5th International Symposium on 
Cyclodextrins , Editions de Sant e , Paris , pp. 328 – 331 . 
53. Liu , F. Y. , Kildsig , D. O. , and Mitra , A. K. ( 1992 ), Complexation of 6 - acyl - O - . - cyclodextrin 
derivatives with steroids, effects of chain length and substitution degree , Drug Del. Ind. 
Pharm. , 18 , 1599 – 1612 . 
54. Zhang , P. , Ling , C. C. , Coleman , A. W. , Parrot - Lopez , H. , and Galons , H. ( 1991 ), Formation 
of amphiphilic cyclodextrins via hydrophobic esterifi cation at the secondary hydroxyl 
face , Tetrahedron Lett. , 32 , 2769 – 2770 . 
55. Zhang , P. , Parrot - Lopez , H. , Tchoreloff , P. , Basakin , A. , Ling , C. C. , De Rango , C. , and 
Coleman , A. W. ( 1992 ), Self - organizing systems based on amphiphilic cyclodextrins diesters 
, J. Phys. Org. Chem. , 5 , 518 – 528 . 
56. Memi o lu , E. , Charon , D. , Duchene , D. , and H . ncal , A. A. ( 1999 ), Synthesis of per(2,3 - 
di - O - hexanoyl) - . - cyclodextrin and characterization amphiphilic cyclodextrins nanoparticles 
, in Torres - Labandeira , J. , and Vila - Jato , J. , Eds., Proceedings of the 9th International 
Symposium on Cyclodextrins , Kluwer Academic , Dordrecht , pp. 622 – 624 . 
57. Lesieur , S. , Charon , D. , Lesieur , P. , Ringerd - Lefebvre , C. , Muguet , V. , Duchene , D. , and 
Wouessidjewe , D. ( 2000 ), Phase behaviour of fully - hydrated DMPC - amphiphilic cyclodextrin 
systems , Chem. Phys. Lip. , 10 , 127 – 144 . 
58. Canceill , J. , Jullien , L. , Lacombe , L. , and Lehn , J. M. ( 1992 ), Channel type molecular 
structures, Part 2, synthesis of bouquet - shaped molecules based on a . - cyclodextrin core , 
Helv. Chim. Acta , 75 , 791 – 812 . 
59. Auzely - Velty , R. , Djedaini - Pilard , F. , Desert , S. , Perly , B. , and Zemb , T. ( 2000 ), Micellization 
of hydrophobically modifi ed cyclodextrins 1. Miceller structures , Langmuir , 16 , 
3727 – 3734 . 
60. Tchoreloff , P. , Boissonnade , M. M. , Coleman , A. W. , and Baszkin , A. ( 1995 ), Amphiphilic 
monolayers of insoluble cyclodextrins at the water/air interface. Surface pressure and 
surface potential studies , Langmuir , 11 , 191 – 196 . 
61. Ringard - Lefebvre , C. , Bochot , A. , Memi o lu , E. , Charon , D. , Duchene , D. , and Baszkin , 
A. ( 2002 ), Effect of spread amphiphilic . - cyclodextrins on interfacial properties of the 
oil/water system , Coll. Surf. B Biointerf. , 25 , 109 – 117 . 
62. Memi o lu , E. , Bochot , A. , en , M. , Charon , D. , Duchene , D. , and Hincal , A. A. ( 2002 ), 
Amphiphilic . - cyclodextrins modifi ed on the primary face: Synthesis, characterization 
and evaluation of their potential as novel excipients in the preparation of nanocapsules , 
J. Pharm. Sci. , 95 ( 1 ), 1214 – 1224 . 
63. Bilensoy , E. , Do an , A. L. , en , M. , and Hincal , A. A. ( 2007 ), Complexation behaviour of 
antiestrogen drug tamoxifen citrate with natural and modifi ed cyclodextrins , J. Inclus. 
Phenom. Macroc. Chem. , 57 , 651 – 655 . 
64. Alexandre , S. , Coleman, A. W. , Kasselouri, A. , and Valleton, J. M. (1996), Scanning force 
microscopy investigation of amphiphilic cyclodextrin Langmuir - Blodgett fi lms , Thin Solid 
Films , 284 – 285 , 765 – 768 . 
REFERENCES 1245 
s g 
s g 
s g S 
g S

1246 CYCLODEXTRIN-BASED NANOMATERIALS IN PHARMACEUTICAL FIELD 
65. Tanaka , M. , Ishizuka , U. , Matsumoto , M. , Nakamura , T. , Yabe , A. , Nakanishi , H. , 
Kawabata , Y. , Takahashi , H. , Tamura , S. , Tagaki , W. , Nakahara , H. , and Fukuda , K. ( 1987 ), 
Host - guest complexes of an amphiphilic . - cyclodextrin and azobenzene derivatives in 
Langmuir - Blodgett fi lms , Chem. Lett. , 1307 – 1310 . 
66. Mazzaglia , A. , Angelini , N. , Darcy , R. , Donohue , R. , Lombardo , D. , Micali , N. , Sciortino , 
M. T. , Villari , V. , and Scolaro , L. M. ( 2003 ), Novel heterotropic colloids of anionic porphyrins 
entangled in cationic amphiphilic cyclodextrins: Spectroscopic investigation and 
intracellular delivery , Chem. Eur. J. , 9 , 5762 – 5769 . 
67. Matsumoto , M. , Matsuzawa , Y. , Noguchi , S. , Sakai , H. , and Abe , M. ( 2004 ), Structure of 
Langmuir - Blodgett fi lms of amphiphilic cyclodextrin and water - soluble benzophenone , 
Mol. Cryst. Liq. Cryst. , 425 , 197 – 204 . 
68. Cryan , S. A. , Holohan , A. , Donohue , R. , Darcy , R. , and O ’ Driscoll , C. M. ( 2004 ), Cell 
transfection with polycationic cyclodextrin vectors , Eur. J. Pharm. Sci. , 21 , 625 – 633 . 
69. Dubes , A. , Bouchu , D. , Lamartine , R. , and Parrot - Lopez , H. ( 2001 ), An effi cient regiospecifi 
c synthetic route to multiply substituted acyl - sulphated . - cyclodextrins , Tetrahedron 
Lett. , 42 , 9147 – 9151 . 
70. Dubes , A. , Degobert, G. , Fessi, H. , and Parrot-Lopez, H. (2003), Synthesis and characterisation 
of sulfated amphiphilic alpha - , beta - and gamma - cyclodextrins: Application to the 
complexation of acyclovir , Carbohydr. Res. , 338 , 2185 – 2193 . 
71. Granger , C. E. , Feliz, C. P. , Parrot-Lopez, H. , and Langlois , B. R. (2000), Fluorine containing 
. - cyclodextrin: A new class of amphiphilic carriers , Tetrahedron Lett. , 41 , 9257 – 
9260 . 
72. P e roche , S. , Degobert , G. , Putaux , J. L. , Blanchin , M. G. , Fessi , H. , and Parrot - Lopez , H. 
( 2005 ), Synthesis and characterization of novel nanospheres made from amphiphilic 
perfl uoroalkylthio - . - cyclodextrins , Eur. J. Pharm. Biopharm. , 60 , 123 – 131 . 
73. Skiba , M. , Skiba - Lahiani , M. , and Arnaud , P. ( 2002 ), Design of nanocapsules based on 
novel fl uorophilic cyclodextrin derivatives and their potential role in oxygen delivery , 
J. Inclus. Phenom. Macroc. Chem. , 44 , 151 – 154 . 
74. Fessi , H. C. , Devissaguet , J. P. , Puisieux , F. , and Thies , C. ( 1997 ), Process for the preparation 
of dispersible colloidal systems of a substance in the form of nanoparticles, U.S. 
Patent 5,118,528 . 
75. Wouessidjewe , D. , Skiba , M. , Leroy - Lechat , F. , Lemos - Senna , E. , Puisieux , F. , and Duchene , 
D. ( 1996 ), A new concept in drug delivery based on “ skirt - shaped cyclodextrins aggregates 
” present state and future prospects , STP Pharma. Sci. , 6 , 21 – 26 . 
76. Lemos - Senna , E. , Wouessidjewe , D. , Lesieur , S. , Puisieux , F. , Couarrazze , G. , and Duchene , 
D. ( 1998 ), Evaluation of the hydrophobic drug loading characteristics in nanoprecipitated 
amphiphilic cyclodextrins nanospheres , Pharm. Dev. Technol. , 3 , 1 – 10 . 
77. Lemos - Senna , E. , Wouessidjewe , D. , Lesieur , S. , and Duchene , D. ( 1998 ), Preparation of 
amphiphilic cyclodextrin nanospheres using the emulsion solvent evaporation method, 
infl uence of the surfactant on preparation and hydrophobic drug loading , Int. J. Pharm. , 
170 , 119 – 128 . 
78. Lemos - Senna , E. ( 1998 ), Contribution a l ’ etude pharmacotechnique et physicochimique 
de nanospheres de cyclodextrins amphiphiles comme transporteurs de principes actifs, 
Ph.D. thesis, University of Paris, Paris. 
79. Skiba , M. , Wouessidjewe , D. , Fessi , H. , Devissaguet , J. P. , Duchene , D. , and Puisieux , F. 
( 1992 ), Preparation et utilizations des nouveau systemes colloidaux dispersibles a base 
de cyclodextrines sous forme de nanocapsules, French Patent 92 - 07285. 
80. Memisoglu - Bilensoy , E. , Do an , A. L. , and Hincal , A. A. ( 2006 ), Cytotoxic evaluation of 
injectable amphiphilic cyclodextrin nanoparticles on fi broblasts and polymorphonuclear 
cells: Surfactant effect , J. Pharm. Pharmacol. , 58 ( 5 ), 585 – 589 . 
g

81. Memi o lu - Bilensoy , E. , en , M. , and Hincal , A. A. ( 2006 ), Effect of drug physicochemical 
properties on in vitro characteristics of amphiphilic cyclodextrin nanospheres and nanocapsules 
, J. Microencapsul. , 23 ( 1 ), 59 – 68 . 
82. Memi o lu - Bilensoy , E. , Vural , . , Renoir , J. M. , Bochot , A. , Duchene , D. , and Hincal , 
A. A. ( 2005 ), Tamoxifen citrate loaded amphiphilic . - cyclodextrin nanoparticles: In vitro 
characterization and cytotoxicity , J. Controlled Release , 104 , 489 – 496 . 
83. Vural , . , Memi o lu - Bilensoy , E. , Renoir , J. M. , Bochot , A. , Duchene , D. , and Hincal , A. 
A. (2005), Transcription effi ciency of tamoxifen citrate loaded . - cyclodextrin nanoparticles 
, J. Drug Deliv. Sci. Technol. , 15 ( 5 ), 339 – 342 . 
84. Bilensoy , E. , G u rkaynak , O. , Ertan , M. , en , M. , and Hincal , A. A. , Development of non - 
surfactant cyclodextrin nanoparticles loaded with anticancer drug paclitaxel, J. Pharm. 
Sci. , DOI: 10.1002/jps.2111, Published online . 
85. Bilensoy , E. , G u rkaynak , O. , Do an , A. L. , Duman , M. , and H . ncal , A. A. , Safety and 
effi cacy of amphiphilic cyclodextrin nanoparticles for paclitaxel delivery, Int. J. Pharm. , 
DOI: 10.1016/j.ijpharm.2007.06.05 . 
86. C . rpanl . , Y. , Bilensoy , E. , C al . , S. , and Hincal , A. A. ( 2006 ), Camptothecin inclusion 
complexes with natural and modifi ed . - cyclodextrins, in Proceedings of the 33rd Annual 
Meeting and Exhibition on Controlled Release Society , Vienna, July, 22 – 26 pp. 988 – 989 . 
87. C . rpanl . . Y. , Bilensoy , E. , C al . , S. , and Hincal , A. A. ( 2007 ), Development of camptothecin 
loaded nanoparticles from amphiphilic . - cyclodextrin derivatives, paper presented at the 
Pharmaceutical Sciences World Congress PSWC, Amsterdam, April, 22 – 25. 
88. Sortino , S. , Mazzaglia , A. , Scolaro , L. M. , Merlo , F. M. , Volveri , V. , and Sciortino , M. T. 
( 2006 ), Nanoparticles of cationic amphiphilic cyclodextrins entangling anionic porphyrins 
as carrier - sensitizer system in photodynamic cancer therapy , Biomaterials , 27 , 4256 – 
4265 . 
89. Skiba , M. , Morvan , C. , Duchene , D. , and Puisieux , F. , ( 1995 ), Evaluation of the gastrointestinal 
behaviour in the rat of amphiphilic . - cyclodextrin nanocapsules loaded with 
indomethacin , Int. J. Pharm. , 126 , 275 – 279 . 
90. Memisoglu - Bilensoy , E. , and Hincal , A. A. ( 2006 ), Sterile injectable cyclodextrin nanoparticles: 
Effects of gamma irradiation and autoclaving , Int. J. Pharm. , 311 , 203 – 208 . 
REFERENCES 1247 
s g S 
s g I 
I s g 
S 
g 
s 
s


1249 
7.2 
NANOTECHNOLOGY IN 
PHARMACEUTICAL 
MANUFACTURING 
Yiguang Jin 
Beijing Institute of Radiation Medicine, Beijing, China 
Contents 
7.2.1 Introduction 
7.2.2 Nanomaterials 
7.2.2.1 Types of Nanomaterials 
7.2.2.2 Manufacturing and Processing of Nanomaterials 
7.2.3 Nanotechnology for Drug Delivery 
7.2.3.1 Nanocarriers 
7.2.3.2 Nanosuspensions 
7.2.3.3 Self - Assembled Drug Nanostructures 
7.2.4 Nanomedicine 
7.2.5 Perspective 
References 
7.2.1 INTRODUCTION 
Nanotechnology is the ability to produce and process nanosized materials or manipulate 
objects within the nanoscale. The nanoscale commonly indicates the range 
from 1 to 100 nm [1] . However, some scientists regard the nanoscale range from 1 
to 200 nm [2] , even to 1000 nm [3] . Making a comparison with a human hair, it is 
about 80,000 nm wide. Nanotechnology is a broad, highly interdisciplinary, and still 
evolving fi eld which involves the production and application of physical, chemical, 
and biological systems. Nanotechnology is likely to have a profound impact on our 
economy and society in the early twenty - fi rst century, perhaps comparable to that 
of information technology or advances in cellular and molecular biology. Science 
and engineering research in nanotechnology promises breakthroughs in areas such 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

1250 NANOTECHNOLOGY IN PHARMACEUTICAL MANUFACTURING 
as materials, manufacturing, electronics, medicine, health care, energy, environment, 
biotechnology, information technology, and national security. It is widely felt 
that nanotechnology will lead to the next industrial revolution [4] . 
The idea of nanotechnology was fi rst presented by physicist Richard Feynman. 
His lecture entitled “ Room at the Bottom ” in 1959 unveiled the possibilities available 
in the molecular world. Because bulk matter is built of so many atoms, there 
is a remarkable amount of space within which to build. Feynman ’ s vision spawned 
the discipline of nanotechnology, and his dream is now coming true [5] . Along with 
continually increasing multidisciplinary applications of nanotechnology, many new 
terms with nanotechnology characteristics appear, for example, nanomechanics [6] , 
nanooptics [7] , nanoelectronics [8] , nanochemistry [9] , nanomedicine [10] , nanobiotechnology 
[5, 11] , nanolithography [12] , nanoengineering [13] , nanofabrication [14] , 
and nanomanufacturing [15] . A very broad sense term, nanoscience is often used. 
More and more new words with nano as a prefi x will be created to fi t for the nowadays 
nanoworld. In fact, applications of nanotechnology in medicine and biotechnology 
have made great progresses in the recent two decades. 
All developed countries including the United States, Japan, and Europe invest 
a great deal of money in nanotechnology. The National Science Foundation (NSF) 
of the United States is a leading agency in the national nanotechnology initiative, 
funding nanotechnology investments at $ 373 million in 2007, an increase of 8.6% 
from 2006 and of nearly 150% since 2001 [16] . Developing countries such as China 
and India also invest a lot in this increasing fi eld so as not to stay far behind developed 
countries. Cancer therapy and research are hottest applied fi elds of bionanotechnology. 
In 2004, the U.S. National Cancer Institute (NCI) launched a $ 144 
million cancer nanotechnology initiative, and the investment increased largely in 
the following two years [17] . At the same time, investment from public resources 
or companies is much higher than that from governments. 
The application of nanotechnology in pharmacy has a long history, before the 
prevalence of the nanoconcept. It was well known 50 years ago that very small drug 
particles have a high solubility in solvents, resulting from the too large surface area 
when particle size decreased to a very small level, that is, the nanoscale, although 
this scale had not been mentioned yet. In 1965, Banham created liposomes (lipid 
vesicles) consisting of phospholipids which had a small size, typically ranging from 
10 nm to several micrometers. It was soon found that liposomes were excellent drug 
carriers, and more importantly they had site - specifi c distribution capability in vivo 
depending on their size. It is well known that nanosized liposomes are inclined to 
deposit in the mononuclear phagocyte system (MPS), including liver, spleen, lung, 
and marrow. Therefore, nanotechnology was introduced in drug delivery very long 
ago. Now various nanomaterials are used to deliver drugs, and some nanosystems 
delivering active agents are available on the market. Undoubtedly, nanotechnology 
plays a key role in future pharmaceutical development and pharmacotherapy. 
7.2.2 NANOMATERIALS 
7.2.2.1 Types of Nanomaterials 
Nanomaterial is a general term. Although nanomaterials are defi ned as solid or 
liquid materials at the nanoscale, the nanoscale range remains unclear. Many scien

NANOMATERIALS 1251 
tists regard materials that are one dimensional and 1 nm to less than 100 nm as 
nanomaterials. However, some scientists treat larger materials (e.g., less than 
200 nm) as nanomaterials [2] . In spite of the different views, nanomaterials show 
unique characteristics that are different from those of bulk materials. Rapid development 
of nanotechnology in varied disciplines helps to create various kinds of 
nanomaterials. In terms of shape differences, nanomaterials can be classifi ed as 
nanospheres, nanovesicles, nanoshells, nanotubes, nanohorns, nanofi bers, nanowires, 
nanoribbons, nanorods, nanosticks, nanohelices, and so on, and they can 
appear in any shape imagined. In terms of state differences, nanomaterials can also 
be classifi ed as nanoparticles with solid cores, nanoemulsions with liquid cores, and 
nanobubbles with air cores. Images of some nanomaterials of various shapes are 
shown in Figure 1 . 
Nanoparticle is the most usually used term, having a broad meaning. From a 
narrow sense, nanoparticles are always used to indicate all ball - like nanomaterials, 
and therein the term nanosphere is also used. Nanocapsules are core – shell nanoparticles, 
wherein trapped drugs are gathered in a core coated with a hard shell, though 
generally nanoparticles have uniformly dispersed drugs within the whole particle. 
Nanovesicle is not a familiar term, for example, liposomes have an inner phase and 
an outer phase (dispersing medium) that exist together in nanovesicles [18] . In light 
of drug nature, especially solubility, drugs are entrapped in an inner phase or bilayers 
(shells). In addition, nanosuspension often appears in the pharmaceutical fi eld, 
meaning drug nanocrystal dispersion in liquid media [19] . Needle - shaped nanocrystals 
are more common than globe - shaped ones. Nanogels are newly developed 
based on hydrogels, being similar to nanoparticles after lyophilization [20] . Recently, 
a special kind of nanomaterial consisting of drugs was created for drug delivery, 
called self - assembled drug nanostructure (SADN), which is formed by the self - 
assembly of amphiphilic prodrugs in aqueous media [21, 22] . 
Some special nanomaterials are of great interest due to their unique properties. 
Dendrimers are versatile, well - defi ned, nanosized monodispersing macromolecules 
which are hyperbranched synthesized polymers constructed by repetitive monomer 
units. They are perfect nanoarchitectures with size from 1 nm to more than 10 nm 
depending on the synthesis generation. Drugs can be entrapped into the branches 
FIGURE 1 Some typical nanomaterials. 
Nanospheres Nanorods Nanovesicles 
Nanotubes Nanofibers Nanohelices

1252 NANOTECHNOLOGY IN PHARMACEUTICAL MANUFACTURING 
of dendrimers or conjugated with them on the high reactive surfaces [23] . The fi rst 
fullerene discovered was the buckyball, also known as buckminsterfullerene. It was 
discovered by Smalley, Curl, and Kroto in 1985 [24] , who shared a Nobel Prize in 
1996 for the discovery. Buckyball is roughly spherical cages of 60 carbon atoms 
(C60 ) arranged in interlocking hexagons and pentagons, like the patches on a soccer 
ball. Fullerenes have attracted considerable research interest, partly because of 
their unique structures and further because, once suitably dissolved, they display a 
diverse range of biological activity [25] . Quantum dots (QDs) are semiconductor 
nanocrystals commonly consisting of CdSe or ZnS. Besides their utilization as 
electronic materials, QDs have recently been applied to biomedical areas after 
modifi cation. The new generations of QDs have far - reaching potential for the study 
of intracellular processes at the single - molecule level, high - resolution cellular 
imaging, long - term in vivo observation of cell traffi cking, tumor targeting, and 
diagnostics [26] . 
Although many types of nanomaterials are created continually, the most important 
and basic issues are nanoscale effects and the subsequent particular functions. 
Nanomaterials with varied shapes and components provide different platforms to 
achieve more functions. In the area of pharmaceutical manufacturing, people focus 
on the drug delivery function of nanomaterials. Furthermore, the rapid development 
of modern medicine has led to the belief that traditional drug dosage forms 
such as tablets, capsules, and injections may not treat some vital diseases well, 
perhaps not at all. Some advanced techniques developed in other disciplines should 
be considered to apply to medicine. Nanomaterials can load and deliver drugs in 
vivo as well as display special properties such as high dispersion, adhesive property, 
and site - specifi c distribution in vivo. Modifi ed nanomaterials further possess new 
functions, for example, they may be thermally sensitive, pH sensitive, magnetically 
sensitive, and ultrasound sensitive. 
Nanotechnology has a great effect on pharmaceutical manufacturing. The unique 
functions of nanomaterials promise considerable benefi t to pharmacotherapy over 
traditional drug preparations. When drug - loaded nanomaterials go through the 
gastrointestinal tract, high dispersion and adhesion can lead to tight contact of 
nanomaterials with mucous membranes, enhancing drug absorption. Nanomaterials 
have been applied in all routes of administration, including oral, injection (intravenous, 
subcutaneous, intramuscular, intra - articular cavity, and other possible injection 
sites), intranasal, pulmonary inhalation, conjunctiva, topical, and transdermal, 
possibly showing various required effects. Some of the characteristics and pharmaceutical 
applications of nanomaterials are given in Table 1 . More applications will 
continue to be developed. 
7.2.2.2 Manufacturing and Processing of Nanomaterials 
When material dimensions reach the nanoscale, quantum mechanical and thermodynamic 
properties that are insignifi cant in bulk materials dominate, causing these 
nanomaterials to display new and interesting properties. The manufacturing and 
processing of nanomaterials may become diffi cult due to the unique properties. The 
very small size of nanomaterials produces a very large surface - to - volume ratio, that 
is, a great number of molecules/atoms locate on surfaces. High surface energy leads 
to nanomaterials easily agglomerating to diminish energy unless enough hindrance 

NANOMATERIALS 1253 
TABLE 1 Characteristics and Applications of Some Nanomaterials in Pharmacy 
Types of 
Nanomaterials Characteristics 
Applications in 
Pharmacy References 
Nanoparticles Solid nanosized particles 
consisting of polymers, 
lipids, or inorganic 
materials spherically 
shaped most of the 
time, entrapped 
compounds dispersing 
in the whole particle 
Loading all kinds 
of active agents, 
including drugs, 
vaccines, diagnostic 
agents, and imaging 
agents for good 
bioavailability, 
targeted delivery, and 
controlled release 
27, 28 
Nanocapsules Core – shell nanoparticles 
with entrapped 
compounds gathering 
in the core 
Loading all kinds of 
active agents for 
same aims as 
nanoparticles, 
possibly protecting 
entrapped agents 
29 
Liposomes Lipid vesicles with 
entrapped compounds 
in inner phase or 
bilayers depending on 
physicochemical 
property 
Loading all kinds of 
active agents for 
good bioavailability, 
targeted delivery, and 
controlled release 
30, 31 
Niosomes Nonionic surfactant 
vesicles with similar 
property as liposomes 
Loading all kinds of 
active agents for 
same aims as for 
liposomes 
32 
Nanoemulsions Nanoscale emulsions Loading drugs, as a 
method to prepare 
nanoparticles 
33, 34 
Polymeric 
micelles 
Micelles consisting of 
amphiphilic polymers 
Loading hydrophobic 
drugs in the core for 
solubilization, 
targeted delivery, and 
controlled release 
35, 36 
Nanogels Nanosized hydrogels 
consisting of cross - 
linked hydrophilic 
polymers 
Loading various 
compounds for 
controlled release 
or targeting 
20 
Dendrimers Well - defi ned, nanosized, 
monodispersing 
macromolecules with 
hyperbranched 
structures 
Loading all kinds of 
active agents for 
good bioavailability, 
targeted delivery, and 
controlled release 
23 

1254 NANOTECHNOLOGY IN PHARMACEUTICAL MANUFACTURING 
Types of 
Nanomaterials Characteristics 
Applications in 
Pharmacy References 
Carbon 
nanotubes 
(CNTs) 
Nanosized tubes as if 
rolling up a single 
layer of graphite sheet 
(single - walled CNTs; 
SWNTs) or by rolling 
up many layers to 
form concentric 
cylinders (multiwalled 
CNTs; MWNTs) with 
diameters of . 1 nm 
and large length – 
diameter ratio 
Linking a wide variety 
of active molecules 
with functionalized 
CNTs 
37 
Fullerenes Very tiny balls consisting 
of 60 carbon atoms 
with diameter of 
. 0.7 nm 
Water - soluble 
carboxylic acid C 60 
derivatives acting as 
antimicrobials, being 
linked to a variety of 
active molecules 
25, 38, 39 
Quantum dots Tiny nanocrystals 
commonly consisting 
of semiconductor 
materials in the range 
of 2 – 10 nm, glowing 
upon ultraviolet (UV) 
light 
Mainly as probes to 
track antibodies, 
viruses, proteins, or 
deoxyribonucleic acid 
(DNA) in vivo 
26, 40 
Nanosuspensions Drug nanocrystals 
dispersing in aqueous 
media commonly 
stabilized by 
surfactants 
Suitable for insoluble 
drugs to obtain good 
bioavailability and 
targeting 
19 
Self - assembled 
drug 
nanostructures 
Nanostructures 
consisting of 
amphiphilic prodrugs 
Suitable for hydrophilic 
drugs to obtain good 
bioavailability, 
targeting, and 
controlled release 
21, 22 
prevents them from agglomeration. As a result, manufacturing and processing of 
nanomaterials become hard issues. Anyway, many successful methods have been 
found to manufacture stable nanomaterials. 
“ Top down ” and “ bottom up ” are two basic ways to manufacture nanomaterials. 
From its apparent meaning, the top - down method starts with a bulk material and 
then breaks it into smaller pieces using mechanical, chemical, or other forms of 
energy. Microchip manufacturing is the most common example of the top - down 
approach to produce nanomaterials. While this is an effi cient approach for some 
industries, the process is generally labor and cost intensive. In contrast, the bottom - 
up method produces nanomaterials from atomic or molecular species via chemical 
reactions or physicochemical interactions such as self - assembly, allowing the precur- 
TABLE 1 Continued

NANOMATERIALS 1255 
sor molecules/particles to grow in size. Self - assembly leads to gaining the lowest 
energy state of molecules and makes molecules reorient naturally to obtain ordered 
aggregates. Carbon nanotubes, liposomes, and the SADNs are examples of nanomaterials 
that are manufactured using the bottom - up approach. A deep understanding 
of chemical and physical properties of precursor molecules/particles is needed 
to design and manufacture nanomaterials using the bottom - up approach. Both top - 
down and bottom - up approaches can be performed in gas, liquid, supercritical fl uid, 
solid state, or vacuum. Anyway, when bulk materials corrupt, energy is required, 
and certainly the obtained nanoscale materials stay at a higher energy state than 
their parents. Whereas in the bottom - up approach molecules self - assemble into 
ordered aggregates with controlled behavior. Considering the higher energy of self - 
assembling monomolecules dispersing in media, their aggregation should be an 
energy - diminishing procedure and proceed spontaneously (Figure 2 ). 
One of the largest hurdles of nanomanufacturing is how to scale up production. 
In the laboratory, manufacturing nanomaterials is diffi cult enough as highly 
advanced tools and carefully clean environments are required. Therefore, scale - up 
manufacturing in factories becomes a great challenge, hard to achieve. The most 
successful mass nanomanufacturing to date has occurred with computer microprocessors 
where companies have been able to etch circuit boards at 65 nm or smaller. 
Most manufacturers are interested in the ability to control (a) particle size, (b) 
particle shape, (c) size distribution, (d) particle composition, and (e) degree of particle 
agglomeration. Neither the top - down nor bottom - up approach is superior at 
the moment. Each has its advantages and disadvantages. However, the bottom - up 
approach may have the potential to be more cost - effective in the future. 
Clinical applications require that biomedical nanomaterials have good biocompatibility 
or biodegradability. Therefore, biodegradable polymers (synthetic or 
natural), small molecules such as lipids, and some bioabsorptive inorganic salts such 
as calcium phosphate are preferred. Other materials such as poly(ethylene glycol) 
(PEG) is eventually excreted from body so they can also be selected. Materials that 
are nonbiodegradable or not easily removed from the body, such as carbon nanotubes 
and quantum dots, should be carefully considered as drug carriers, although 
they have already been used to deliver drugs or genes. More importantly, before 
any nanomaterial can be used in a clinic, the acute and long - term toxicity and side 
effects must be estimated in detail. So a novel discipline, nanotoxicology, is of great 
interest [41] . In addition, problems of large - scale production of nanomaterials, for 
example, the uniformity and stability of products, cannot be ignored. Some nanomaterials, 
including liposomes, polymeric or lipid nanoparticles, nanosuspensions, 
and SADNs, are described in detail in the following sections. The common manufacturing 
methods of pharmaceutical nanomaterials are listed in Table 2 , though 
some are only used in the laboratory. 
FIGURE 2 Two basic methods to manufacture nanomaterials. 
Top down Bottom up 
Nanomaterials Atoms or molecules Bulk materials

1256 NANOTECHNOLOGY IN PHARMACEUTICAL MANUFACTURING 
TABLE 2 Manufacturing Methods of Some Nanomaterials in Pharmacy 
Types of 
Nanomaterials Materials Involved a Manufacturing Methods References 
Nanoparticles 
Polymeric 
nanoparticles 
Various natural polymers, 
e.g., albumin, gelatin, 
alginate, collagen, 
chitosan; biodegradable 
synthetic polymers, e.g., 
poly(lactic acid) (PLA), 
poly(lactide - co - 
glycotide) (PLGA), 
poly( . - caprolactone) 
(PCL), poly(methyl 
methacrylate), 
and poly(alkyl 
cyanoacrylate); 
derivatives of 
cyclodextrin and starch; 
some modifi ed 
polymers (e.g., 
PEGylated polymers) 
also used 
Monomer 
polymerization, 
precipitation, solvent 
evaporation, salting 
out 
42 – 44 
Solid lipid 
nanoparticles 
(SLNs) 
Mainly glycerides and 
fatty acids, surfactants 
also used 
High - pressure 
homogenization, 
microemulsion 
technique, solvent 
evaporation 
27 
Inorganic 
nanoparticles 
Calcium salts (e.g., 
calcium carbonate and 
calcium phosphate), 
gold 
Precipitation 45 – 47 
Nanocapsules Various polymers, e.g., 
poly( iso - 
butylcyanoacrylate) 
(PIBCA), PLA, PLGA, 
PCL 
Interfacial 
polymerization of 
monomers or 
interfacial 
nanodeposition of 
polymers 
29, 48, 49 
Liposomes Phospholipids and 
cholesterol, 
phospholipid 
derivatives, e.g., PEG – 
polyethylene (PE), also 
added 
Many methods used, 
mainly fi lm 
hydration, reverse - 
phase evaporation, 
injection, freeze 
drying 
50 
Niosomes Noionic surfactants, e.g., 
sorbitan monostearate 
(Span 60) 
As for liposomes 32 
Nanoemulsions Oil and surfactants High - pressure 
homogenization, 
ultrasonic 
emulsifi cation, phase 
inversion 
34 

NANOMATERIALS 1257 
Types of 
Nanomaterials Materials Involved a Manufacturing Methods References 
Polymeric 
micelles 
Poloxamer - like block 
copolymers; PEG and 
lipophilic polymer 
copolymers; PEGylated 
lipids 
Dialysis, emulsifi cation, 
or fi lm method 
35, 36 
Nanogels Cross - linked hydrophilic 
copolymers, e.g., 
Pluronic – 
poly(ethylenimine) 
(PEI) and polyethylene 
oxide (PEO) – PEI 
Covalent conjugation of 
polymers 
20, 51, 52 
Dendrimers Dendritic macromolecules 
with repetitive moieties 
Divergent or 
convergent synthesis 
23, 53 
Carbon 
nanotubes 
Carbon, but only the 
water - soluble 
derivatives of CNTs 
used in pharmacy 
CNTs formed by 
chemical vapor 
deposition (CVD) 
in presence of Fe 
catalyst, water - 
soluble CNT 
derivatives obtained 
by acid processing 
followed by 
conjugation with 
drugs 
54, 55 
Fullerenes (C 60 ) Carbon, but only the 
water - soluble 
derivatives of C 60 used 
in pharmacy 
C 60 obtained by arc 
discharge method 
using graphite 
electrodes or in a 
benzene fl ame, 
water - soluble C 60 
derivatives obtained 
by acid processing 
followed by 
conjugation with 
drugs 
38, 56 
Quantum dots Water - soluble derivatives 
of semiconductor 
materials (e.g., ZnS, 
PbS, CdSe, InP) used in 
pharmacy 
QDs obtained via 
pyrolysis of 
organometallic 
precursors, water - 
soluble QD 
derivatives obtained 
by chemical reaction 
57 – 59 
Nanosuspensions Pure drugs and stabilizers 
(including surfactants 
or polymers) 
Precipitation, wet 
milling, 
homogenization 
19, 60, 61 
Self - assembled 
drug 
nanostructures 
Polar drugs with proper 
conformation and lipids 
with long chains (e.g., 
glycerides, fatty acids, 
cholesterol) 
Amphiphilic prodrugs 
obtained by synthesis, 
subsequently SADNs 
obtained by injection 
method 
21, 22 
a Organic solvents may be involved and subsequently removed. 
TABLE 2 Continued

1258 NANOTECHNOLOGY IN PHARMACEUTICAL MANUFACTURING 
7.2.3 NANOTECHNOLOGY FOR DRUG DELIVERY 
7.2.3.1 Nanocarriers 
High - throughput screening technologies in drug discovery present an effi cient way 
to fi nd new potential active agents. But in recent years it has become evident that 
the development of new drugs alone is not suffi cient to ensure progress in pharmacotherapy. 
Poor water solubility of potential active molecules, insuffi cient bioavailability, 
fl uctuating plasma levels, and high food dependency are the major and 
common problems. Major efforts have been spent on the development of customized 
drug carriers to overcome the disappointing in vivo fate of those potential 
drugs. For drug carriers the followings are considered: nontoxicity (acute and 
chronic), suffi cient drug - loading capacity, possibility of drug targeting, controlled - 
release characteristic, chemical and physical storage stability (for both drugs and 
carriers), and feasibility of scaling up production with reasonable overall costs. 
Nanocarriers have attracted great interest because they are desirable systems to 
fulfi ll the requirements mentioned above. 
Over the past decade nanocarriers as nanoparticulate pharmaceutical carriers 
have been shown to enhance the in vivo effi ciency of many drugs both in pharmaceutical 
research and the clinical setting, including liposomes, micelles, nanocapsules, 
polymeric nanoparticles and lipid nanoparticles. They perform various 
therapeutically or diagnostically important functions. More importantly, many 
useful modifi cations have been made, including the increased stability and half - life 
of nanocarriers in the circulation, required biodistribution, passive or active targeting 
into the required pathological zone, responsiveness to local physiological stimuli 
such as pathology - associated changes in local pH and/or temperature, and ability 
to serve as imaging/contrast agents for various imaging modalities (gamma scintigraphy, 
magnetic resonance imaging, computed tomography, ultrasonography). In 
addition, multifunctional pharmaceutical nanocarriers have already made a promising 
progress [62] . Some of those pharmaceutical carriers have already found their 
way into clinics, while others are still under preclinical investigation. This section 
presents two of the most promising nanocarriers, that is, liposomes and nanoparticles, 
especially their manufacturing, characteristics, and applications. 
Liposomes Liposomes (lipid vesicles) have a relative long history, fi rst discovered 
by Banham in 1965 [63] . In the following decades, liposomes rapidly became a useful 
drug carrier. During the 1990s, many liposome - based drugs reached the market in 
the United States and Europe. The history of liposomes is the procedure of nanotechnology 
application to biomedicine. Phospholipids have particular structural 
conformation, leading to their self - assembly into bilayers with lipid chains inside 
and polar head groups outside during hydration. Importantly, phospholipids are the 
primary components of cell membranes so that liposomes have good biocompatibility 
without toxicity. The formation of liposomes is almost spontaneous, wherein a 
bottom - up procedure is involved [64] . When relatively free phospholipid molecules 
meet water, their polar head groups have affi nity with water while lipid chains 
repulse water, which subsequently leads to their aggregation due to hydrophobic 
interaction, and then bilayers consisting of phospholipids are formed spontaneously. 
Closed vesicles are further formed by bilayer bending (Figure 3 ). Before phospho

FIGURE 3 Structures of phospholipids and formation of liposomes. 
H3C 
CH3 
N+
CH3 
O 
O O 
O– 
P 
O 
O 
O 
O 
CH3 
H3C 
Dipalmitoyl phosphatidylcholine 
(DPPC) 
3D optimized structure of DPPC 
Polar head group 
Lipid chains 
Self-assembly 
Bilayer 
Vesicle 
(liposome) 
lipids become “ free, ” bulk phospholipids must be dispersed throughout, forming a 
thin fi lm, dissolution or emulsifi cation, wherein additional energy is sometimes 
needed. Liposomes may have a size ranging from 10 nm to more than 10 . m mainly 
depending on composition and manufacturing approaches. A number of reports 
about the preparation of liposomes can be found in the literature and a detailed 
description of liposomes is in Chapter 7.1 of this handbook. In this section the 
NANOTECHNOLOGY FOR DRUG DELIVERY 1259

1260 NANOTECHNOLOGY IN PHARMACEUTICAL MANUFACTURING 
preparation, characteristics, and applications of nanosized liposomes are presented 
as well as some modifi ed liposomes and recent progress. 
Although various methods of manufacturing liposomes are reported, three types 
are usually involved: hydration of lipid fi lm, interface aggregation of lipid molecules 
by emulsion - like process, and lipid solutions dispersing into nonsolvents by an 
injection - like process or controlled mixture. Practical methods are thin - fi lm hydration 
[65] , reverse - phase evaporation [66] , ethanol injection [67] , polyol dilution [68] , 
double emulsions [69] , proliposome method [70] , and high - pressure homogenization 
[71] . Liposomes may have various morphologies related to manufacturing methods, 
mainly multilamellar vesicles (MLVs), large unilamellar vesicles (LUVs), and small 
unilamellar vesicles (SUVs). Liposomes can be further processed by sonication, 
detergent depletion, membrane fi ltration [72] , and lyophilization [73] to make them 
fi ner and more uniform or stable. For example, MLVs are sonicated to SUVs. 
The composition of liposomes is a key factor in their manufacturing. Phospholipids 
are major components of liposomes. In terms of resources, phospholipids are 
classifi ed as natural, semisynthetic, and wholly synthetic phospholipids. Natural 
phospholipids also have different resources (e.g., soybean, egg yolk). In terms of 
polar head groups, phospholipids are classifi ed as phosphatidylcholine (PC), phosphatidylethanolamine 
(PE), phosphatidylserine (PS), phosphatidylinositol (PI), 
phosphatidylglycerol (PG), and phosphatidic acid (PA), where PC and PE are the 
most used. Different polar head groups result in varied surface charged liposomes 
that then infl uence the stability and in vivo distribution. Because PS, PI, PG, and PA 
have negative charges, the liposomes containing them are negatively charged. Sometimes, 
other lipids such as N,N. - dioleoyl - N,N. - dimethylammonium chloride 
(DODAC) and stearylamine are mixed with phospholipids to prepare positively 
charged liposomes. Cholesterol is commonly used with phospholipids because cholesterol 
can make liposomal membranes stronger [50] . The mole percentage of 
cholesterol in the liposomal composition is commonly not more than 50%. Lecithin 
(an often used term in the lipid fi eld) as a phospholipid from natural resources (e.g., 
soybean lecithin and egg lecithin) is often used to manufacture liposomes, which is 
actually a mixture composed of various kinds of phospholipids though PC dominates. 
The long - chain fatty acids constituting phospholipids also have many types, 
such as lauric (C12), myristic (C14), palmitic (C16), and stearic (C18). In general, 
unsaturated fatty acids occur in natural phospholipids. Dimyristoyl phosphatidylcholine 
(DMPC), dipalmitoyl phosphatidylcholine (DPPC), distearoyl phosphatidylcholine 
(DSPC), and dipalmitoyl phosphatidylethanolamine (DPPE) are the most 
common synthetic phospholipids. The length of the lipid chain infl uences the gel – 
liquid crystalline phase transition temperature ( Tc ) of phospholipids, wherein longer 
chained lipids lead to higher Tc . For example, DPPC has a Tc of 41 ° C while DSPC 
has a Tc of 54 ° C [50] . 
Drug entrapment is an important parameter in manufacturing liposomes which 
is infl uenced by many factors: the types, molecular weights, and physicochemical 
properties of drugs; the types, sizes, and compositions of liposomes; and the manufacturing 
methods. In addition, entrapped drugs may leak during storage. Drugs may 
be entrapped in one of two parts of liposomes, the inner phase or bilayers, depending 
on the physicochemical property of the drugs. Water - soluble drugs prefer the 
aqueous inner phase while lipophilic drugs prefer the hydrophobic environment of 
bilayers. Macromolecules such as peptides and proteins can adsorb onto bilayers, 

wherein electrostatic interaction can infl uence entrapment. Those drugs insoluble 
in both water and oil are hard to entrap. Rather than common manufacturing 
approaches, more promising methods are adopted to improve drug entrapment. 
Ionic gradient methods can increase the entrapment effi cacy of some ionic drugs, 
including the pH gradient method [74] , the ammonium sulfate gradient method [75, 
76] , the acetate gradient method [77] , and the manganese ion gradient method [78] . 
Lyophilization of liposomes is a good preservation method which can prevent 
entrapped drugs from leaking, liposome precipitation and agglomeration due to 
gravity and thermal movement, and possible hydrolysis of phospholipids (resulting 
in production of toxic lyso - phospholipids). Generally, lipophilic drugs have a high 
entrapment effi cacy, though drug loading is limited, because they insert into bilayers 
tightly. Therefore, an effi cient method increasing entrapment effi cacy is to prepare 
the lipophilic derivatives of hydrophilic drugs [79] . 
The manufacturing of nanosized liposomes can be performed using the methods 
mentioned above. However, the small size of nanoliposomes is diffi cult to achieved 
by methods such as fi lm hydration. Molecular self - assembly occurs in the injection 
method, and then the size and morphology of obtained liposomes can be well controlled. 
In fact, liposomes that result from the injection method are uniform and 
small enough, to the nanoscale, and usually SUVs are obtained. Because of the very 
low toxicity of ethanol, the ethanol injection method is usually used and is described 
as follows to show the process of manufacturing liposomes [50] . A scale - up manufacturing 
process of the ethanol injection method has been established [80 – 82] . The 
obtained liposome size is mostly less than 300 nm: 
(a) Handling and storage of lipids is important. Store organic solutions of phospholipids 
in a sealed glass container layered with argon or nitrogen below 
. 20 ° C, preferably at . 78 ° C. When transferring a portion of the material, allow 
it to reach room temperature before opening the bottle. Saturated phospholipids, 
that is, lipids composed of completely saturated fatty acids, such as 
DPPC, are stable as powders. However, storage of these lipids as described 
above is highly recommended. Unsaturated phospholipids are extremely 
hygroscopic as powders, which will quickly absorb moisture and become 
gummy upon opening the storage container. Always dissolve such lipids in a 
suitable solvent (preferably chloroform) and store it in a glass container at 
. 78 ° C. 
(b) Prepare materials such as phospholipids, cholesterol, other additives, ethanol, 
injector, beaker, agitation machine, and evaporation device before manufacturing. 
Calculate the amount of these agents according to the request of the 
last products. A fi ne - gauge needle to a 1 - mL glass syringe is preferred. Dissolve 
lipid components (including lipophilic drugs) in ethanol. Dissolve water 
- soluble drugs in water or aqueous media as dispersing media. 
(c) Rapidly inject the ethanol solutions into agitated aqueous media with the 
tip under the surface. A homogeneous and almost transparent liquid will be 
obtained. Repeat this process and notice that the percentage of ethanol in the 
last product is not more than 7.5%. Collect all liquids and remove ethanol by 
evaporation, dialysis, or gel fi ltration. The last liposomal suspensions can be 
further concentrated through evaporating water. Sterilize them by autoclave. 
They may be lyophilized when needed. 
NANOTECHNOLOGY FOR DRUG DELIVERY 1261

1262 NANOTECHNOLOGY IN PHARMACEUTICAL MANUFACTURING 
Separation of nonentrapped drugs from liposomes (or purifi cation of liposomes) is 
an important process after manufacturing. The size difference between liposomes 
and unincorporated materials is the basis of separation. Gel chromatography, dialysis, 
and centrifugation are usual approaches. The drug entrapment percentage of 
liposomes can be obtained after separation. The whole drugs in liposomal suspensions 
or the entrapped drugs can be determined by dissolving liposomes with organic 
solvents or solubilizing liposomes with detergents to release drugs. The morphology 
of liposomes can be investigated by negatively stained transmission electron microscopy, 
cryo - electron microscopy, or freeze - fracture electron microscopy. The size 
distribution of liposomes is usually analysized by photon correlation spectroscopy 
(laser light scattering) [50] . 
Beyond conventional liposomes, functional liposomes are designed to achieve 
various therapeutic effects. Conventional liposomes manufactured by natural phospholipids 
or commonly used synthetic phospholipids such as PC and PE are negatively 
charged. However, cationic liposomes can form complexes with peptides or 
nucleic acids through electrostatic interaction and prefer to adsorb onto the surfaces 
of cell membranes, subsequently improving interaction with cells and penetrating 
into cytosol or phagocytosis. Therefore, cationic liposomes have become a standard 
transfection agent in cell manipulation [83] . Furthermore, they become primary 
nonviral gene delivery carriers [84] . 
Liposomes show site - specifi c distribution in the MPSs after intravenous (IV) 
administration due to opsonization by the complement system [85] . The diseases in 
MPSs can benefi t from the drug targeting. But this is a bad result for diseases in 
other tissues. Long - circulating liposomes are then developed for targeting to non - 
MPS tissues. The long - circulating effect results from hydrophilic polymers coated 
on liposomes. For example, the half - life of the long - circulating liposomes can be 
extended to 20 h in rat. They are also called sterically stable liposomes or Stealth 
liposomes. The lipid conjugate of PEG, PEG – DSPE, is commonly used and inserts 
into bilayers and hinders plasma protein adsorption. The enhanced permeability and 
retention (EPR) effect of solid tumors makes long - circulating liposomes a very 
useful tool for anticancer therapy [86] . However, in recent years it was reported that 
in most cases PEGylated liposomes were cleared very rapidly from circulation with 
repeated injection. But doxorubicin PEGylated liposome is an exception. The production 
of anti - PEG immunoglobulin (Ig) M following injection is the major reason, 
and the spleen also plays a key role [87] . However, a more recent case has appeared. 
A modifi ed phospholipid – methoxy(polyethylene glycol) conjugate was recently 
synthesized through the methylation of phosphate oxygen moiety which could 
prevent PEGylated liposomes from being activated by a complement system in vivo 
followed by achieving a true long - circulating effect [88] . 
Other functional liposomes are mainly stimuli - responsive liposomes. The 
pH - sensitive liposomes contain pH - sensitive lipids such as 1,2 - dioleoyl - sn - 3 - 
phosphatidylethanolamine (DOPE) showing an inverted hexagonal confi guration 
in a low - pH environment and release entrapped drugs in the low - pH environment 
of tumor tissues due to liposomal membrane destabilization [89] . Temperature - sensitive 
liposomes are prepared from special lipids such as DPPC whose phase transition 
temperature ( Tc = 41 ° C) is proper to perform clinical anticancer therapy. When 
up to Tc , the fl uidity of liposomal membranes increases sharply, followed by 

entrapped drugs releasing [90] . Some thermosensitive polymers can also be used to 
manufacture temperature - sensitive liposomes [91] . Magnetoliposomes load ultra- 
fi ne magnetite, preferring to accumulate in the local tissue within the magnetic fi eld 
[92] . Immunoliposomes load attached monoclonal antibodies to treat some severe 
diseases such as cancer [93] . 
Liposomes have been successfully applied to many drugs, diagnostic agents, 
imaging agents, transfection agents, vaccines, and so on. Liposomes have been tried 
in almost all routes of administration: oral, injection (intravenous, subcutaneous, 
intramuscular, intra - articular cavity, and other possible injection sites), intranasal, 
pulmonary inhalation, conjunctiva, topical, and transdermal. The most signifi cant 
application fi eld of liposomes is still anticancer therapy. After a long - time research 
for 30 years, some liposomal products have reached the market (Table 3 ). The major 
problems in manufacturing liposomes are scale - up production, effi cient sterilization, 
and stable storage. 
TABLE 3 Liposomal Drugs Approved for Clinical Application 
Drug Product Name 
Composition of 
Liposomes and 
Other Major 
Excipients Indication Company 
Daunorubicin DaunoXome DSPC , cholesterol Kaposi ’ s 
sarcoma 
Gilead Sciences 
Doxorubicin Mycet Egg PC, 
cholesterol 
Combinational 
therapy of 
recurrent 
breast cancer 
Zeneus 
Doxorubicin Doxil/Caelyx MPEG – DSPE, 
HSPC, 
cholesterol, 
ammonium 
sulfate, sucrose, 
histidine 
Refractory 
Kaposi ’ s 
sarcoma; 
ovarian 
cancer; 
recurrent 
breast cancer 
Alza/SP Europe 
Amphotericin 
B 
AmBisome 
(lyophilized 
product) 
HSPC, cholesterol, 
DSPG, . - 
tocopherol, 
sucrose, 
disodium 
succinate 
Fungal 
infections 
Gilead Sciences 
Cytarabine DepoCyt DOPC, DPPG, 
cholesterol, 
triolein 
Lymphomatous 
meningitis 
SkyePharma 
Morphine DepoDur DOPC, DPPG, 
cholesterol, 
tricaprylin, 
triolein 
Pain following 
major 
surgery 
SkyePharma 
MPEG = methyl PEG; HSPC = hydrogenated soy phosphatidylcholine; DSPG = disteroylphosphatidylglycerol; 
DOPC = dioleoylphosphatidylcholine; DPPG = dipalmitoylphosphatidylglycerol. 
NANOTECHNOLOGY FOR DRUG DELIVERY 1263

1264 NANOTECHNOLOGY IN PHARMACEUTICAL MANUFACTURING 
Nanoparticles Nanoparticles attract much attention of pharmaceutical scientists 
because of their controllable manufacturing, uniform preparations, and low cost. 
The major difference between nanoparticles and liposomes is that the former has a 
solid core while the latter only has inner aqueous phase and thin bilayers. In addition, 
in the case of liposomes, the entrapped water - soluble drugs exist only in solutions 
of the inner phase, while lipophilic drugs are only limited in the small space 
of bilayers. Therefore, the drug - loading effi ciency (drug – lipid ratio) of liposomes is 
always limited. In the case of nanoparticles, drugs exist in the solid state, and high 
drug loading is possibly achieved. Unlike liposomes, nanoparticles may be composed 
of various materials, and biodegradable materials are preferably used. Furthermore, 
modifi ed materials based on traditional natural and synthetic materials 
are also frequently used to manufacture nanoparticles to achieve more functions, 
which then highly enlarges the lists of used materials. In addition, more manufacturing 
methods of nanoparticles are optional than liposomes. Therefore, nanoparticles 
are relatively ideal nanocarriers for most drugs. 
Nanoparticles can be classifi ed as three types, polymeric nanoparticles, lipid 
nanoparticles, and inorganic nanoparticles, depending on the major components. 
Polymeric nanoparticles are exploited earlier, while lipid nanoparticles are of great 
interest in recent years due to very good biocompatibility. The development of 
polymeric nanoparticles is highly related to polymer science. Besides a great deal 
of natural polymers, more and more biodegradable polymers are synthesized, which 
allows pharmaceutical scientists to have enough optional subjects. Solid lipid 
nanoparticles (SLNs) composed of solid lipids have a profound advantage of no 
biotoxicity [94] . Inorganic nanoparticles are currently exploited only a little [45 – 47] , 
and the major problems may be their poor biodegradability and relatively low drug - 
loading effi ciency. 
Polymeric Nanoparticles Polymeric materials for manufacturing nanoparticles 
include synthetic poly(lactic acids) (PLA), poly(lactide - co - glycolide) (PLGA), 
poly(. - caprolactone) (PCL), poly(methyl methacrylates), and poly(alkyl cyanoacrylates); 
natural polymers (albumin, gelatin, alginate, collagen), and modifi ed 
natural polymers (chitosan, starch). Polyesters, alone and in combination with other 
polymers, are most commonly used for the formulation of nanoparticles. PLGA 
and PLA are highly biocompatible and biodegradable. They have been used since 
the 1980s for numerous in vivo applications (biodegradable implants, controlled 
drug release). The U.S. Food and Drug Administration (FDA) has approved PLGA 
for human therapy [95] . More recently, formulations based on natural polymers 
have been developed and are on the market. For example, a wonderful natural 
polymer, chitosan, has permeability enhancer abilities, allowing the preparation of 
organic solvent free mucoadhesive particles [42] . 
Nanoparticles of synthetic polymers are usually manufactured by dispersion of 
preformed polymers. Although many methods can be used, they may be classifi ed 
as monomer polymerization, nanoprecipitation, emulsion diffusion/solvent evaporation, 
and salting out. An appropriate method is selected mainly depending on 
polymer and drug natures. Polymerization of polymer monomers has been developed 
usually using poly(alkyl cyanoacrylate) [96, 97] . Organic solvents are usually 
used in polymerization. A detailed description of this method is not provided 
here. 

The nanoprecipitation method is commonly adopted to entrap lipophilic drugs, 
and low polydispersity is probably achieved [42] . In general, the organic solution 
containing drugs and polymers is added a nonsolvent to lead to polymers precipitating 
together with drugs. The size of formed nanoparticles can be adjusted by the 
polymer and nonsolvent amounts in the organic phase. Nanoparticles can be separated 
from solvents and unincorporated drugs with centrifugation followed by spray 
drying or freeze drying when needed. The stability and drug recovery yield of 
nanoparticles depend on the ratio of drugs to polymers [98] . Recently, this technique 
has also been used to entrap hydrophilic compounds into PLGA and PLA 
nanoparticles [99, 100] , especially peptides and proteins [101] . 
Another common method to manufacture polymeric nanoparticles is the emulsion 
diffusion or solvent evaporation technique, which is used to entrap hydrophobic 
or hydrophilic drugs. Generally, the polymer and hydrophobic drugs are dissolved 
in a partially water miscible organic phase (e.g., benzyl alcohol, propylene carbonate, 
and ethyl acetate). The organic solution is emulsifi ed in aqueous media containing 
a suitable surfactant [i.e., anionic sodium dodecyl sulfate (SDS), nonionic 
poly(vinyl alcohol) (PVA) or cationic didodecyl dimethyl ammonium bromide 
(DMAB)] under stirring. The diffusion of the organic solvent and the counterdiffusion 
of water into the emulsion droplets induce polymeric nanoparticle formation. 
The organic solvent is evaporated. Also hydrophilic drugs could be entrapped into 
a water - in - oil (W/O) emulsion containing polymers and then undergo the above 
process. Then a water - in - oil - in - water (W/O/W) emulsion is obtained. After evaporation 
of total organic solvent, the drug - loaded nanoparticles can be separated. 
Polymer nature, polymer concentration, solvent nature, surfactant molecular mass, 
viscosity, phase ratio, stirring rate, temperature, and fl ow of water all affect nanoparticle 
size [102] . 
The salting - out method is also used. Polymers are dissolved in water - miscible 
organic solvents such as acetone or tetrahydrofuran (THF). The organic phase is 
emulsifi ed in an aqueous phase that contains the emulsifi er and salts of high concentration. 
Typically, the salt solution used contains 60% (w/w) magnesium chloride 
hexahydrate or magnesium acetate tetrahydrate with a polymer - to - salt ratio of 1 : 3. 
In contrast to the emulsion diffusion method, no diffusion of solvents occurs due 
to the presence of high concentrated salts. The fast addition of pure water to the 
O/W emulsion under mild stirring reduces the ionic strength and leads to the migration 
of the organic solvent to the aqueous phase, inducing nanoparticle formation. 
The fi nal step is purifi cation by cross - fl ow fi ltration or centrifugation to remove the 
salting - out agent. Common salting - out agents are electrolytes (sodium chloride, 
magnesium acetate, or magnesium chloride) or nonelectrolytes, such as sucrose. 
Polymer concentration and molecular weight, stirring rate and time, and the nature 
and concentration of surfactant and solvent are all important parameters. This 
method would allow avoiding the use of organic chlorinated solvents and large 
amounts of surfactants [102] . Furthermore, formulation of nanoparticles with 
natural polymers is performed by ionic gelation (chitosan), coacervation (chitosan, 
gelatin), and desolvation (gelatin) [102, 103] . These mild methods have the advantage 
of producing organic solvent - free formulations. 
Additional advantages can be obtained by changing nanoparticle surface properties, 
for example, good stability, mucoadhesion, and long circulation time. 
For example, the in vivo long - circulating effect is achieved either by coating 
NANOTECHNOLOGY FOR DRUG DELIVERY 1265

1266 NANOTECHNOLOGY IN PHARMACEUTICAL MANUFACTURING 
nanoparticle surfaces with hydrophilic polymers/surfactants or by incorporating biodegradable 
copolymers containing a hydrophilic moiety. Like long - circulating liposomes, 
PEG - containing polymers are frequently used to manufacture long - circulating 
nanoparticles. PEGylated copolymers (PLA – PEG, PLGA – PEG and PCL – PEG) are 
used [42, 104] , and the long - circulating effect also results from the adsorption or covalent 
conjugation of some hydrophilic polymers with the hydrophobic surface of 
nanoparticles [105, 106] . Moreover, the active targeting of nanoparticles can be 
achieved by incorporating the conjugate of the polymer and target - directed molecule, 
such as (Arg - Gly - Asp) RGD , (trans - activator transcription) TAT peptides [107] , and 
monoclonal antibody [108] . Recently, self - assembled nanoparticles have aroused 
great interest, consisting of amphiphilic macromolecules, such as hydrophobically 
modifi ed glycol chitosan, which can also entrap drugs or peptides [109] . 
Before nanocarriers go into clinical applications, some issues must be considered, 
including drug - loading capacity, possibility of drug targeting, in vivo fate of the 
carrier (interaction with the biological surrounding, degradation rate, accumulation 
in organs), acute and chronic toxicity, scaling up of production, physical and chemical 
storage stability, and overall costs. A certain advantage of polymer systems 
is the wealth of possible chemical modifi cations. Possible problems of polymeric 
nanoparticles derive from residues of organic solvents used in the production 
process, polymer cytotoxicity, and the scaling up of production. Polymer hydrolysis 
during storage has to be taken into account and lyophilization is often required to 
prevent polymer degradation [94] . 
Solid Lipid Nanoparticles The outstanding advantage of lipid nanoparticles is 
perfect biocompatibility because their raw materials are the components of our 
body, preferring to be used or degraded by the body. Solid lipids are usually used 
as the major component of lipid nanoparticles — hence the name solid lipid nanoparticles. 
However, the used solid lipids generally become liquid at a high temperature 
to adapt to the preparation of SLNs. Compared with polymeric nanoparticles, the 
materials used for SLNs are simpler. The frequently used lipids are glycerides 
of various fatty acids, which also exist in the emulsions for parenteral nutrition. 
Large - scale production of SLNs can be achieved in a cost - effective and relatively 
simple way using high - pressure homogenization and microemulsion. Another useful 
method is solvent emulsifi cation/evaporation. The SLN introduced in 1991 represents 
an alternative carrier system to traditional colloidal carriers, such as liposomes 
and polymeric nanoparticles. SLNs combine advantages of the traditional systems 
but avoid some of their major disadvantages [27] . The proposed advantages of SLNs 
include [94] : 
• Possibility of controlled drug release and drug targeting 
• Increased drug stability 
• High drug payload 
• Incorporation of lipophilic and hydrophilic drugs feasible 
• No biotoxicity of the carrier 
• Avoidance of organic solvents 
• No problems with respect to large - scale production and sterilization 

Solid lipids, emulsifi ers, and water are generally the ingredients involved for 
manufacturing SLNs. The term lipids is used in a broader sense and includes triglycerides 
(e.g., stearin), partial glycerides (e.g., Imwitor), fatty acids (e.g., stearic 
acid), steroids (e.g., cholesterol), and waxes (e.g., cetyl palmitate). All categories of 
emulsifi ers may be used to stabilize the lipid dispersion, and the combination 
of emulsifi ers prevents particle agglomeration more effi ciently. The choice of the 
emulsifi er depends on the administration route and is more limited for parenteral 
administration. 
High - pressure homogenization (HPH) has emerged as a reliable and powerful 
technique for the preparation of SLNs. Homogenizers of different sizes are commercially 
available from several manufacturers at reasonable prices. In contrast 
to other techniques, scaling up of HPH is out of the question in most cases. High - 
pressure homogenizers push a liquid with high pressure (100 – 2000 bars) through a 
narrow gap (in the range of a few micrometers). The fl uid accelerates over a very 
short distance to very high velocity (over 1000 km/h). Very high shear stress and 
cavitation forces disrupt the particles down to the submicrometer range. Typical 
lipid contents are 5 – 10%, though higher lipid concentrations (up to 40%) may be 
used. Two general approaches of HPH, hot and cold homogenization, can be used 
for manufacturing SLNs (Figure 4 ) [27] . 
Microemulsions (transparently appearing with droplet size less than 100 nm) are 
thermodynamically stable systems, and the choice of optimal formulation containing 
oil, surfactant, cosurfactant, and oil – water ratio is key [110] . Generally, the solid 
lipid of low melting point (e.g., stearic acid) melts at a high temperature (e.g., 65 – 
70 ° C), and then hot microemulsions are prepared using it. SLNs can be obtained 
after the hot microemulsions are rapidly cooled by injecting them into cold water 
(e.g., 2 ° C) under stirring. Emulsifi ers in a formulation typically include Tween 
20/60/80, lecithin, and sodium taurodeoxycholate, and coemulsifi ers include alcohols 
and sodium monooctylphosphate. The typical volume ratios of hot microemulsions 
to cold water are from 1 : 25 to 1 : 50. The very low solid concentration of SLN 
suspensions is the disadvantage of the microemulsion method. Rapid temperature 
decrease in hot microemulsions is key to obtaining homogeneous and small - sized 
nanoparticles. A high temperature gradient can also ensure rapid lipid crystallization 
and prevent aggregation [94] . 
The solvent emulsifi cation/evaporation method involves lipid precipitation in 
O/W emulsions. Solid lipids are dissolved in a water - immiscible organic solvent 
(e.g., cyclohexane) followed by emulsifi cation in an aqueous medium. Upon evaporation 
of the solvent, the nanoparticle dispersion is formed due to lipid precipitation. 
Residue of organic solvents is the major problem of this method [94] . However, 
the microemulsion and solvent emulsifi cation/evaporation methods can be performed 
conveniently in the laboratory without specifi c apparatuses. 
During research of SLNs, some problems have continually appeared, for example, 
very low drug loads, drug expulsion during storage, and high water content of SLN 
dispersions. The . and .. crystallines of higher energy state mainly appear in conventional 
SLNs manufactured by hot - homogenization technique. However, these 
crystallines prefer to transform to the more ordered . modifi cation of low energy 
state during storage. The high ordered degree improves the crystal imperfections, 
diminishing further to lead to drug expulsion. To solve this problem, a new kind of 
lipid nanoparticle was developed, called a nanostructured lipid carrier (NLC). NLCs 
NANOTECHNOLOGY FOR DRUG DELIVERY 1267

1268 NANOTECHNOLOGY IN PHARMACEUTICAL MANUFACTURING 
are composed of spatially very different lipid molecules, that is, solid lipids and 
liquid lipids (oils). The matrix remains solid at body temperature though its melting 
point is lower than one of the original solid lipids. No crystallization happens in 
NLCs so that the drug loads can be increased and the expulsion during storage is 
avoided [111] . 
New functions can be obtained by modifi cations of SLNs. Incorporation of Tween 
80 and Poloxamer 188 can stabilize SLNs to achieve long - circulating or crossing 
blood – brain barrier effects [112] . Recently, novel nanoparticles called polymer – 
lipid hybrid nanoparticles (PLNs) were developed [113] . They can entrap cationic 
anticancer agents (e.g., doxorubicin) effectively by incorporation of an anionic 
lipophilic polymer into lipids to treat multidrug - resistant (MDR) cancers. 
In general, SLNs are used to entrap hydrophobic drugs due to their lipid nature, 
but a few reports show that hydrophilic drugs can also be entrapped. A hydrophilic 
peptide, gonadorelin, and monostearin were dissolved in acetone and ethanol at 
50 ° C followed by pouring the resultant organic solution into an aqueous medium 
containing 1% PVA under agitation to obtain peptide - loaded SLNs that were sub- 
FIGURE 4 High - pressure homogenization for manufacturing SLNs. 
Melt lipid and dissolve/disperse 
drugs in lipid 
Hot homogenization technique Cold homogenization technique 
Disperse 
drug-loaded lipid in 
hot surfactant aqueous 
solution 
Mix thoroghly using 
stirrer to form coarse 
preemulsions 
High-pressure 
homogenization at 
temperature above 
melting point of 
lipid to form hot o/w 
nanoemulsions 
Solidification of 
nanoemulsions by 
cooling down to room 
temperature
Solid lipid nanoparticles (SLNs) 
High-pressure 
homogenization at 
room temperature or 
below 
Disperse powder 
in surfactant 
aqueous solution rind 
in powder mill 
(50–100 .m) 
Grind in powder 
mill (50–100 .m) 
Solidification of 
drug-loaded lipid in 
liquid nitrogen or dry 
ice

sequently separated by centrifugation. Up to 69% of gonadorelin was incorporated. 
The in vitro release of gonadorelin from SLNs was slow [114] . The W/O/W multiple - 
emulsion technique was also used to manufacture peptide - loaded SLNs. Insulin is 
a model peptide located in the inner water phase of the W/O/W emulsion, tripalmitin 
is the core of SLNs, and the surfaces are modifi ed with PEG 2000 – stearate. The 
insulin - loaded SLNs show good stability upon the low pH of the gastric medium 
and the pancreatic enzymes in intestinal medium [115] . 
Perspective of Nanoparticles As drug nanocarriers, nanoparticles have unique 
advantages: for example, high dispersing, adhesive property, targeting in vivo. Like 
liposomes, anticancer therapy is a major function of nanoparticles [116] . Easy modi- 
fi cation of nanoparticles also makes them platforms to perform more functions, for 
example, delivering drugs across the blood – brain barrier (BBB) [117] , lymphatic 
targeting [118] , and gene delivery [119] . 
Abraxane is a successful paradigm of nanoparticle application. It is an albumin 
nanoparticle loading paclitaxel developed by American Pharmaceutical Partners 
(APP) and American BioScience. The outstanding advantage of Abraxane is no 
signifi cant side effects, not like the traditional paclitaxel preparation with Cremophor 
EL (polyethoxylated castor oil) and ethanol. More nanoparticle products will 
reach the market in the future. 
Other Nanocarriers 
Nanoemulsions Lipid nanoemulsions were introduced in the 1950s as parenteral 
nutrition. Vegetable oils (e.g., soy oil) or middle - chain triglycerides are used, typically 
occupying 10 – 20% of the emulsion. Other ingredients include phospholipids 
as stabilizers and glycerol as osmolar regulation agent. In recent years this system 
has been further developed to load lipophilic drugs and several formulations 
are commercialized. Examples are etomidate (Etomidat - Lipuro), diazepam 
(Diazepam - Lipuro and Stesolid), propofol (Disoprivan), and dexamethasone palmitate 
(Lipotalon). In comparison to previous, solubilization - based formulations of 
these drugs, reduction of the local and systemic side effects (e.g., pain during injection) 
has been achieved. The possibility of controlled drug release from nanoemulsions 
is restricted due to the small size and the liquid state of the carrier. Most drugs 
show a rapid release from them. Advantages of nanoemulsions include toxicological 
safety and a high content of the lipid phase as well as the possibility of large - scale 
production by high - pressure homogenization [94] . 
Microemulsions Microemulsions are nanoemulsions which are optically isotropic, 
transparent or translucent, low - viscous, and thermodynamically stable liquid solutions, 
mainly containing tiny liquid droplets less than 100 nm. The manufacturing 
of microemulsions as a self - formed system is relatively simple. They are bicontinuous 
systems essentially composed of water and oil with surfactant and cosurfactant 
separating. A very low interfacial tension to 0 mN/m is found in microemulsions 
despite the large oil – water interfacial areas. A prominent example is the Sandimmun 
Optoral/Neoral preconcentrate for microemulsions. Now microemulsions are 
usually limited to dermal and peroral applications due to their high surfactant 
content. Because they only exist in narrow regions of phase diagrams, they are very 
restricted in tolerance to quantitative formulation changes [120] . 
NANOTECHNOLOGY FOR DRUG DELIVERY 1269

1270 NANOTECHNOLOGY IN PHARMACEUTICAL MANUFACTURING 
Polymeric Micelles Polymeric micelles composed of amphiphilic copolymers, that 
is, polymers consisting of a hydrophobic block and a hydrophilic block, are gaining 
increasing attention. They show high stability both in vitro and in vivo and good 
biocompatibility, and more importantly they can solubilize a broad variety of poorly 
soluble drugs in their inner core. Many of these drug - loaded micelles are currently 
at different stages of preclinical and clinical trials. Due to their hydrophilic shell 
and small size, they prefer to exhibit prolonged circulation times in vivo and can 
accumulate in tumor tissues. Polymeric micelles are formed by block copolymers 
consisting of hydrophilic and hydrophobic monomer units with the length of a 
hydrophilic block exceeding to some extent that of a hydrophobic one. If the length 
of a hydrophilic block is too high, copolymers exist in water as unimers (individual 
molecules), while molecules with very long hydrophobic block prefer to form structures 
with nonmicellar morphology, such as rods and lamellae. Diblock copolymers 
with an A – B structure and tri - or multiblock copolymers such as poly(ethylene 
oxide) – poly(propylene oxide) – poly(ethylene oxide) (PEO – PPO – PEO) (A – B – A) 
may self - organize into micelles. The amphiphilic copolymers commonly have 
the critical micelle concentration (CMC) values as low as 10 . 6 M , which is about 
two orders of magnitude lower than that of such surfactants as Tween 80. As potential 
drug carriers, the hydrophobic core of polymeric micelles generally consists of 
a biodegradable polymer such as poly( . - benzyl - l - aspartate) (PBLA), PLA, or PCL 
which serves as a reservoir for an insoluble drug, protecting it from contact with the 
aqueous environment. The core may also consist of a water - soluble polymer [e.g., 
poly(aspartic acid; P(Asp)], which is rendered hydrophobic by the chemical conjugation 
of a hydrophobic drug or is formed through the association of two oppositely 
charged polyions (polyion complex micelles). Another special group of polymeric 
micelles is formed by lipid - core micelles, that is, micelles formed by conjugates of 
soluble copolymers with lipids (e.g., PEG – PE) [35, 36] . 
Three methods are used to manufacture polymeric micelles: dialysis, emulsifi cation, 
and fi lm methods. In the dialysis method, the drug and copolymer are dissolved 
together in a water - miscible solvent (e.g., ethanol) followed by dialysis against 
water. During the process (possibly several days), the insoluble drugs are incorporated 
into the formed micellar core. In the emulsifi cation method, an O/W emulsion 
is fi rst prepared using an aqueous solution of the copolymer and the drug solution 
in a water - insoluble volatile solvent (e.g., chloroform). The drug - loaded micelle is 
formed as solvent evaporation. In the fi lm method, the copolymer solution and the 
drug solution are dissolved separately in miscible volatile organic solvents and are 
mixed followed by evaporating solvents to form a polymer/drug fi lm. The fi lm is 
hydrated in water or buffers, and then the micelle is formed by intensive shaking. 
If the amount of a drug exceeds the solubilization capacity of micelles, the excess 
drug precipitates in a crystalline form and is removed by fi ltration. The loading 
effi ciency for different compounds varies from 1.5 to 50% by weight. The major 
driving force behind self - association of amphiphilic polymers is the decrease of free 
energy of the system due to removal of hydrophobic fragments from the aqueous 
surroundings with the formation of micelle core stabilized by hydrophilic blocks 
exposed to water [35, 36] . 
Various drugs, for example, diazepam and indomethacin, doxorubicin, anthracycline 
antibiotics, and polynucleotides, were effectively solubilized in polymeric 
micelles. Also polymeric micelles can carry various reporter (contrast) groups and 

become the imaging agents. Besides targeted drug delivery due to the EPR effect 
of tumor, specifi c polymeric micelles having stimuli - responsive amphiphilic block 
copolymers, targeting ligand molecules, or monoclonal antibody molecules are also 
manufactured [35, 36] . 
Nanogels Nanogels are colloidal stable particles made from hydrogels with 
nanosized hydrophilic polymeric networks. Hydrogels are the simple gels swelling 
strongly in aqueous media, typically composed of hydrophilic polymer components 
cross - linked into a network by either covalent (chemical cross - linking) or noncovalent 
(physical cross - linking) interactions. It is the cross - linking that provides for 
dimensional stability, while the high solvent content gives rise to the fl uidlike transport 
properties. Cross - links are important to maintain the network structure of the 
hydrogels and prevent dissolution of the hydrophilic chains [121] . 
Two methods, emulsifi cation – evaporation and the micelle/nanoparticle approach, 
are used to manufacture nanogels. In the former method, bis - activated PEG in 
dichloromethane is added dropwise to the aqueous solution of polyethylenimine 
(PEI) and then sonicated. The resulting white emulsion is evaporated in vacuum, 
producing a clear, slightly opalescent solution. This solution is stirred for less than 
one day at room temperature and much debris is separated by centrifugation. The 
nanogel suspension is obtained after dialysis against water [51] . This procedure is 
convenient except for using organic solvents, a vacuum evaporation step, and the 
obtained particles with a wide size distribution. Another method involves surface 
preactivated micelles or nanoparticles followed by reaction with other polymers on 
the surface in aqueous media. None of the organic solvents involved are of benefi t. 
For example, a Pluronic block copolymer both ends of which are activated by 1,1 . - 
carbonyldiimidazole is dissolved in water at a concentration above its CMC. A 
diluted aqueous solution of PEI is then added dropwise to the micellar solution, 
stirring overnight. During this procedure a covalently linked cationic polymer PEI 
layer is formed on the Pluronic micelles, producing nanogels with narrow size distribution. 
After dialysis the resulting nanogel suspension can be further lyophilized. 
Using this procedure, the nanogels based on Pluronic P85/PEG and F127/PEG are 
obtained with fi nal yields of 55 and 70% by weight and average hydrodynamic 
diameters of 100 and 180 nm, respectively [20, 52] . 
Many drugs can be entrapped into nanogels, for example, valproic acid, nucleoside 
analogues, antisense oligonucleotides, adenosine triphosphate (ATP), and small 
interfering ribonucleic acid (siRNA). Because macromolecular drugs such as peptides 
and proteins need to locate in a hydrophilic environment to maintain their 
activity, the particular hydrophilic property of nanogels would be of benefi t. Special 
functions such as cellular targeting, crossing the BBB, and controlled release may 
also be achieved by the nanogel technique. In addition, the nanogel materials should 
be biodegradable. In cationic nanogels, PEI and PEG are cross - linked via urethane 
bonds, usually considered as stable links. However, due to the presence of highly 
protonated PEI, hydrolysis of these bonds was signifi cantly accelerated, and the 
polymer network of nanogels could rapidly degrade in aqueous solution at the 
physiological pH during a period of about two weeks [20] . 
Dendrimers Dendrimers attracted much attention after they were fi rst investigated 
by Tomalia 20 years ago [122, 123] , and they have become the star molecules 
NANOTECHNOLOGY FOR DRUG DELIVERY 1271

1272 NANOTECHNOLOGY IN PHARMACEUTICAL MANUFACTURING 
in recent years. Dendrimers possess perfect nanoarchitectures from 1 nm to more 
than 10 nm, consisting of repetitive chemical moieties with tree architecture. According 
to repetitive folds, dendrimers with the same basic cores are divided into a series 
of generations. The higher generation of dendrimers represents more repetitive 
units. Dendrimers are hyperbranched macromolecules that can be subdivided into 
three architectural parts: (a) the multivalent surface, with a high number of potential 
reactive sites; (b) the “ outer shell ” just beneath the surface, having a well - 
defi ned microenvironment protected from the outside by the dendrimer surface; 
and (c) the core, which in higher generation dendrimers is protected from the surroundings, 
creating a microenvironment surrounded by dendritic branches. Therefore, 
the interior of dendrimers is well suited for entrapment of guest molecules. 
The multivalent surfaces on a higher generation dendrimer can contain a very high 
number of functional groups. This makes the dendritic surfaces and outer shell well 
suited to host – guest interactions. Dendrimers can be tailored specifi cally for the 
desired purposes, for example, as dendritic sensors, drug vehicles, or even drugs 
[23] . 
Dendrimers are synthesized through a stepwise repetitive reaction sequence, 
wherein a convergent or divergent approach is used. On the one hand, the most 
divergent dendrimer syntheses require excess monomer loading and lengthy chromatographic 
separations, particularly at higher generations. On the other hand, 
convergent synthesis strategies are generally limited to the construction of only 
lower generation dendrimers due to the nanoscale steric issues that are encountered 
when attaching the dendrons to the molecular - level core [124] . Currently much of 
the work on dendrimers has been based on the commercially available Starburst 
poly(amidoamine) (PAMAM) dendrimers that are extensively studied as drug carriers. 
PAMAM may be synthesized from an ammonia or ethylenediamine core 
(EDA) by the divergent approach, involving Michael addition followed by amidation 
with methyl acrylate and resulting in the production of a dendrimer family 
(G = 0 – 7), and half - generation dendrimers are carboxyl terminated and full - generation 
dendrimers are amine terminated (e.g., G = 5.0, 5.3 nm in size) [122, 123] . 
Dendrimers have been evaluated as drug nanocarriers, gene transfection agents 
imaging agents, and nanodrugs [124] . Also many surface - modifi ed dendrimers have 
been synthesized to obtain more functions such as active targeting and gene delivery. 
Dendrimers may be used as drugs for antibacterial and antiviral treatment and 
as antitumor agents. VivaGel, a topical water - based gel based on sulfonated naphthyl 
- modifi ed poly(lysine) dendrimers, has been evaluated against human immunodefi 
ciency virus (HIV) and other sexually transmitted diseases (STDs). The cationic 
dendrimers prefer to destabilize cell membranes and cause cell lysis and the cytotoxicity 
is generation dependent with higher generation dendrimers being the most 
toxic. The degree of substitution as well as the type of amine functionality is important, 
with primary amines being more toxic than secondary or tertiary amines. 
Another common dendrimer, poly(propylenimine) (PPI), shows similar behavior. 
However, anionic dendrimers show signifi cantly lower cytotoxicity than cationic 
ones. PEG or fatty acid surface - modifi ed dendrimers can reduce the cytotoxicity of 
cationic dendrimers [124] . 
Carbon Nanotubes, Fullerenes, and Quantum Dots Carbon nanotubes and fullerenes 
are carbon - based nanomaterials, and quantum dots are semiconductor nano

crystals. All of them show hydrophobic property. The possibility of cytotoxicity of 
these materials with inorganic nature should not be ignored although low toxicity 
is shown [25, 40, 125, 126] . However, these seemingly good results may be partly 
attributed to their poor solubility in polar solvents, which subsequently makes 
investigation of their biological properties diffi cult. They hardly load any drugs 
unless the surface is modifi ed hydrophilically. Because these nanomaterials are 
mainly produced in laboratories with the special devices, their modifi cations and 
subsequent pharmacological investigations are limited. However, a number of functional 
derivatives have been synthesized, and it is found that the modifi ed products 
have potent and selective pharmacological effects on organs, cells, enzymes, and 
nucleic acids [25, 37, 57] . 
7.2.3.2 Nanosuspensions 
Nanosuspensions of drugs are submicrometer colloidal dispersions of pure particles 
of drug which are stabilized by surfactants. A surprisingly large proportion of new 
drug candidates emerging from drug discovery programs are water insoluble, and 
therefore poorly bioavailable, leading to development efforts being abandoned. 
More than 40% of active substances during formulation development by the pharmaceutical 
industry are poorly water soluble. A substantial factor that prevents the 
development of such substances is the limited dissolution rate. Nanosuspensions are 
promising in addressing these so - called brickdust candidates. During the process of 
overcoming issues involving solubility, the additional pharmacokinetic benefi ts of 
drugs formulated in nanosuspensions come to be appreciated [19, 61] . 
Nanosuspensions can be used for those water - insoluble and oil - soluble compounds 
(high log P ), although other lipidic carriers such as liposomes and emulsions 
can be used to formulate these compounds as well. However, nanosuspensions can 
be used to address other problems, such as compounds that are insoluble in both 
water and oil. Nanosuspensions can maintain the drug in a preferred crystalline 
state of size suffi ciently small for pharmaceutical acceptability. For reasons of convenience 
to the patients, aqueous nanosuspensions can also be transformed to 
tablets or capsules after spray drying or freeze drying. Moreover, utilization of the 
dense, solid state confers an additional advantage of higher mass per volume loading. 
This is crucial when high dosing is required, for example, low - volume intramuscular 
and ophthalmic applications. Conventional approaches often attempt to solubilize 
insoluble drugs with the use of excessive amounts of cosolvents, but this often brings 
problems of toxicity. Besides, very large doses of drugs must be administered to 
animals when acute toxicity is investigated in preclinical research. As a result, the 
interference of toxic side effects caused by cosolvents cannot be ignored if using 
them [19] . 
Nanosuspensions are not nanocarriers so that what is emphasized during manufacturing 
is not materials but the manufacturing techniques. Only drugs and stabilizers 
(usually surfactants) participate in manufacturing nanosuspensions so that the 
process may be simple depending on drug instincts but sometimes it is not easy. 
The bottom - up and top - down approaches may be used in manufacturing nanosuspensions 
depending on drug nature and in - house devices. 
Antisolvent precipitation is a bottom - up method wherein two phases are involved: 
the initial creation of crystal nuclei of drugs and the subsequent growth. Formation 
NANOTECHNOLOGY FOR DRUG DELIVERY 1273

1274 NANOTECHNOLOGY IN PHARMACEUTICAL MANUFACTURING 
of a stable suspension with the smallest particle size requires a high nucleation rate 
but low growth rate. Both process rates are dependent on temperature and supersaturation. 
The optimum temperature for nucleation might lie below that for growth. 
A high - supersaturation condition is achieved by adding small amounts of a water - 
miscible organic solution of the drug to the nonsolvent (water) under rapid mixing, 
which leads to spontaneous nucleation. At high - supersaturation levels, the crystal 
habit or external appearance is changed to a needlelike or dendritic morphology. 
These crystals are easily broken, forming new smaller nuclei. Rapidly grown crystals 
tend to be more imperfect and often incorporate impurities and dislocations. This 
effect is more pronounced for fl exible molecules that have many degrees of freedom 
[19] . The presence of stabilizing surfactants is generally necessary to assist in forming 
submicrometer particles, and hydrophilic groups in the surfactants lead to rapid 
wetting of the high - surface - area particles in aqueous media, for example, in the case 
of oral administration. It is well known that the unprotective surfaces of nanoparticles 
show a high energy that leads to particle agglomeration. Therefore, the 
nanoparticles must be protected by, for example, steric hindrance and electrostatic 
pulsion. In the case of itraconazole (ITZ) nanosuspension manufacturing, a mixture 
solution of ITZ and Poloxamer (P407) in THF at room temperature was mechanically 
injected into a P407 aqueous solution at 3 ° C. Magnetic stirring was utilized to 
enhance heat and mass transfer. Nanosuspensions containing sub - 300 - nm particles 
were obtained with drug loads as high as 86% [60] . 
Top - down methods are also commonly used to manufacture nanosuspensions, 
including wet milling and homogenization. In pearl/ball milling, the active agent, in 
the presence of surface stabilizer(s), is comminuted by milling media. Particle size 
is determined by stress intensity and the number of contact points. The drug macrosuspensions 
are poured into a milling container containing milling pearls from, 
for example, glass, zircon oxide or special polymers such as hard polystyrene derivatives. 
The drugs are ground to nanocrystals between the pearls. The nanosus pension - 
derived products, Rapamune (sirolimus tablets) and Emend (aprepitant capsules), 
were approved by the FDA and launched in 2000 and 2003, respectively. They are 
manufactured by Elan ’ s NanoCrytal technology using a proprietary wet - milling 
technique. A general problem of pearl mills is potential erosion of materials from 
the milling pearls leading to product contamination. A polymer as substitution may 
minimize erosion. Scaling up with pearl mills is possible; however, there is a certain 
limitation in the size of the mill due to its weight. Up to about two - thirds of the mill 
volume are the pearls lead to heavy weight of the machinery, thus limiting the 
maximum batch size [127] . 
Homogenization can be divided into two types. One is the forcing of a suspension 
under pressure through a narrow - aperture valve (microfl uidization). The other is 
high - pressure homogenization of particles in water or other media (piston gap). 
Microfl uidization is a jet stream principle. The suspension is accelerated and passes 
a specially designed homogenization chamber with a high velocity. In the Z - type 
chamber, the suspension changes the direction of its fl ow a few times, leading to 
particle collision and shear forces, while in the Y - type chamber, the suspension 
stream is divided into two streams colliding frontally. Sometimes it is necessary to 
pass through the microfl uidizer many times to minimize particle size [127] . 
In piston - gap homogenization, suspension contained in a cylinder passes a very 
thin gap with an extremely high velocity. Bubbles of water vapor are produced for 

compensation followed by collapsing in the valve. Cavitation - induced shock waves 
occur and crack the particles. Homogenization can also be utilized to further reduce 
the size of particles made by precipitation. Commonly dendritic crystals made by 
precipitation are more susceptible to rupture by the subsequent mechanical shock 
of homogenization. In addition, the mechanical energy supplied by the homogenizer 
can change the initially formed, unstable amorphous particles to a stable state 
through subsequent crystallization. The size of drug nanocrystals depends mainly 
on (a) power density of the homogenizer, (b) number of homogenization cycles, 
and (c) temperature. Another important determining factor for the fi nal size of drug 
nanocrystals is the hardness of drugs. A relatively soft drug, paclitaxel, can reach 
250 nm in size, which is less than harder drugs. The size should be homogeneous as 
achieved with a homogenizer to avoid physical destabilization. Stabilizers have an 
effect on long - term physical stability but not on maximum dispersity or the nanocrystal 
shape. Contamination from the production equipment is typically below 
1 ppm, which is within a suitable range. Besides water, water - free media and water 
mixtures are used preferably due to advantages of easy evaporation or homogenization 
at higher temperature (with subsequent more cavitation). Oils, propylene 
glycol, and PEG with varieous molecular weights can be used. For PEG being solid 
at room temperature (e.g., PEG 1000, 6000), the obtained drug nanocrystals disperse 
in PEG particles at room temperature and can conveniently be put into hard 
capsules [19, 127] . 
The lyophilized drug nanosuspensions can be transferred to a fi nal dry oral 
dosage form such as tablets or reconstituted prior to administration. Drug nanosuspensions 
can be directly used as parenteral products. A shelf life of up to three years 
was shown for selected nanosuspensions. Sterilization can be achieved by aseptic 
processing of previously sterilized components, membrane fi ltration for parti cles 
suffi ciently small or for drugs that can withstand it, steam sterilization, or 
. - irradiation. 
7.2.3.3 Self - Assembled Drug Nanostructures 
It is well known that liposomes are composed of amphiphilic phospholipids. The 
formation of liposomes is actually a procedure of molecular self - assembly. Furthermore, 
great amounts of amphiphilic compounds, natural or synthesized rather than 
phospholipid - like surfactants, can also self - assemble into ordered aggregates in 
aqueous media or organic solvents [128 – 130] . The formed aggregates are mostly 
nanoarchitectures with various shapes such as vesicle, rod, ribbon, fi ber, tube, or 
helix [131 – 134] . They can remain relatively stable in certain environments. Many 
of them may become drug nanocarriers, such as liposomes, or even perform as active 
agents [109, 135, 136] . The research of self - assembled nanocarriers seems to go into 
the fi eld of supramolecular chemistry. But these results also give us some useful 
information for developing new approaches of drug delivery. A novel idea may 
relate to why we do not try to construct a nanostructure from drugs themselves. 
Twenty years ago the cardiovascular drug pindolol was conjugated with stearyl 
glycerol via succinyl as linker followed by forming maleate salt to obtain pindolol 
diglyceride. Vaizoglu and Speiser used the word “ pharmacosomes ” to describe the 
colloidal dispersions prepared from drug – lipid conjugates with or without additional 
surfactants [137] . Pindolol pharmacosomes (vesicle - like) were prepared from 
NANOTECHNOLOGY FOR DRUG DELIVERY 1275

1276 NANOTECHNOLOGY IN PHARMACEUTICAL MANUFACTURING 
pindolol diglyceride, which showed good stability and useful pharmacokinetic 
parameters. Unfortunately, no more detailed research is being done about pharmacosomes, 
possibly because no appropriate theory supports the new dosage form and 
no proper drugs and lipids are selected. 
The idea of manufacturing nanostructures from drugs may be resourced from 
liposomes, pharmacosomes, and other molecular self - assemblies in supramolecular 
chemistry. More importantly, this novel idea resulted from long - term efforts to 
work on drug delivery and to solve the disadvantages of current drug carriers. 
Almost all current delivery systems (usually called carriers) passively load drugs so 
as to always lead to low entrapment effi ciency and possible drug leakage in preparation, 
preservation, and transportation in vivo [30, 73] , and these carriers might have 
been destroyed in vivo before reaching target sites. In addition, lipophilic biomembranes, 
including cell membranes, usually prevent hydrophilic drugs from entering 
into target sites. If carriers cannot override cell membranes except for endocytosis/ 
phagocytosis by cells/macrophages, the loaded hydrophilic drugs are probably 
released only on target surfaces, not entering. In summary, a majority of drugs could 
not eventually reach and get into target sites due to poor properties of carriers and 
drugs. 
A novel technology involving prodrug, molecular self - assembly, and nanotechnology 
was developed to address the problems of drugs and classical carriers. The 
nanostructures are formed by molecular self - assembly of amphiphilic prodrugs in 
aqueous media generally without additional excipients. The self - assembled drug 
nanostructures not only possess the amphiphilic property of monomolecular drugs, 
benefi ting to cross biomembranes, but also deliver themselves in vivo without “ carriers 
” and then prefer to release active parent agents with a sustained rate. They 
may overcome some defi ciencies of traditional nanocarriers such as liposomes, for 
example, low effi ciency of drug entrapment and loading, rapid drug leakage in 
vitro/in vivo, and bad stability [22] . 
Self - assembled drug nanostructure is not a proprietary term in pharmacy currently. 
Herein this term is defi ned as the ordered nanosized self - aggregates of 
amphiphilic drugs in aqueous media. It is abbreviated as SADN. Another term, 
self - assembled drug delivery system (SADDS), introduced by Jin [22] obviously 
includes SADN. Unfortunately, most current drugs do not occupy an amphiphilic 
and self - assembling nature [138] , so they must be modifi ed in chemical structures 
before manufacturing SADNs. Then the prodrug technique is selected. 
In contrast to nanosuspension technology, SADN technology is mainly applied 
to hydrophilic or polar drugs. These drugs are rationally modifi ed to their amphiphilic 
prodrugs by lipid derivation. Molecular self - assembly in aqueous media is the 
key to manufacturing SADNs. According to the principles of supramolecular chemistry, 
the amphiphilic molecules forming self - assemblies should have proper structural 
conformation. The morphology of assemblies also depends on the structure 
of amphiphiles and the surrounding environment. Some parameters, including the 
optimal head area ao , the volume v of fl uid hydrophobic chain, and the maximum 
effi cient chain length lc , are used to describe the conformation of amphiphiles. The 
critical packing parameter (CPP), equal to v / aolc , can be applied to direct self - 
assembly behavior. The amphiphiles prefer to form vesicles when the CPP is – 1. 
Generally, single - chain lipids with small head group areas (e.g., SDS in a low - salt 
solution) are cone shaped, prone to form spherical micelles, while double - chain 

lipids with large head group area and fl uid chains (e.g., phosphatidylcholine) are 
truncated - cone shaped, prone to form fl exible bilayers, vesicles [139] . Therefore, 
the lipids used for drug covalent conjugation are rationally selected from long - chain 
alkyl lipids, for example, fatty acids, lipid alcohols, lipid amines, long - chain glycolipids, 
and cholesterol. Furthermore, too large or small polar drugs are not appropriate 
for preparation of self - assembling prodrugs. 
Antiviral nucleoside analogues such as acyclovir, didanosine, and zidovudine 
were used to prepare their long - chain glyceride or cholesteryl derivatives in Jin ’ s 
laboratory [140 – 142] . All the derivatives showed amphiphilic property and some of 
them self - assembled into ordered aggregates in water. Amphiphilic prodrugs were 
subsequently used to manufacture self - assemblies using the bottom - up approach, 
such as liposomes, and the self - assembly may be driven by a hydrophobic interaction, 
hydrogen bonding, and so on [21, 143] . The monomolecular amphiphilic 
prodrug is prone to incorporate into the assemblies and not to depart so that almost 
no drugs leak from SADNs. The whole self - assemblies are nearly composed of 
amphiphilic drugs, leading to high drug loading. When SADNs reach targets in vivo, 
the continual dissociation of aggregates and the sustained degradation of prodrugs 
provide controlled drug release. 
Acyclovir self - assembled nanoparticles as SADNs were manufactured which 
showed strong targeting effect in vivo (mainly in the MPSs) and sustained release 
at target sites [22] . Based on this paradigm, a general process to manufacture 
SADNs is as follows [21, 22] : 
(a) To obtain an amphiphilic prodrug with proper molecular structure, 
stearyl glyceride was selected to conjugate with acyclovir. Succinyl acyclovir 
(SACV) was synthesized and subsequently conjugated with stearyl glyceride 
by acylation reaction. The amphiphilic prodrug stearyl - glyceride - succinyl 
acyclovir (SGSA) was obtained. 
(b) The injection method was used to manufacture SADNs. SGSA was 
dissolved in the water - miscible solvent THF. The solution containing 
5 mg/mL SGSA was slowly and continually injected into vortexed water under 
surface via a 100 - . L microsyringe. A homogeneous and slightly opalescent 
suspension was obtained, which was acyclovir self - assembled nanoparticles 
(SANs). 
(c) The organic solvent was removed from the suspension through evaporation 
by heating, and the suspension can further be concentrated by removing 
water under heating until the appropriate prodrug concentration is obtained. 
The concentrated suspension was transferred into ampoules and sealed. It 
may be sterilized by autoclave. 
(d) Acyclovir SANs were characterized. They were cuboidlike shaped based on 
transmission electron microscopy and were nanoscale with an average size 
of 83 nm based on dynamic light scattering. The zeta potential of . 31 mV 
indicated the nanoparticles had negative surface charge. Hydrophobic interaction 
of alkyl chains improves SGSA molecules to form bilayers, and then 
cuboidlike nanoparticles were achieved by layer - by - layer aggregation based 
on inter - bilayer hydrogen bonding. The gel – liquid crystalline phase transition 
was about 50 ° C, and the mechanism of confi guration changes on phase 
transition was analyzed [144] . 
NANOTECHNOLOGY FOR DRUG DELIVERY 1277

1278 NANOTECHNOLOGY IN PHARMACEUTICAL MANUFACTURING 
(e) The in vitro and in vivo behavior of acyclovir SANs was investigated. The 
SANs kept the physical state stable upon centrifugation or exposure of 
some common additives Autoclave and bath heat for sterilization hardly in- 
fl uenced the state of SANs. SGSA in SANs showed good chemical stability 
in weak acidic or neutral buffers, although they were very sensitive to alkaline 
solutions and carboxylester enzymes. The SANs were rapidly removed 
from blood circulation after bolus IV administration to rabbits and mainly 
distributed in liver, spleen, and lung followed by slow elimination in these 
tissues. 
Because nucleoside analogues are important and plentiful agents in antiviral and 
anticancer therapy, other polar drugs can simulate the above process to manufacture 
SADNs. In addition, macrophages as the reservoirs of HIV or other viruses 
prefer to carry viruses throughout the whole body even to the central nervous 
system. How to deliver drugs to macrophages has become a key issue in antiviral 
therapy [145] . SADNs prefer to show macrophage - specifi c distribution. Therefore, 
the antiviral SADNs show the advantages of high drug loading, controlled release, 
and targeting macrophage, which may provide a useful and promising way to treat 
increasing viral diseases. In the future SADNs will be modifi ed to get more functions, 
such as long circulating effect, pH sensitivity, and use in antiviral, anticancer, 
and gene therapy. 
7.2.4 NANOMEDICINE 
Nanomedicine is a concept with broad implications. According to the defi nition of 
the European Science Foundation (ESF), the fi eld of nanomedicine is the science 
and technology of diagnosing, treating, and preventing disease and traumatic injury, 
of relieving pain, and of preserving and improving human health using molecular 
tools and molecular knowledge of the human body. It is perceived as embracing 
fi ve main subdisciplines that in many ways are overlapping and underpinned by the 
following common technical issues: (a) analytical tools, (b) nanoimaging, (c) nanomaterials 
and nanodevices, (d) novel therapeutics and drug delivery systems, and 
(e) clinical, regulatory, and toxicological issues. The ESF ’ s scientifi c forward look 
on nanomedicine warns that nanomedicine benefi ts will be lost without major 
investment and calls for a coordinated European strategy to deliver new nanotechnology 
- based medical tools for diagnostics and therapeutics [146] . From a view of 
narrow sense, nanomedicine can be defi ned as the use of nanoscale or nanostructured 
materials in medicine that have unique medical effects according to their 
structure. In addition, nanostructures up to 1000 nm in size are adopted because 
from a technical point of view the control of materials in this size range not only 
results in new medical effects but also requires novel, scientifi cally demanding 
chemistry and manufacturing techniques [147] . 
The increasing research in nanomedicine has led to many publications, accounting 
for about 4% of publications on nanotechnology research (about 34,300 documents 
in 2004) worldwide. Also commercialization efforts in nanomedicine are 
increasing. About 207 companies (including 158 small - and medium - size enter

prises) visibly pursue nanomedicine activities and devote either all or a signifi cant 
share of their business to the development of nanomedicines. A characterizing 
feature of nanotechnology is its enabling function to add new functionality to existing 
products, making them more competitive. For example, Ambisome (Gilead), 
a liposomal formulation of the fungicide Fungizone (Bristol - Myers Squibb) that 
shows reduced kidney toxicity, had total sales of $ 212 million in 2004. The total 
sales of the 38 identifi ed nanomedicine products from all sectors of nanomedicine 
are estimated to be $ 6.8 billion in 2004. The market is predicted to further grow 
to . $ 12 billion by the year 2012. Currently, nanomedicine is dominated by drug 
delivery systems, accounting for more than 75% of the total sales. Twenty - three 
nanoscale drug delivery systems are available on the market, but within this group, 
three polymer therapeutics alone account for sales of $ 3.2 billion: (i) Neulasta 
(pegfi lagrastim; recombinant methionyl human granulocyte colony stimulating 
factor and PEG), (ii) Pegasys (PEGylated interferon . 2a), and (iii) PEG - Intron 
(PEGylated interferon . 2a), all protein therapeutics to which nanoscale polymer 
strings of PEG have been attached to reduce immunogenicity and to prolong 
plasma half - life. The most widely used nanotechnology product in the fi eld of in 
vitro diagnostics is colloidal gold in lateral fl ow assays, rapid tests for pregnancy, 
ovulation, HIV, and other indications. Magnetic nanoparticles are also used for 
cell - sorting applications in clinical diagnostics. In the fi eld of biomaterials, the commercial 
status of nanotechnology - based dental restoratives is most advanced. Furthermore, 
nanohydroxyapatite - based products for the repair of bone defects have 
been successfully commercialized. Nanotechnology - based contrast agents are a 
market with estimated sales of about $ 12 million. All of the marketed contrast 
agents consist of superparamagnetic iron oxide nanoparticles for magnetic resonance 
imaging. Nanostructured electrodes are used to improve the electrode tissue 
contact, and nanomaterials are used to increase the biocompatibility of implant 
housings. Pacemakers with nanostructured (fractal) electrodes are the only active 
implants currently on the market that contain a nanotechnology - enabled component 
[147] . 
In spite of the great success, the safety of nanomedicine is maintained as a 
worrying issue. A new discipline appears to exploit the toxicological problem in 
nanotechnology applications, called nanotoxicology. Nanotoxicology can be 
defi ned as safety evaluation of engineered nanostructures and nanodevices. 
Nanomaterials could be deposited in all regions of the respiratory tract after 
inhalation. The small size facilitates uptake into cells and transcytosis across 
epithelial and endothelial cells into the blood and lymph circulation to reach 
potentially sensitive target sites such as bone marrow, lymph nodes, spleen, and 
heart. Access to the central nervous system and ganglia via translocation along 
axons and dendrites of neurons has also been observed. Nanomaterials could 
also penetrate the skin via uptake into lymphatic channels [41] . Although possible 
damages of those biodegradable nanomaterials for drug delivery need consideration, 
too much fear is needless. Usually they would be ultimately degraded 
nearly without any trace. However, hard or nonbiodegradable materials, including 
carbon nanotubes, fullerenes, quantum dots, polystyrene, and metal nanoparticles, 
should be thoroughly investigated about their toxic effects on our body 
before clinical application. 
NANOMEDICINE 1279

1280 NANOTECHNOLOGY IN PHARMACEUTICAL MANUFACTURING 
7.2.5 PERSPECTIVE 
Nanotechnology has had a great effect on pharmaceutical manufacturing and 
strongly improves it, rapidly progressing. No one suspects the key role nanotechnology 
will have in future pharmaceutical research and manufacturing. The continually 
increasing achievements in nanotechnology will result in exciting changes in the 
pharmaceutical industry. Now it has gone into an era of controlling the behavior of 
drugs in vitro/in vivo. Although some problems such as toxicity are not addressed, 
the tremendous advantages that result from nanotechnology are obvious. More and 
more potent medicines will be manufactured and diseases such as cancer, HIV, cardiovascular 
diseases, and nervous system diseases may well be cured or better 
treated in the future by nanomedicine technology. 
REFERENCES 
1. Silva , G. A. ( 2006 ), Neuroscience nanotechnology: Progress, opportunities and challenges 
, Nat. Rev. Neurosci ., 7 , 65 – 74 . 
2. Wang , Z. L. ( 2000 ), Characteriaztion of Nanophase Materials , Wiley - VCH , Weinheim, 
Germany , pp. 1 – 6 . 
3. Vasir , J. K. , Reddy , M. K. , and Labhasetwar , V. D. ( 2005 ), Nanosystems in drug targeting: 
Opportunities and challenges , Curr. Nanosci ., 1 , 47 – 64 . 
4. Lane , N. F. ( 2004 ), in Bhushan , B. , Ed., Springer Handbook of Nanotechnology , 
Springer - Verlag , Berlin . 
5. Goodsell , D. S. ( 2004 ), Bionanotechnology: Lessons from Nature , Wiley - Liss , Hoboken, 
NJ , pp. 1 – 6 . 
6. Fritz , J. , Baller , M. K. , Lang , H. P. , Rothuizen , H. , Vettiger , P. , Meyer , E. , Guntherodt , 
H. J. , Gerber , C. , and Gimzewski , J. K. ( 2000 ), Translating biomolecular recognition 
into nanomechanics , Science , 288 , 316 – 318 . 
7. Xu , X. H. N. , Chen , J. , Jeffers , R. B. , and Kyriacou , S. ( 2002 ), Direct measurement of 
sizes and dynamics of single living membrane transporters using nanooptics , Nano Lett ., 
2 , 175 – 182 . 
8. Li , J. , Papadopoulos , C. , and Xu , J. ( 1999 ), Nanoelectronics — Growing Y - junction 
carbon nanotubes , Nature , 402 , 253 – 254 . 
9. Sergeev , G. B. ( 2006 ), Nanochemistry , Elsevier , Amsterdam, The Netherlands . 
10. Moghimi , S. M. , Hunter , A. C. , and Murray , J. C. ( 2005 ), Nanomedicine: Current status 
and future prospects , FASEB J ., 19 , 311 – 330 . 
11. Whitesides , G. M. ( 2003 ), The “ right ” size in nanobiotechnology , Nat. Biotechnol ., 21 , 
1161 – 1165 . 
12. Lee , K. B. , Park , S. J. , Mirkin , C. A. , Smith , J. C. , and Mrksich , M. ( 2002 ), Protein 
nanoarrays generated by dip - pen nanolithography , Science , 295 , 1702 – 1705 . 
13. Liu , G. Y. , and Amro , N. A. ( 2002 ), Positioning protein molecules on surfaces: A nanoengineering 
approach to supramolecular chemistry , Proc. Nat. Acad. Sci. U.S.A. , 99 , 
5165 – 5170 . 
14. Quake , S. R. , and Scherer , A. ( 2000 ), From micro - to nanofabrication with soft 
materials , Science , 290 , 1536 – 1540 . 
15. Xia , Y. N. , McClelland , J. J. , Gupta , R. , Qin , D. , Zhao , X. M. , Sohn , L. L. , Celotta , R. 
J. , and Whitesides , G. M. ( 1997 ), Replica molding using polymeric materials: A practical 
step toward nanomanufacturing , Adv. Mater ., 9 , 147 – 149 . 

16. http://www.whitehouse.gov/omb/budget/fy2007/nsf.html 2007 - 1 - 5 . 
17. Service , R. F. ( 2005 ), Nanotechnology takes aim at cancer , Science , 310 , 1132 – 1134 . 
18. Vauthey , S. , Santoso , S. , Gong , H. , Watson , N. , and Zhang , S. ( 2002 ), Molecular selfassembly 
of surfactant - like peptides to form nanotubes and nanovesicles , Proc. Nat. 
Acad. Sci. U. S. A ., 99 , 5355 – 5360 . 
19. Rabinow , B. E. ( 2004 ), Nanosuspensions in drug delivery , Nat. Rev. Drug Discov ., 3 , 
1 – 12 . 
20. Vinogradov , S. V. ( 2006 ), Colloidal microgels in drug delivery applications , Curr. Pharm. 
Des ., 12 , 4703 – 4712 . 
21. Jin , Y. , Qiao , Y. , Li , M. , Ai , P. , and Hou , X. ( 2005 ), Langmuir monolayers of the long - 
chain alkyl derivatives of a nucleoside analogue and the formation of self - assembled 
nanoparticles , Coll. Surf. B: Biointerf ., 42 , 45 – 51 . 
22. Jin , Y. , Tong , L. , Ai , P. , Li , M. , and Hou , X. ( 2006 ), Self - assembled drug delivery 
systems. 1. Properties and in vitro/in vivo behavior of acyclovir self - assembled nanoparticles 
(SAN) , Int. J. Pharm ., 309 , 199 – 207 . 
23. Boas , U. , and Heegaard , P. M. H. ( 2004 ), Dendrimers in drug research , Chem. Soc. Rev ., 
33 , 43 – 63 . 
24. Kroto , H. W. , Heath , J. R. , O ’ Brien , S.C. , Curl , R. F. , and Smalley , R. E. ( 1985 ), Nature , 
318 , 162 – 163 . 
25. Satoh , M. , and Takayanagi , I. ( 2006 ), Pharmacological studies on fullerene (C60), a 
novel carbon allotrope, and its derivatives , J. Pharm. Sci ., 100 , 513 – 518 . 
26. Michalet , X. , Pinaud , F. F. , Bentolila , L. A. , Tsay , J. M. , Doose , S. , Li , J. J. , Sundaresan , 
G. , Wu , A. M. , Gambhir , S. S. , and Weiss , S. ( 2005 ), Quantum dots for live cells, in vivo 
imaging, and diagnostics , Science , 307 , 538 – 544 . 
27. Muller , R. H. , Mader , K. , and Gohla , S. ( 2000 ), Solid lipid nanoparticles (SLN) for 
controlled drug delivery — A review of the state of the art , Eur. J. Pharm. Biopharm ., 
50 , 161 – 177 . 
28. Groneberg , D. A. , Giersig , M. , Welte , T. , and Pison , U. ( 2006 ), Nanoparticle - based 
diagnosis and therapy , Curr. Drug Targets , 7 , 643 – 648 . 
29. Couvreur , P. , Barratt , G. , Fattal , E. , Legrand , P. , and Vauthier , C. ( 2002 ), Nanocapsule 
technology: A review , Crit. Rev. Ther. Drug Carrier Syst ., 19 , 99 – 134 . 
30. Barenholz , Y. ( 2001 ), Liposome application: Problems and prospects , Curr. Opin. Coll. 
Interf. Sci ., 6 , 66 – 77 . 
31. Torchilin , V. P. ( 2005 ), Recent advances with liposomes as pharmaceutical carriers , Nat. 
Rev. Drug Discov ., 4 , 145 – 160 . 
32. Uchegbu , I. F. , and Vyas , S. P. ( 1998 ), Non - ionic surfactant based vesicles (niosomes) 
in drug delivery , Int. J. Pharm ., 172 , 33 – 70 . 
33. Sadurni , N. , Solans , C. , Azemara , N. , and Garcia - Celma , M. J. ( 2005 ), Studies on the 
formation of O/W nano - emulsions, by low - energy emulsifi cation methods, suitable for 
pharmaceutical applications , Eur. J. Pharm. Sci ., 26 , 438 – 445 . 
34. Solans , C. , Izquierdo , P. , Nolla , J. , Azemara , N. , and Garcia - Celma , M. J. ( 2005 ), Nano - 
emulsions , Curr. Opin. Coll. Interf. Sci ., 10 , 102 – 110 . 
35. Jones , M. - C. , and Leroux , J. - C. ( 1999 ), Polymeric micelles — A new generation of colloidal 
drug carriers , Eur. J. Pharm. Biopharm ., 48 , 101 – 111 . 
36. Torchilin , V. P. ( 2007 ), Micellar nanocarriers: Pharmaceutical perspectives , Pharm. 
Res ., 24 , 1 – 16 . 
37. Bianco , A. , Kostarelos , K. , and Prato , M. ( 2005 ), Applications of carbon nanotubes in 
drug delivery , Curr. Opin. Chem. Biol ., 9 , 674 – 679 . 
REFERENCES 1281

1282 NANOTECHNOLOGY IN PHARMACEUTICAL MANUFACTURING 
38. Foley , S. , Crowley , C. , Smaihi , M. , Bonfi ls , C. , Erlanger , B. F. , Seta , P. , and Larroqueb , 
C. ( 2002 ), Cellular localisation of a water - soluble fullerene derivative , Biochem. Biophys. 
Res. Commun ., 294 , 116 – 119 . 
39. Tsao , N. , Luh , T. - Y. , Chou , C. - K. , Chang , T. - Y. , Wu , J. - J. , Liu , C. - C. , and Lei , 
H. - Y. ( 2002 ), In vitro action of carboxyfullene , J. Antimicrob. Chemother ., 49 , 
641 – 649 . 
40. Ozkan , M. ( 2004 ), Quantum dots and other nanoparticles: What can they offer to drug 
discovery , Drug Discov. Today , 9 , 1065 – 1071 . 
41. Oberdorster , G. , Oberdorster , E. , and Oberdorster , J. ( 2005 ), Nanotoxicology: An 
emerging discipline evolving from studies of ultrafi ne particles , Environ. Health Perspect 
., 113 , 823 – 839 . 
42. des Rieux , A. , Fievez , V. , Garinot , M. , Schneider , Y. - J. , and Preat , V. ( 2006 ), Nanoparticles 
as potential oral delivery systems of proteins and vaccines: A mechanistic approach , 
J. Controlled Release , 116 , 1 – 27 . 
43. Lemos - Senna , E. , Wouessidjewe , D. , Lesieur , S. , and Duchene , D. ( 1998 ), Preparation 
of amphiphilic cyclodextrin nanospheres using the emulsifi cation solvent evaporation 
method. Infl uence of the surfactant on preparation and hydrophobic drug loading , Int. 
J. Pharm ., 170 , 119 – 128 . 
44. Xiao , S. , Tong , C. , Liu , X. , Yu , D. , Liu , Q. , Xue , C. , Tang , D. , and Zhao , L. ( 2006 ), 
Preparation of folate - conjugated starch nanoparticles and its application to tumor - 
targeted drug delivery vector , Chin. Sci. Bull ., 51 , 1693 – 1697 . 
45. Sokolova , V. V. , Radtke , I. , Heumann , R. , and Epple , M. ( 2006 ), Effective transfection 
of cells with multi - shell calcium phosphate - DNA nanoparticles , Biomaterials , 27 , 
3147 – 3153 . 
46. Ueno , Y. , Futagawa , H. , Takagi , Y. , Ueno , A. , and Mizushima , Y. ( 2005 ), Drug - 
incorporating calcium carbonate nanoparticles for a new delivery system , J. Controlled 
Release , 103 , 93 – 98 . 
47. Souza , G. , Christianson , D. R. , Staquicini , F. I. , Ozawa , M. G. , Snyder , E. Y. , Sidman , 
R. L. , Miller , J. R. , Arap , W. , and Pasqualini , R. ( 2006 ), Networks of gold nanoparticles 
and bacteriophage as biological sensors and cell - targeting agents , Proc. Nat. Acad. Sci. 
U.S.A ., 103 , 1215 – 1220 . 
48. Guinebretiere , S. , Briancon , S. , Lieto , J. , Mayer , C. , and Fessi , H. ( 2002 ), Study of the 
emulsion - diffusion of solvent: Preparation and characterization of nanocapsules , Drug 
Dev. Res ., 57 , 18 – 33 . 
49. Hillaireau , H. , Doan , T. L. , Appel , M. , and Couvreur , P. ( 2006 ), Hybrid polymer nanocapsules 
enhance in vitro delivery of azidothymidine - triphosphate to macrophages , 
J. Controlled Release , 116 , 346 – 352 . 
50. New , R. R. C. ( 1990 ), Liposomes: A Practical Approach , Oxford University Press , 
Oxford , pp. 1 – 162 . 
51. Vinogradov , S. , Batrakova , E. , and Kabanov , A. ( 1999 ), Poly(ethylene glycol) - 
polyethylenimine NanoGel particles: Novel drug delivery systems for antisense 
oligonucleotides , Coll. Surf. B: Biointerf ., 16 , 291 – 304 . 
52. Vinogradov , S. V. , Kohli , E. , and Zeman , A. D. ( 2005 ), Cross - linked polymeric nanogel 
formulations of 5 . - triphosphates of nucleoside analogues: Role of the cellular membrane 
in drug release , Mol. Pharm ., 2 , 449 – 461 . 
53. Bosman , A. W. , Janssen , H. M. , and Meijer , E. W. ( 1999 ), About dendrimers: Structure, 
physical properties, and applications , Chem. Rev ., 99 , 1665 – 1688 . 
54. Gagner , J. , Johnson , H. , Watkins , E. , Li , Q. , Terrones , M. , and Majewski , J. ( 2006 ), 
Carbon nanotube supported single phospholipid bilayer , Langmuir , 22 , 10909 – 10911 . 

55. Kam , N. W. S. , Liu , Z. , and Dai , H. ( 2006 ), Carbon nanotubes as intracellular transporters 
for proteins and DNA: An investigation of the uptake mechanism and pathway , 
Ang. Chem. Int. Ed ., 45 , 577 – 581 . 
56. Zakharian , T. Y. , Seryshev , A. , Sitharaman , B. , Gilbert , B. E. , Knight , V. , and Wilson , 
L. J. ( 2005 ), A fullerene - paclitaxel chemotherapeutic: Synthesis, characterization, and 
study of biological activity in tissue culture , J. Am. Chem. Soc ., 127 , 12508 – 12509 . 
57. Akerman, M. E. , Chan, W. C. W. , Laakkonen, P. , Bhatia, S. N. , and Ruoslahti, E. (2002 ), 
Nanocrystal targeting in vivo , Proc. Nat. Acad. Sci. U.S.A ., 99 , 12617 – 12621 . 
58. Boulmedais , F. , Bauchat , P. , Brienne , M. J. , Arnal , I. , Artzner , F. , Gacoin , T. , Dahan , 
M. , and Marchi - Artzner , V. ( 2006 ), Water - soluble pegylated quantum dots: From a 
composite hexagonal phase to isolated micelles , Langmuir , 22 , 9797 – 9803 . 
59. Algar , W. R. , and Krull , U. J. ( 2006 ), Adsorption and hybridization of oligonucleotides 
on mercaptoacetic acid - capped CdSe/ZnS quantum dots and quantum dot - 
oligonucleotide conjugates , Langmuir , 22 , 11346 – 11352 . 
60. Matteucci , M. E. , Hotze , M. A. , Johnston , K. P. , and Williams , III. R. O. ( 2006 ), Drug 
nanoparticles by antisolvent precipitation: Mixing energy versus surfactant stabilization , 
Langmuir , 22 , 8951 – 8959 . 
61. Douroumis , D. , and Fahr , A. ( 2006 ), Nano - and micro - particulate formulations of 
poorly water - soluble drugs by using a novel optimized technique , Eur. J. Pharm. 
Biopharm ., 63 , 173 – 175 . 
62. Torchilin , V. P. ( 2006 ), Multifunctional nanocarriers , Adv. Drug Deliv. Rev ., 58 , 
1532 – 1555 . 
63. Banham , A. D. , Standish , M. M. , and Watkins , J. C. ( 1965 ), Diffusion of univalent ions 
across the lamellae of swollen phospholipids , J. Mol. Biol ., 13 , 238 – 252 . 
64. Lasic , D. D. , Joannic , R. , Keller , B. C. , Frederik , P. M. , and Auvray , L. ( 2001 ), Spontaneous 
vesiculation , Adv. Coll. Interf. Sci ., 89 – 90 , 337 – 349 . 
65. Bhalerao , S. S. , and Harshal , A. R. ( 2003 ), Preparation, optimization, characterization, 
and stability studies of salicylic acid liposomes , Drug Dev. Ind. Pharm ., 29 , 451 – 467 . 
66. Szoka , F. , and Papahadjopoulos , D. ( 1978 ), Procedure for preparation of liposomes with 
large internal aqueous space and high capture by reverse - phase evaporation , Proc. Nat. 
Acad. Sci. U. S. A ., 75 , 4194 – 4198 . 
67. Pons , M. , Foradada , M. , and Estelrich , J. ( 1993 ), Liposomes obtained by the ethanol 
injection method , Int. J. Pharm ., 95 , 51 – 56 . 
68. Kikuchi , H. , Yamauchi , H. , and Hirota , S. A. ( 1994 ), Polyol dilution method for mass 
production of liposomes , J. Liposome Res ., 4 , 71 – 91 . 
69. Wang , T. , Deng , Y. , Geng , Y. , Gao , Z. , Zou , J. , and Wang , Z. ( 2006 ), Preparation of 
submicron unilamellar liposomes by freeze - drying double emulsions , Biochim. Biophys. 
Acta , 1758 , 222 – 231 . 
70. Ning , M. - Y. , Guo , Y. - Z. , Pan , H. - Z. , Yu , H. - M. , and Gu , Z. - W. ( 2005 ), Preparation 
and evaluation of proliposomes containing clotrimazole , Chem. Pharm. Bull ., 53 , 
620 – 624 . 
71. Barnadas - Rodriguez , R. , and Sabes , M. ( 2001 ), Factors involved in the production of 
liposomes with a high - pressure homogenizer , Int. J. Pharm ., 213 , 175 – 186 . 
72. Taira , M. C. , Chiaramoni , N. S. , Pecuch , K. M. , and Alonso - Romanowski , S. ( 2004 ), 
Stability of liposomal formulations in physiological conditions for oral drug delivery , 
Drug Deliv ., 11 , 123 – 128 . 
73. Glavas - Dodov , M. , Fredro - Kumbaradzi , E. , Goracinova , K. , Simonoska , M. , Calis , S. , 
Trajkovic - Jolevska , S. , and Hincal , A. A. ( 2005 ), The effects of lyophilization on the 
stability of liposomes containing 5 - FU , Int. J. Pharm ., 291 , 79 – 86 . 
REFERENCES 1283

1284 NANOTECHNOLOGY IN PHARMACEUTICAL MANUFACTURING 
74. Liang , W. , Levchenko , T. S. , and Torchilin , V. P. ( 2004 ), Encapsulation of ATP into 
liposomes by different methods: Optimization of the procedure , J. Microencapsul ., 21 , 
251 – 261 . 
75. Haran , G. , Cohen , R. , Bar , L. K. , and Barenholz , Y. ( 1993 ), Transmembrane ammonium 
sulfate gradients in liposomes produce effi cient and stable entrapment of amphipathic 
weak bases , Biochim. Biophys. Acta , 1151 , 201 – 215 . 
76. Zhigaltsev , I. V. , Kaplun , A. P. , Kucheryanu , V. G. , Kryzhanovsky , G. N. , Kolomeichuk , 
S. N. , Shvets , V. I. , and Yurasov , V. V. ( 2001 ), Liposomes containing dopamine entrapped 
in response to transmembrane ammonium sulfate gradient as carrier system for dopamine 
delivery into the brain of parkinsonian mice , J. Liposome Res ., 11 , 55 – 71 . 
77. Hwang , S. H. , Maitani , Y. , Qi , X. R. , Takayama , K. , and Nagai , T. ( 1999 ), Remote 
loading of diclofenac, insulin and fl uorescein isothiocyanate labeled insulin into liposomes 
by pH and acetate gradient methods , Int. J. Pharm ., 179 , 85 – 95 . 
78. Abraham , S. A. , Edwards , K. , Karlsson , G. , Hudon , N. , Mayer , L. D. , and Bally , M. B. 
( 2004 ), An evaluation of transmembrane ion gradient - mediated encapsulation of topotecan 
within liposomes , J. Controlled Release , 96 , 449 – 461 . 
79. Gulati , M. , Grover , M. , Singh , S. , and Singh , M. ( 1998 ), Lipophilic drug derivatives in 
liposomes , Int. J. Pharm ., 165 , 129 – 168 . 
80. Wagner , A. , Vorauer - Uhl , K. , and Katinger , H. ( 2002 ), Liposomes produced in a pilot 
scale: Production, purifi cation and effi ciency aspects , Eur. J. Pharm. and Biopharm ., 54 , 
213 – 219 . 
81. Wagner , A. , Vorauer - Uhl , K. , Kreismayr , G. , and Katinger , H. ( 2002 ), The crossfl ow 
injection technique — An improvement of the ethanol injection method , J. Liposome 
Res ., 12 , 259 – 270 . 
82. Wagner , A. , Platzgummer , M. , Kreismayr , G. , Quendler , H. , Stiegler , G. , Ferko , B. , 
Vecera , G. , Vorauer - Uhl , K. , and Katinger , H. ( 2006 ), GMP production of liposomes: 
A new industrial approach , J. Liposome Res ., 16 , 311 – 319 . 
83. Woodle , M. C. , and Scaria , P. ( 2001 ), Cationic liposomes and nucleic acids , Curr. Opin. 
Coll. Interf. Sci ., 6 , 78 – 84 . 
84. Templeton , N. S. ( 2002 ), Cationic liposome - mediated gene delivery in vivo , Biosci. Rep ., 
22 , 283 – 295 . 
85. Ishida , T. , Harashima , H. , and Kiwada , H. ( 2002 ), Liposome clearance , Biosci. Rep ., 22 , 
197 – 224 . 
86. Winterhalter , M. , Frederik , P. M. , Vallner , J. J. , and Lasic , D. D. ( 1997 ), Stealth ® liposomes: 
From theory to product , Adv. Drug Deliv. Rev ., 24 , 165 – 177 . 
87. Ishida , T. , Ichihara , M. , Wang , X. , and Kiwada , H. ( 2006 ), Spleen plays an important 
role in the induction of accelerated blood clearance of PEGylated liposomes , J. Controlled 
Release , 115 , 243 – 250 . 
88. Moghimi , S. M. , Hamad , I. , Andresen , T. L. , Jorgensen , K. , and Szebeni , J. ( 2006 ), 
Methylation of the phosphate oxygen moiety of phospholipid - methoxy(polyethylene 
glycol) conjugate prevents PEGylated liposome - mediated complement activation and 
anaphylatoxin production , FASEB J ., 20 , 2591 – 2593 . 
89. Drummond , D. C. , Zignani , M. , and Leroux , J. - C. ( 2000 ), Current status of pH - sensitive 
liposomes in drug delivery , Prog. Lipid Res ., 39 , 409 – 460 . 
90. Yatvin , M. B. , Weinstein , J. N. , Dennis , W. H. , and Blumenthal , R. ( 1978 ), Liposomes 
and local hyperthermia: Selective delivery of methotrexate to heated tumors , Science , 
202 , 1290 – 1293 . 
91. Kono , K. ( 2001 ), Thermosensitive polymer - modifi ed liposomes , Adv. Drug Deliv. Rev ., 
53 , 307 – 319 . 

92. Zhang , J. Q. , Zhang , Z. R. , Yang , H. , Tan , Q. Y. , Qin , S. R. , and Qiu , X. L. ( 2005 ), 
Lyophilized paclitaxel magnetoliposomes as a potential drug delivery system for breast 
carcinoma via parenteral administration: In vitro and in vivo studies , Pharm. Res ., 22 , 
573 – 583 . 
93. Koning , G. A. , Kamps , J. A. A. M. , and Scherphof , G. L. S. (2002), Effi cient intracellular 
delivery of 5 - fl uorodeoxyuridine into colon cancer cells by targeted immunoliposomes , 
Cancer Detection Prevention , 26 , 299 – 307 . 
94. Mehnert , W. , and Mader , K. ( 2001 ), Solid lipid nanoparticles: Production, characterization 
and applications , Adv. Drug Deliv. Rev ., 47 , 165 – 196 . 
95. Shive , M. S. , and Anderson , J. M. ( 1997 ), Biodegradation and biocompatibility of PLA 
and PLGA microspheres , Adv. Drug Deliv. Rev ., 28 , 5 – 24 . 
96. Sommerfeld , P. , Schroeder , U. , and Sabel , B. A. ( 1997 ), Long - term stability of PBCA 
nanoparticle suspensions suggests clinical usefulness , Int. J. Pharm ., 155 , 201 – 207 . 
97. Sommerfeld , P. , Schroeder , U. , and Sabel , B. A. ( 1998 ), Sterilization of unloaded polybutylcyanoacrylate 
nanoparticles , Int. J. Pharm ., 164 , 113 – 118 . 
98. Chorny , M. , Fishbein , I. , Danenberg , H. D. , and Golomb , G. ( 2002 ), Lipophilic drug 
loaded nanospheres prepared by nanoprecipitation: Effect of formulation variables on 
size, drug recovery and release kinetics , J. Controlled Release , 83 , 389 – 400 . 
99. Govender , T. , Stolnik , S. , Garnett , M. C. , Illum , L. , and Davis , S. S. ( 1999 ), PLGA 
nanoparticles prepared by nanoprecipitation: Drug loading and release studies of a 
water soluble drug , J. Controlled Release , 57 , 171 – 185 . 
100. Bilati , U. , Allemann , E. , and Doelker , E. ( 2005 ), Development of a nanoprecipitation 
method intended for the entrapment of hydrophilic drugs into nanoparticles , Eur. J. 
Pharm. Sci ., 24 , 67 – 75 . 
101. Bilati , U. , Allemann , E. , and Doelker , E. ( 2005 ), Nanoprecipitation versus emulsion - 
based techniques for the encapsulation of proteins into biodegradable nanoparticles and 
process - related stability issues , AAPS Pharm. Sci. Tech ., 6 , 74 . 
102. Astete , C. E. , and Sabliov , C. M. ( 2006 ), Synthesis and characterization of PLGA 
nanoparticles , J. Biomater. Sci. Polym. Ed ., 17 , 247 – 289 . 
103. Zwiorek , K. , Kloechner , J. , Wagner , E. , and Coester , C. ( 2004 ), Gelatin nanoparticle as 
a new and simple gene delivery system , J. Pharm. Pharm. Sci ., 7 , 22 – 28 . 
104. Cheng , J. , Teply , B. A. , Sherifi , I. , Sung , J. , Luther , G. , Gu , F. X. , Levy - Nissenbaum , 
E. , Radovic - Moreno , A. F. , Langer , R. , and Farokhzad , O. C. ( 2007 ), Formulation of 
functionalized PLGA - PEG nanoparticles for in vivo targeted drug delivery , Biomaterials 
, 28 , 869 – 876 . 
105. Hillery , A. M. , and Florence , A. T. ( 1996 ), The effect of adsorbed poloxamer 188 and 
407 surfactants on the intestinal uptake of 60 - nm polystyrene particles after oral administration 
in the rat , Int. J. Pharm ., 132 , 123 – 130 . 
106. Peracchia , M. T. , Vauthier , C. , Puisieux , F. , and Couvreur , P. ( 1997 ), Development of 
sterically stabilized poly(isobutyl 2 - cyanoacrylate) nanoparticles by chemical coupling 
of poly(ethylene glycol) , J. Biomed. Mater. Res ., 34 , 317 – 326 . 
107. Suk , J. S. , Suh , J. , Choy , K. , Lai , S. K. , Fu , J. , and Hanes , J. ( 2006 ), Gene delivery to 
differentiated neurotypic cells with RGD and HIV Tat peptide functionalized polymeric 
nanoparticles , Biomaterials , 27 , 5143 – 5150 . 
108. Aktas , Y. , Yemisci , M. , Andrieux , K. , Gursoy , R. N. , Alonso , M. J. , FernAndez - Megia , 
E. , Novoa - Carballal , R. , Quinoa , E. , Riguera , R. , Sargon , M. F. , Celik , H. H. , Demir , 
A. S. , Hincal , A. A. , Dalkara , T. , Capan , Y. , and Couvreur , P. ( 2005 ), Development and 
brain delivery of chitosan - PEG nanoparticles: Functionalized with the monoclonal antibody 
OX26 , Bioconjugate Chem ., 16 , 1503 – 1511 . 
REFERENCES 1285

1286 NANOTECHNOLOGY IN PHARMACEUTICAL MANUFACTURING 
109. Park , J. H. , Kwon , S. , Nam , J. - O. , Park , R. - W. , Chung , H. , Seo , S. B. , Kim , I. - S. , Kwon , 
I. C. , and Jeong , S. Y. ( 2004 ), Self - assembled nanoparticles based on glycol chitosan 
bearing 5h - cholanic acid for RGD peptide delivery , J. Controlled Release , 95 , 579 – 
588 . 
110. Klier , J. , Tucker , C. J. , Kalantar , T. H. , and Green , D. P. ( 2000 ), Properties and applications 
of microemulsions , Adv. Mater ., 12 , 1751 – 1757 . 
111. Muller , R. H. , Radtke , M. , and Wissing , S. A. ( 2002 ), Solid lipid nanoparticles (SLN) 
and nanostructured lipid carriers (NLC) in cosmetic and dermatological preparations , 
Adv. Drug Deliv. Rev ., 54 , S131 - S155 . 
112. Goppert , T. M. , and Muller , R. H. ( 2003 ), Plasma protein adsorption of Tween 80 - 
and Poloxamer 188 - stabilized solid lipid nanoparticles , J. Drug Target ., 11 , 225 – 
231 . 
113. Wong , H. L. , Bendayan , R. , Rauth , A. M. , and Wu , X. Y. ( 2006 ), Simultaneous delivery 
of doxorubicin and GG918 (Elacridar) by new polymer - lipid hybrid nanoparticles 
(PLN) for enhanced treatment of multidrug - resistant breast cancer , J. Controlled 
Release , 116 , 275 – 284 . 
114. Hu , F. Q. , Hong , Y. , and Yuan , H. ( 2004 ), Preparation and characterization of solid 
lipid nanoparticles containing peptide , Int. J. Pharm ., 273 , 29 – 35 . 
115. Garcia - Fuentes , M. , Torres , D. , and Alonso , M. J. ( 2002 ), Design of lipid nanoparticles 
for the oral delivery of hydrophilic macromolecules , Coll. Surf. B Biointerf ., 27 , 
159 – 168 . 
116. Brannon - Peppas , L. , and Blanchette , J. O. ( 2004 ), Nanoparticle and targeted systems 
for cancer therapy , Adv. Drug Deliv. Rev ., 56 , 1649 – 1659 . 
117. Lockman , P. R. , Mumper , R. J. , Khan , M. A. , and Allen , D. D. ( 2002 ), Nanoparticle 
technology for drug delivery across the blood - brain barrier , Drug Dev. Ind. Pharm ., 28 , 
1 – 13 . 
118. Nishioka , Y. , and Yoshino , H. ( 2001 ), Lymphatic targeting with nanoparticulate system , 
Adv. Drug Deliv. Rev ., 47 , 55 – 64 . 
119. Csaba , N. , Caamano , P. , Sanchez , A. , Dominguez , F. , and Alonso , M. J. ( 2005 ), PLGA: 
Poloxamer and PLGA:Poloxamine blend nanoparticles: New carriers for gene delivery , 
Biomacromolecules , 6 , 271 – 278 . 
120. Bagwe , R. P. , Kanicky , J. R. , Palla , B. J. , Patanjali , P. K. , and Shah , D. O. ( 2001 ), 
Improved drug delivery using microemulsions: Rationale, recent progress, and new 
horizons , Crit. Rev. Ther. Drug Carrier Syst ., 18 , 77 – 140 . 
121. Nayak , S. , and Lyon , L. A. ( 2005 ), Soft nanotechnology with soft nanoparticles , Ang. 
Chem. Int. Ed ., 44 , 7686 – 7708 . 
122. Tomalia , D. A. , Baker , H. , Dewald , J. R. , Hall , M. , Kallos , G. , Martin , S. , Roeck , J. , 
Ryder , J. , and Smith , P. ( 1985 ), A new class of polymers: Starburst - dendritic macromolecules 
, Polym. J ., 17 , 117 – 132 . 
123. Tomalia , D. A. , Baker , H. , Dewald , J. R. , Hall , M. , Kallos , G. , Martin , S. , Roeck , J. , 
Ryder , J. , and Smith , P. ( 1986 ), Dendritic macromolecules: Synthesis of starburst dendrimers 
, Macromolecules , 19 , 2466 – 2468 . 
124. Svenson , S. , and Tomalia , D. A. ( 2005 ), Dendrimers in biomedical applications — 
Refl ections on the fi eld , Adv. Drug Deliv. Rev ., 57 , 2106 – 2129 . 
125. Lacerda , L. , Bianco , A. , Prato , M. , and Kostarelos , K. ( 2006 ), Carbon nanotubes as 
nanomedicines: From toxicology to pharmacology , Adv. Drug Deliv. Rev ., 58 , 1460 – 
1470 . 

126. Smart , S. K. , Cassady , A. I. , Lu , G. Q. , and Martin , D. J. ( 2006 ), The biocompatibility 
of carbon nanotubes , Carbon , 44 , 1034 – 1047 . 
127. Keck , C. M. , and Muller , R. H. ( 2006 ), Drug nanocrystals of poorly soluble drugs produced 
by high pressure homogenisation , Eur. J. Pharm. Biopharm ., 62 , 3 – 16 . 
128. Guerin , C. B. E. , and Szleifer , I. ( 1999 ), Self - assembly of model nonionic amphiphilic 
molecules , Langmuir , 15 , 7901 – 7911 . 
129. Choi , S. K. , Vu , T. K. , Jung , J. M. , Kim , S. J. , Jung , H. R. , Chang , T. , and Kim , B. H. 
( 2005 ), Nucleoside - based phospholipids and their liposomes formed in water , ChemBio- 
Chem , 6 , 432 – 439 . 
130. Snip , E. , Shinkai , S. , and Reinhoudt , D. N. ( 2001 ), Organogels of a nucleobase - bearing 
gelator and the remarkable effects of nucleoside derivatives and a porphyrin derivative 
on the gel stability , Tetrahedr. Lett ., 42 , 2153 – 2156 . 
131. Engberts , J. B. F. N. , and Hoekstra , D. ( 1995 ), Vesicle - forming synthetic amphiphiles , 
Biochim. Biophys. Acta , 1241 , 323 – 340 . 
132. Giulieri , F. , and Krafft , M. P. ( 2003 ), Tubular microstructures made from nonchiral 
single - chain fl uorinated amphiphiles: Impact of the structure of the hydrophobic chain 
on the rolling - up of bilayer membrane , J. Coll. Interf. Sci ., 258 , 335 – 344 . 
133. Giorgi , T. , Lena , S. , Mariani , P. , Cremonini , M. A. , Masiero , S. , Pieraccini , S. , Rabe , 
J. P. , Samori , P. , Spada , G. P. , and Gottarelli , G. ( 2003 ), Supramolecular helices via 
self - assembly of 8 - oxoguanosines , J. Am. Chem. Soc ., 125 , 14741 – 14749 . 
134. Yanagawa , H. , Ogawa , Y. , Furuta , H. , and Tsuno , K. ( 1989 ), Spontaneous formation of 
superhelical strands , J. Am. Chem. Soc ., 111 , 4567 – 4570 . 
135. Zemel , A. , Fattal , D. R. , and Ben - Shaul , A. ( 2003 ), Energetics and self - assembly of 
amphipathic peptide pores in lipid membranes , Biophys. J ., 84 , 2242 – 2255 . 
136. Chabaud , P. , Camplo , M. , Payet , D. , Serin , G. , Moreau , L. , Barthelemy , P. , and 
Grinstaff , M. W. ( 2006 ), Cationic nucleoside lipids for gene delivery , Bioconjugate 
Chem ., 17 , 466 – 472 . 
137. Vaizoglu , M. O. , and Speiser , P. P. ( 1986 ), Pharmacosomes — A novel drug delivery 
system , Acta Pharm. Suec ., 23 , 163 – 172 . 
138. Schreier , S. , Malheiros S. V. P. , and de Paula , E. ( 2000 ), Surface active drugs: Self - 
association and interaction with membranes and surfactants. Physicochemical and biological 
aspects , Biochim. Biophys. Acta , 1508 , 210 – 234 . 
139. Israelachvili , J. N. ( 1992 ), Intermolecular and Surface Forces with Applications to Colloidal 
and Biological Systems , Academic , London . 
140. Jin , Y. , Li , M. , Tong , L. , Wang , L. , and Peng , T. ( 2003 ), Lipid derivatives of nucleoside 
analogues and their salts, China Patent CN1259331. 
141. Jin , Y. , and Ai , P. ( 2004 ), Cholesteryl derivatives of nucleoside analogues, China Patent 
CN1566130. 
142. Jin , Y. , Du , L. , Xing , L. , and Xin , R. ( 2006 ), Cholesteryl phosphoryl derivatives of 
nucleoside analogues, China Patent Application 2006101122957. 
143. Jin , Y. , Qiao , Y. , and Hou , X. ( 2006 ), The effects of chain number and state of lipid 
derivatives of nucleosides on hydrogen bonding and self - assembly through the investigation 
of Langmuir - Blodgett fi lms , Appl. Surf. Sci ., 252 , 7926 – 7929 . 
144. Jin , Y. ( 2007 ), Effect of temperature on the state of the self - assembled nanoparticles 
prepared from an amphiphilic lipid derivative of acyclovir , Coll. Surf. B Biointerf ., 54 , 
124 – 125 . 
REFERENCES 1287

1288 NANOTECHNOLOGY IN PHARMACEUTICAL MANUFACTURING 
145. Aquaro , S. , Calio , R. , Balzarini , J. , Bellocchi , M. C. , Garaci , E. , and Perno , C. F. ( 2002 ), 
Macrophages and HIV infection: Therapeutical approaches toward this strategic virus 
reservoir , Antivir. Res ., 55 , 209 – 225 . 
146. European Science Foundation ( 2005 ), Nanomedicine, An ESF - European Medical 
Research Council (EMRC) forward look report, European Science Foundation, 
Strasbourg. 
147. Wagner , V. , Dullaart , A. , Bock , A. - K. , and Zweck , A. ( 2006 ), The emerging nanomedicine 
landscape , Nat. Biotechnol ., 24 , 1211 – 1217 . 

1289 
7.3 
PHARMACEUTICAL 
NANOSYSTEMS: MANUFACTURE, 
CHARACTERIZATION, AND 
SAFETY 
D. F. Chowdhury 
University of Oxford, Oxford, United Kingdom 
Contents 
7.3.1 Defi nition 
7.3.1.1 Top - Down and Bottom - Up Approaches to Nanotechnology 
7.3.2 Taxonomy of Nanomedicine Technologies 
7.3.3 Nano – Pharmaceutical Systems 
7.3.4 Description of Nanosystems 
7.3.4.1 Polymeric Systems 
7.3.4.2 Quantum Dots and Quantum Confi nement 
7.3.4.3 Metal Nanoparticles and Surface Plasmon Resonance 
7.3.4.4 Self - Assembled Systems 
7.3.4.5 Nanostructures Based on Carbon 
7.3.5 Manufacturing Technologies 
7.3.5.1 Nanoscale Assembly Methods 
7.3.5.2 Nano - structuring processes for polymeric materials 
7.3.6 Characterization Techniques 
7.3.6.1 Nanoparticle Characterization Methods and Tools 
7.3.6.2 Scanning Probe Technologies 
7.3.7 Toxicology Considerations 
7.3.7.1 Lung Toxicity 
7.3.7.2 Systemic Uptake 
7.3.7.3 Skin Permeation of Nanoparticles 
References 
Suggested Reading 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

1290 PHARMACEUTICAL NANOSYSTEMS 
7.3.1 DEFINITION 
Nanotechnology is an enabling technology and one which is generally manifest at 
the primary level in the form of nanomaterials. The defi nition of nanotechnology 
therefore focuses on materials and how manipulation at the nanoscale leads to novel 
properties and therefore potentially new uses. The pharmaceutical industry has yet 
to adopt strict guidelines for what falls under the remit of nanotechnology, with 
numerous defi nitions in existence. For the purpose of this chapter, the current U.S. 
Food and Drug Administration (FDA) defi nition for nanotechnology as applied to 
pharmaceuticals is deemed most appropriate. The FDA describes nanotechnology 
as technology that includes the following [1] : 
1. Research and technology development or products regulated by the FDA that 
are at the atomic, molecular or macromolecular levels and where at least one 
dimension that affects the functional behavior of the product is in the length 
scale range of approximately 1 – 100 nm 
2. Creating and using structures, devices, and systems that have novel properties 
and functions because of their small and/or intermediate size 
3. Ability to control or manipulate at the atomic scale 
Nanotechnology is therefore essentially about understanding and manipulating 
materials at the atomic, molecular, and macromolecular level in a way that imparts 
properties to the material that would otherwise not exist either as individual atoms 
or as bulk processed macroscopic systems. 
Properties that can be exploited to provide novel and unique properties to materials 
include surface and quantum effects, for example, van der Waals forces; electrostatic 
interaction; ionic, covalent and hydrogen bonding; and quantum confi nement. 
Additionally nonconventional means of molecular assembly and atomic manipulation 
can lead to novel material properties. Control and exploitation of these effects 
can lead to new and useful changes to the thermal, magnetic, electrical, optical and 
mechanical, and biological and physicochemical properties of materials. 
7.3.1.1 Top - Down and Bottom - Up Approaches to Nanotechnology 
There are generally two approaches to nanotechnology, the top - down and bottom - 
up approaches. As the names suggest, the top - down approach utilizes ultraprecision 
machining and nanolithographic techniques among others to achieve very high 
defi nition structures with nanolevel accuracy, usually either by removing material 
from the surface of a larger structure until the desired structure with desired features 
is achieved or through deposition of material with almost atomic - scale precision 
and control. The bottom - up approach involves the assembly of atoms, molecules, 
or nanoscale components to assemble a larger structure within the nanoscale range. 
There are numerous methods by which this can be achieved, including conventional 
bulk chemical processing methods and exploitation of chemical and biological self - 
assembly techniques. The pharmaceutical industry is primarily involved in the application 
of nanomaterials rather than the discovery and development of new materials, 
though as this chapter will indicate, there are often areas of overlap between what 
is a new material and a construct of a novel material. 

7.3.2 TAXONOMY OF NANOMEDICINE TECHNOLOGIES 
A useful starting point would be to gauge the breadth of technologies falling under 
the classifi cation of nanomedicine. Table 1 provides a means of classifying materials 
and processes derived from nanotechnology as relating to pharmaceuticals and 
medicine in general. 
7.3.3 NANO – PHARMACEUTICAL SYSTEMS 
Having gauged the huge scope of nanotechnology in medicine, the scope of this 
chapter is limited to pharmaceuticals. The term pharmaceutical is intended in the 
context of systems pertaining to drugs or dosage formulations. Nano – pharmaceutical 
systems generally imply products that may in their own right or in combination 
with another moiety bring about therapeutic benefi t. They may also include engineered 
nanostructured systems that may act as a carrier for drugs or a delivery 
vehicle or a delivery system for drugs and therapeutic agents. The defi nition is 
extended to include imaging systems which may be used alone or in conjunction 
with therapeutic agents given the numerous nanosystems that have found application 
in diagnostics and imaging. 
The various nanoscale architectures that can be achieved using nanotechnology 
include spheres (solid or hollow), tubes, porous particles, solid particles, and branched 
structures, and with the rapid evolution of lithographic techniques, three - 
dimensional objects of almost any desired shape can be achieved from both metals 
and polymers. Given the vast spectrum of materials of construction, size, shape, and 
form covered by nanosystems, a simple means of classifi cation is needed for effective 
differentiation between systems. Nanosystems can be classifi ed in a number of ways, 
for example, according to their elemental composition, according to size, structure, 
and function, or perhaps according to a structure – function relationship. It can be 
said that nanosystems fall into the broader category of nanostructures, which can 
be generalized into the following categories: 
Particulate nanostructures 
Capsular nanostructures 
Crystalline nanostructures 
Polymeric nanostructures 
It can be readily appreciated that there will be an element of overlap between 
the broad categories above and in particular with evolving complex hybrid systems. 
The classifi cation is intended as a point of reference and for ease of understanding 
the vast possibilities that exist with nanosystems without the need for constant 
reclassifi cation as far as possible. These structures may be further differentiated 
according to their primary composition: 
Organic 
Inorganic 
Organic/inorganic hybrid 
Carbon based 
NANO-PHARMACEUTICAL SYSTEMS 1291

1292 PHARMACEUTICAL NANOSYSTEMS 
TABLE 1 Partial Nanomedicine Technologies Taxonomy 
Raw nanomaterials 
Nanoparticle coatings 
Nanocrystalline materials 
Nanostructured materials 
Cyclic peptides 
Dendrimers 
Detoxifi cation agents 
Fullerenes 
Functional drug carriers 
Magnetic resonance (MR) scanning 
(nanoparticles) 
Nanobarcodes 
Nanoemulsions 
Nanofi bers 
Nanoparticles 
Nanoshells 
Carbon nanotubes 
Quantum dots 
Artifi cial binding sites 
Artifi cial antibodies 
Artifi cial enzymes 
Artifi cial receptors 
Molecularly imprinted polymers 
Control of surfaces 
Artifi cial surfaces, adhesive 
Artifi cial surfaces, nonadhesive 
Artifi cial surfaces, regulated 
Biocompatible surfaces 
Biofi lm suppression 
Engineered surfaces 
Pattern surfaces (contact guidance) 
Thin - fi lm coatings 
Nanopores 
Immunoisolation 
Molecular sieves and channels 
Nanofi ltration membranes 
Nanopores 
Separations 
Biological research 
Nanobiology 
Nanoscience in life sciences 
Drug delivery 
Drug discovery 
Biopharmaceuticals 
Drug delivery 
Drug encapsulation 
Smart drugs 
Synthetic biology and early nanodevices 
Dynamic nanoplatform “ nanosome ” 
Tecto - dendrimers 
Artifi cial cells and liposomes 
Polymeric micelles and polymersomes 
Nanorobotics 
DNA - based devices and nanorobots 
Diamond - based nanorobots 
Cell repair devices 
Cell simulations and cell diagnostics 
Cell chips 
Cell stimulators 
DNA manipulation, sequencing, 
diagnostics 
Genetic testing 
Deoxyribonucleic acid (DNA) 
microarrays 
Ultrafast DNA sequencing 
DNA manipulation and control 
Tools and diagnostics 
Bacterial detection systems 
Biochips 
Biomolecular imaging 
Biosensors and biodetection 
Diagnostic and defense applications 
Endoscopic robots and microscopes 
Fullerene - based sensors 
Imaging (e.g., cellular) 
Lab on chip 
Monitoring 
Nanosensors 
Point - of - care diagnostics 
Protein microarrays 
Scanning probe microscopy 
Intracellular devices 
Intracellular assay 
Intracellular biocomputers 
Intracellular sensors/reporters 
Implant inside cells 
BioMEMS 
Implantable materials and devices 
Implanted bio — microelectromechanical 
systems (MEMSs), chips, and electrodes 
MEMS/nanomaterial - based prosthetics 
Sensory aids (e.g.,) 
Microarrays 
Microcantilever - based sensors 
Microfl uidics 
Microneedles 
Medical MEMS 
MEMS surgical devices 

Molecular medicine 
Genetic therapy 
Pharmacogenomics 
Artifi cial enzymes and enzyme control 
Enzyme manipulation and control 
Nanotherapeutics 
Antibacterial and antiviral nanoparticles 
Fullerene - based pharmaceuticals 
Photodynamic therapy 
Radiopharmaceuticals 
Biotechnology and biorobotics 
Biological viral therapy 
Virus - based hybrids 
Stem cells and cloning 
Tissue engineering 
Artifi cial organs 
Nanobiotechnology 
Biorobotics and biobots 
Source : From ref. 2 . 
TABLE 1 Continued 
7.3.4 DESCRIPTION OF NANOSYSTEMS 
There is clear evidence from Table 2 that some of the nanosystems indicated are 
based on conventional colloidal chemistry, and their characteristics are well established 
and understood. The descriptions below deal mainly with systems deemed 
nonconventional and cover some of the key novel properties derived from or utilized 
as part of their construction. 
7.3.4.1 Polymeric Systems 
Polymer - based systems offer numerous advantages, such as biocompatibility, biodegradability, 
and ability to incorporate functional groups for attachment of drugs. 
Drugs can be incorporated into the polymer matrix or in the cavity created by the 
polymeric architecture, from which the drug molecule can be released with an 
element of temporal control, and controlled pharmacokinetic profi le with almost 
zero - order release achievable. 
Dendrimers are large complex globular polymeric molecules [46] with well - 
defi ned chemical structure, size, and shape [47] . They consist of characteristic three - 
dimensional branched structures. The key components of dendrimers are their core, 
branches, and end groups, and precise control over these features is possible during 
the bottom - up synthesis process, thus allowing control over size composition, and 
fi nal chemical reactivity. A more advanced form of dendrimer is the hyperbranched 
dendrimer, where precision control over the architectural construct is lost during 
the synthesis process [48] . Dendrimers are produced from monomers through an 
iterative sequence of reaction steps [49] using either convergent [50, 51] or divergent 
[52, 53] step growth polymerization [54 – 56] and have potential applications in gene 
and cancer therapy and drug delivery through complexation or encapsulation 
[57 – 59] . 
7.3.4.2 Quantum Dots and Quantum Confi nement 
Quantum dots are inorganic semiconductor nanocrystals that possess physical 
dimensions smaller than the exciton Bohr radius, giving rise to the unique phenom- 
DESCRIPTION OF NANOSYSTEMS 1293

1294 PHARMACEUTICAL NANOSYSTEMS 
enon known as quantum confi nement. Quantum confi nement is the spatial confi nement 
of charge carriers (i.e., electrons and holes) within materials. It leads to unique 
optical and electrical properties that are not common for bulk solids. 
Quantum dots have novel and unique optical, magnetic [60] , and electronic properties, 
exceptional imaging properties due to high color intensity, with up to 20 fl uorophores, 
high resistance to photobleaching, and narrow spectral line widths. Their 
size and composition allow for tunable emission that can be excited using a single 
wavelength [61 – 64] . These properties have led to uses as fl uorescent imaging probes, 
detection of cell signaling pathways, and cell targeting. Low depth of light penetration 
and relatively high background fl uorescence are the key limitations of quantum 
dots in in vivo clinical applications. Quantum dots are generally of the size rage 2 – 
8 nm in diameter [65] and have large molar extinction coeffi cients [66] , thus making 
them very bright in vivo probes. 
TABLE 2 Classifi cation of Nanostructures According to Composition and Perceived 
Applications 
Composition Type of Nanostructure Applications 
Organic Polymer micelles [3, 4] Drug delivery 
Polymeric spheres [5] Drug delivery 
Polymer nanoparticles [6, 7] Drug delivery 
Polymer vesicles/containers [8 – 10] Drug delivery 
Lipid nanovesicles [11] Drug delivery 
Lipid emulsions [12] Drug delivery 
Ring peptides [13] Drug delivery 
Lipid nanospheres [14, 15] Drug delivery 
Lipid nanoparticles [16, 17] Drug delivery 
Lipid nanotubes [18] Drug delivery 
Peptide nanoparticles [19, 20] Drug delivery 
Nanobodies [21] Therapeutic, diagnostic 
Dendrimers [22 – 24] Drug delivery, gene delivery 
Inorganic Palladium/platinum nanoparticles [25] Drug delivery 
Silicon nanoneedles [26] Drug delivery 
Porous silicon [27, 28] Drug delivery 
Gold nanoparticles [29] Drug delivery 
Iron oxide nanoparticles [30] Imaging 
Gold nanoshells [31] Imaging agent, thermal ablation 
Quantum dots [32] Imaging, cell targeting 
Metallic nanoshells [33] Imaging, thermal ablation 
Nanocrystals [34, 35] Drug delivery 
Organic/ 
inorganic 
hybrid 
Nanocomposites [36] Drug delivery 
Nanosphere – metallic particle 
composite [37] 
Drug delivery, imaging 
Carbon nanotube clusters [38] Drug delivery, imaging, thermal 
ablation 
Core – shell structures [39, 40] Imaging, thermal ablation 
Carbon 
based 
Fullerenes [41 – 43] Drug delivery, prodrug 
Carbon nanotubes [44, 45] Drug delivery, imaging, thermal 
ablation

Applications of quantum dots include optical detection of genes and proteins in 
animal models and cell assays and tumor and lymph node visualization through 
imaging [67, 68] . 
7.3.4.3 Metal Nanoparticles and Surface Plasmon Resonance 
Surface plasmons, also known as surface plasmon polaritons or packets of electrons, 
are surface electromagnetic waves that propagate parallel along a metal – dielectric 
interface [69, 70] . Surface plasmons exist where the complex dielectric constants of 
the two media are of opposite sign. The excitation of surface plasmons by light of 
a wavelength matching the resonant frequency of the electrons is termed surface 
plasmon resonance (SPR) for planar surfaces and localized surface plasmon resonance 
where nanometer - sized metallic structures are concerned [71, 72] . 
Surface plasmon effects result in useful photothermal effects [73] and have been 
used to enhance the surface sensitivity of various spectroscopic measurements [74] , 
including fl uorescence, Raman scattering, and second - harmonic generation. 
Metallic nanostructures exhibiting SPR are composed of a dielectric core and 
metallic shell, for example, gold sulfi de dielectric core and gold shell. By varying the 
core – shell thickness ratio, the surface plasmon resonance is shifted from the visible 
to the infrared range [75] , spanning a range that is mostly transparent to human 
tissue, that is, has a high physiological transmissivity. Additionally, control over the 
particle diameter allows control over light scattering and light absorption at particle 
diameters below approximately 75 nm. Potential applications of the photothermal 
effects of engineered nanoparticles include the following: 
Controlled drug delivery [76] 
Analysis of controlled drug release from a matrix [77] 
DNA sensor [78, 79] 
Deep tissue tumor cell thermal ablation [80] 
Real - time assessment of drug action [81, 82] 
Immunosensor applications [83, 84] 
7.3.4.4 Self - Assembled Systems 
Molecular self - assembly is a synthetic technique that has been widely used to 
produce nano - and microstructures in a quick and effi cient manner. It has become 
all the more crucial to the formation of nanostructures due to the control attainable 
over the end product and the relative ease with which nanostructures of defi ned 
structure and function can be produced using bulk manufacturing methods. 
The basic principle of self - assembly is based on the simultaneous coexistence of 
two parallel forces [85, 86] , long - range repulsive forces and short - range attractive 
interactions. 
The types of structures attainable using molecular self - assembly are referred to 
as micellar structures [87] and can take on various sizes and shapes: 
Direct spherical micelles [88] 
Inverse spherical micelles [89, 90] 
DESCRIPTION OF NANOSYSTEMS 1295

1296 PHARMACEUTICAL NANOSYSTEMS 
Lamellar sheets [91] 
Vesicles (hollow or concentric) [92] 
Body - centerd - cubic [92] 
Hexagonally packed cylinders/tubes [92] 
Gyroids [93] 
Hollow spheres [94] 
These systems have found widespread use as drug delivery vehicles, and the more 
advanced nanosystems are termed smart nano - objects due to their ability to sense 
local variations in physiological conditions, such as pH and temperature, and respond 
to the stimulus accordingly. 
7.3.4.5 Nanostructures Based on Carbon 
Nanotubes The structure of carbon nanotubes as observed by scanning tunneling 
microscopy is that of rolled grapheme sheets where endpoints of a translation vector 
are folded one onto another [95] . Single - walled carbon nanotubes (SWCNTs) were 
fi rst reported by Iijima and Ichihashi [96] in 1993. Enormous interest in CNTs has 
centered around their unique properties, including high electrical conductivity, 
thermal conductivity, high strength and aspect ratio, ultralight weight, and excellent 
chemical and thermal stability. 
The most common method for the production of carbon nanotubes is hydrocarbon 
- based chemical vapor deposition (CVD) [97] and adaptations of the CVD 
process [98, 99] , where the nanotubes are formed by the dissolution of elemental 
carbon into metal nanoclusters followed by precipitation into nanotubes [100] . The 
CVD method is used to produce multiwalled carbon nanotubes (MWCNTs) [101] 
and double - walled carbon nanotubes (DWCNTs) [102] as well as SWCNTs [103] . 
The biomedical applications of CNTs have been made possible through surface 
functionalization of CNTs, which has led to drug and vaccine delivery applications 
[104, 105] . 
Fullerenes Fullerenes were fi rst discovered in 1985 [106] and are large molecules 
composed exclusively of carbon atoms and manifest physically in the form of hollow 
spherical cagelike structures. The cages are in the region of 7 – 15 A in diameter with 
the most common form being C 60 , though other forms exist too, such as C 70 , C 76 , and 
C84 , depending on the number of carbon atoms making up the cage. Fullerenes can 
be produced using combustion [107] and arc discharge methods [108] . 
Fullerenes offer numerous points of attachment and allow precise bonding of 
active chemical groups in three - dimensional (3D) conformations and positional 
control with respect to matching conjugated fullerene compounds with a given 
target. Water - soluble fullerenes have shown low biological toxicity both in vitro 
[109] and in vivo [110] . Some of the potential applications of fullerenes in pharmaceuticals 
include their use in neurodegenerative and other disease conditions where 
oxidative stress is part of the pathogenesis due to their powerful antioxidant properties 
[111] and in nuclear medicine for binding of toxic metals ions, increasing therapeutic 
potency of radiation therapy and reducing adverse events as fullerenes do 
not undergo biochemical degradation within the body. Fullerene applications in 
photodynamic tumor therapy have also been shown [112] . 

7.3.5 MANUFACTURING TECHNOLOGIES 
There are a diverse range of technologies being applied to the manufacture of 
nanosystems for pharmaceutical applications. Some of these are derived from 
conventional pharmaceutical technologies, such as colloidal processing, and many 
have been adopted from the semiconductor industry, whereby precision spatial 
control is achieved over the production of nanosystems and particles using fabrication 
techniques. To add to this, new technologies are constantly evolving through 
the adaptation and amalgamation of existing technologies in different fi elds or 
through pure innovation leading to completely new processes. It is outside the 
remit of this chapter to cover in any depth all those manufacturing technologies 
that may be applied to pharmaceutical manufacturing. The summary in Table 3 
provides a detailed synopsis of the different types of manufacturing processes and 
types of technologies for each process. This is followed by a brief introduction to 
some of the technologies, with the omission of silicon - and carbon - based fabrication 
processes, which are beyond the scope of this chapter, to provide the reader 
with a starting point for further detailed study and investigation into those processes 
and technologies that may be most suited to their particular product or 
concept. 
TABLE 3 Summary of Manufacturing Processes and Technologies for Producing 
Nanosystems 
Manufacturing Process Technology 
Nanoscale assembly Self - assembling micellar structures [113, 114] 
Bio - self - assembly and aggregation [115, 116] 
Nanomanipulation [117, 118] 
Soft lithography [119, 120] 
Molecular imprinting [121, 122] 
Layer - by - layer electrostatic deposition [123, 124] 
Chemical vapor deposition [125] 
Nanostructuring processes for 
polymeric materials 
Mold replication [126, 127] 
Colloidal lithography [128, 129] 
X - ray lithography [130] 
Interfacial polymerization [131, 132] 
Nanoprecipitation [133, 134] 
Emulsion solvent evaporation [135] 
Nanoimprinting [136] 
Electrospinning [137, 138] 
Nanostructuring processes for silicon Photolithographic fabrication 
X - ray lithography [130] 
Electron beam lithography [139] 
Chemical etching [140] 
Physical and chemical vapor deposition [141] 
Nanostructuring processes for carbon Electric arc discharge [142, 143] 
Laser ablation [144, 145] 
Chemical vapor deposition [146] 
Combustion [147] 
MANUFACTURING TECHNOLOGIES 1297

1298 PHARMACEUTICAL NANOSYSTEMS 
7.3.5.1 Nanoscale Assembly Methods 1 
Self-Assembly through Micelle Formation Self - assembly at the nanoscale is 
deemed important to be able to produce commercially viable products and processes, 
since it offers a mode of bulk production with control over features such as 
size, shape, and morphology at the nanoscale. The basic principle of self - assembly 
is based on the simultaneous coexistence of two parallel forces: 
Long - range repulsive interactions between incompatible domains 
Short - range attractive interactions 
If we take the example of an amphiphilic diblock copolymer, the polymer is 
composed of two blocks, a hydrophobic block and a hydrophilic block. When introduced 
to a solvent beyond a minimum concentration, the critical micelle concentration 
(CMC), the monomers begin to orientate such that the block that is soluble in 
the solvent orients itself toward the periphery, in contact with the continuous media, 
and the insoluble portion turns toward the core in an attempt to minimize contact 
with the continuous phase, thus leading to the formation of a micelle. The long - range 
repulsive forces arise from the relative solubilities of the blocks in the solvent, and 
the short - range attractive forces arise from the covalent link between the two blocks. 
The basic theory of micelle formation using block copolymers is outlined below 
since nanosystem and nano - object self - assembly is likely to be facilitated by such 
polymer systems, and similar principles will apply or aid toward developing self - 
assembling systems. 
Key factors that affect micelle formation are as follows: 
Equilibrium constant 
Solvent type 
Solvent quality 
Critical micelle temperature (CMT) 
Critical micelle concentration 
Overall molar mass of the micelle, MW 
Micelle aggregation number, Z 
Copolymer architecture 
Relative block lengths 
Relative geometries of copolymer blocks 
Polymer composition 
Core – corona interfacial tension 
These key factors will infl uence the following micelle characteristics: 
Hydrodynamic radius of micelles formed, RH 
Radius of gyration, RG 
1 The description in this section has been summarized and adapted from J. Rodriguez - Hernandez et al., 
Toward “ smart ” nano - objects by self - assembly of block copolymers in solution, Progress in Polymer 
Science , 30 (2005), 691 – 724 [148] . 

Ratio of hydrodynamic radius, RH , to radius of gyration, RH 
Micelle core radius, RC 
Micelle corona thickness, C 
Micellar structures can be produced either by addition of the polymer solution 
or addition of the powdered material to the desired solvent and stirring at the 
optimum temperature and monomer concentration. The CMC can be determined 
by ultraviolet (UV) absorption or light scattering techniques such as static light 
scattering (SLS), dynamic light scattering (DLS), or small - angle X - ray scattering 
(SAXS). At the concentration at which monomers form micelles, there will be a 
radical drop in monomer concentration in the bulk. 
The stability of micellar systems depends upon the ability to ensure the aggregated 
monomers do not deaggregate and that individual micelles do not coalesce 
to form larger aggregates. This is inevitable over a period of time, but steps can be 
taken to prolong the stability of the systems through various techniques, and those 
listed below are some of the methods used for spherical micellar systems: 
Steric stabilization using emulsifi ers and surfactants 
Shell or core cross - linking 
Viscosity - enhanced stabilization 
Amine cross - linked stabilization 
Thermodynamic stabilization 
The types of systems that can be produced using micelle formation include spheres, 
shells, capsules, vesicles, clusters, and particles of various shapes and sizes, such as 
spheres, rods, planar structures, and layered structures. Further processing can be 
undertaken to add rate - controlling polymer membranes to the outer shell and to 
incorporate different molecules to the surface (e.g., for receptor recognition). 
Biological Self -Assembly Using DNA as Construction Tool This is a technique 
that has been adopted to produce 2D or 3D nanosystems by utilizing the base - 
pairing affi nity of DNA [149, 150] . 
Biological self - assembly using DNA can be described as a process that allows the 
systematic assembly of molecules with high levels of precision and accuracy without 
external constraints or infl uences. This allows the construction of nanoscale objects 
to the desired structure, conformation, and composition very rapidly and without 
the need for complex processing techniques and conditions. 
DNA is a copolymer composed of a phosphate and sugar backbone and four 
types of bases that branch off from the backbone, A (adenine), G (guanine), C 
(cytosine), and T (thymidine). During DNA replication two strands of DNA come 
together to form a helical structure through complementary base pairing which is 
highly specifi c, whereby thymidine pairs with adenine and guanine with cytosine. 
When strands of DNA come together where the ends of the strands are noncomplementary, 
a portion of the strand extends beyond the complementary base - paring 
region leading to an overhang, otherwise known as a “ sticky end ” . 
The natural mechanism of DNA base pairing can be used to assemble synthetic 
sequences of DNA molecules by synthesizing DNA molecules such that they form 
MANUFACTURING TECHNOLOGIES 1299

1300 PHARMACEUTICAL NANOSYSTEMS 
stable branches [150, 151] with arms that form sticky ends that in turn can assemble 
to form supramolecular structures [152, 152a] . This approach may be used to produce 
complex 3D assemblies through sequential or layer - by - layer self - assembly and may 
incorporate other materials, such as particles and proteins [153 – 156] . The advantages 
of this method are as follows: 
Specifi city and geometry of intermolecular interactions that can be predicted 
Precision control over the fi nal structure at the nanoscale 
Simple manufacturing process without external restrictions 
Complex structures that can be built with defi ned topologies 
Potential for creation of nanodevices 
The types of structures that may be constructed are Branched planar/2D quadrilateral 
structures [157] , cubes [158] , octahedrons [159] , and complex 2D and 3D 
periodic structures [160] . 
DNA Synthesis for Nanoconstruction Single strands of DNA, otherwise known as 
oligomers, are most commonly produced using a solid - support synthesis process [161, 
162] . This is a cyclic process where each nucleotide is sequentially coupled to form a 
nucleotide chain (working from the 3 . end to the 5 . end). The 3 . end is initially covalently 
linked to a solid support and the nucleotide monomers are added sequentially. 
This is a well - established process and its key parameters and critical process steps are 
well documented in the literature [163, 164] . The DNA strands can be tailored according 
to the desired nanoconstruction scheme and target structure [165] . 
Nano Manipulation As the name suggests, this is quite literally a technique for 
physically manipulating matter at the nanoscale. Scanning probe microscopy (SPM) 
techniques have been most widely used to achieve this using the scanning probe tip 
as an implement for assembling atoms, molecules, or nanoparticles according to the 
desired spatial conformation [166 – 169] . 
Soft Lithography Lithography is essentially a process for printing features on a 
planar surface. Nanolithography tools, commonly referred to as soft lithography, 
allow precisely defi ned nanoscale features to be produced on a substrate, which can 
be removed from the substrate as free - standing 3D nano - objects. A number of 
techniques fall within the fi eld of soft lithography, primarily for construction of 
micrometer - sized objects: 
Replica molding 
Micromolding in capillaries (MIMIC) 
Microtransfer molding 
Solvent - assisted microcontact molding (SAMIM) 
Microcontact printing 
Near - fi eld phase shift lithography is a soft lithographic technique used to produce 
geometric shapes with size features at the nanoscale (approximately 40 – 80 nm). This 
involves the production of a polymer mask containing the desired pattern to be 

replicated on the substrate, with nanoscale features usually patterned by X - ray or 
electron beam exposure. The mask is then placed on the surface of the substrate 
and exposed to near - fi eld light, the intensity of which leads to replication of the 
pattern on the mask on to the substrate. Complex geometries, shapes, and features 
can be produced on the substrate which can subsequently be removed to give free - 
standing particles or objects [170, 171] . 
Molecular Imprinting Molecular Imprinting is a process used to imprint or copy 
recognition sites from desired molecules on to polymer structures [172, 173] . The 
recognition sites can be produced on organic or inorganic polymers and inorganic 
materials such as silica and biomaterials such as proteins. A template molecule is 
dissolved in solvent with polymerizable monomers which undergo bond formation 
with the template molecule forming either noncovalent bonds through electrostatic 
interactions, hydrogen bonds or hydrophobic interactions, or reversible covalent 
bonds. The monomers are then polymerized to form a cast or semirigid polymeric 
structure which maintains the steric conformation of the molecule template and its 
recognition site upon removal of the template molecule. As a result, the molecular 
template affi nity for molecules and analyte is mimicked by the “ imprinted ” polymer 
[174] . This has applications in chromatography and drug discovery and potential 
applications in targeted drug delivery. 
Layer -by -Layer Electrostatic Deposition Electrostatic deposition utilizes the 
electrostatic bonding affi nities of materials imparted by their surface charge to build 
highly ordered multilayered fi lms or structures on a substrate. The process involves 
the successive deposition of oppositely charged polyions, exploiting the Coulombic 
long - range electrostatic interactions between the oppositely charged molecules, 
allowing formation of multilayers over a large distance. This technique can be used 
to build multilayer composite fi lms on particles incorporating molecular fragments 
such as polymer – polymer, polymer – organic, polymer bimolecular, and polymer – 
mineral composition [175 – 177] . 
Chemical Vapor Deposition CVD is a crystal growth process whereby a solid 
material is deposited from the gas phase onto a controlled substrate using a suitable 
mixture of volatile precursor materials which react to produce the desired deposit 
on the substrate surface (Table 4 ). Types of fi lms and structures that can be produced 
include the following: 
Polycrystalline 
Amorphous 
Epitaxial silicon 
Carbon fi ber 
Filaments 
Carbon nanotubes 
Silicon dioxide 
Tungsten 
Silicon nitride 
Titanium nitride 
MANUFACTURING TECHNOLOGIES 1301

1302 PHARMACEUTICAL NANOSYSTEMS 
7.3.5.2 Nanostructuring Processes for Polymeric Materials 
Numerous microfabrication techniques have been used to produce a wide range of 
implantable and oral drug delivery systems using materials ranging from silicon, 
glass, silicone elastomer, and plastics. Fabrication techniques have rapidly evolved 
to produce nanoscale objects and therapeutic systems using polymeric materials as 
the substrate due to their biodegradable nature. There are a number of different 
synthetic polymer systems that have been developed for this type of application, 
and the most common ones are listed below: 
Poly( d - lactic acid) (PDLA) 
Poly( . - caprolactone) (PCL) 
Poly(vinyl alcohol) (PVA) 
Polyalkylcyanoacrylates (PACA) 
Poly( l - lactide) (PLLA) 
Poly(lactide - co - glycolide) (PLGA) 
Polymethylcyanoacrylate (PMCA) 
Techniques for the production of micrometer - sized features using polymers are 
well established and apply primarily to device construction. The techniques listed 
TABLE 4 Chemical Vapour Deposition Methods and Their Key Features 
CVD Method Key Features 
Atmospheric pressure CVD [178, 
179] 
Operates at atmospheric pressure 
Atomic layer CVD (atomic layer 
epitaxy) [180, 181] 
High - precision fi lm thickness and uniformity 
requirements 
Aerosol - assisted CVD [182, 183] For use with involatile precursors 
Direct liquid injection CVD [184, 
185] 
High fi lm growth rates possible 
Hot - wire CVD [186] High growth rate, low temperature, and use 
of inexpensive materials such as plastics 
as substrate 
Low - pressure CVD [187] Improved fi lm uniformity 
Metal organic CVD [188] Uniform and conformal deposition 
Microwave plasma - assisted CVD 
[189] 
No external heating required 
Plasma - enhanced CVD [190] Reduced substrate temperatures can be 
used 
Rapid thermal CVD [191] Conformal coverage over high - aspect - ratio 
features is possible, i.e., improved control 
of interfacial properties 
Remote plasma - enhanced CVD 
[192] 
Excellent conformal coverage of complex 
structures 
Can produce multilayer and graded layers 
with tailored functional group attachment 
Ultrahigh vacuum CVD [193] Reduced surface contamination 

below focus primarily on attaining submicrometer, nanoscale features, and geometries 
using polymers such as those listed above. 
Nanomold Replication A physical mold is produced that has nanoscales on the 
order of tens or a few hundred nanometers. To achieve such fi ne features with 
precision and repeatability, electrodeposition is used to produce the molds, 
otherwise referred to as a nanostamp [194] . The stamp is then use as a master 
stamp to duplicate the image or object by casting or embossing the polymeric 
material. 
Colloidal Lithography Colloidal lithography is a process whereby an electrostatically 
self - assembled array of monodispersed colloidal nanospheres is used as a mask 
to construct nanoscale objects and features through deposition or etching processes. 
The monodisperse colloidal spheres, for example, surface - charged latex, self - 
organize or assemble into periodic arrays on the substrate, glass, for example, and 
do not aggregate due to the surface charge repulsion. This method can be used to 
produce 2D [195, 196] and 3D [197] nanostructures, arrays of rings, dots, honeycomb 
structures, pillars, and chemical patterns [198] with a high level of control over 
structure and conformation. 
Interfacial Polymerization Interfacial polymerization is a process whereby very 
thin fi lms or membranes, on the order of nanometer thickness, are produced by 
reacting two monomers at the interface between two immiscible solutions [199] . 
Nanoparticles [200] and aqueous core capsules with very thin membranes have been 
produced using this method for drug delivery applications. 
Nanoprecipitation Nanoprecipitation is a self - assembly directed nanoparticle formation 
method. There are three key steps involved in this process: rapid micromixing 
of the solutes, the creation of a high level of supersaturation to instigate rapid 
nucleation and growth of precipitate, and the kinetic control and termination of 
growth using copolymer stabilizers. One of the drawbacks using this method is the 
poor incorporation of water - soluble drugs [201] . However, the main advantage 
associated with the production of nanospheres for drug delivery using this technique 
is the high degree of control attainable over particle size [202] . 
Emulsion Solvent Evaporation The basic concept of the emulsion solvent evaporation 
technique producing nanoparticles is very straightforward. The particles are 
formed as an emulsion of a polymer – surfactant mixture and dispersed in an organic 
solvent. The solvent is then evaporated to leave behind the individual emulsion 
droplets which form stable free nanoparticles [203] . This method is far easier and 
more preferable over methods such as spray drying and homogenization and operates 
under ambient conditions and mild emulsifi cation conditions. The size and 
composition of the fi nal particles are affected by variables such as phase ratio of 
the emulsion system, organic solvent composition, emulsion concentration, apparatus 
used, and properties of the polymer [204] . 
Nanoimprinting This is a lithographic technique similar to soft contact lithography 
discussed earlier, with the main difference being that nanoimprinting uses a 
MANUFACTURING TECHNOLOGIES 1303

1304 PHARMACEUTICAL NANOSYSTEMS 
hard mold to produce nanoscale features down to sub - 10 nm resolution [205] by 
directly imprinting onto the polymer surface at high temperatures. More recently, 
molds produced from carbon nanotubes have been used to achieve molecular - scale 
resolution. Molds are generally made using electron beam lithography; however, 
high - defi nition molds are produced using molecular beam epitaxy. Some of the 
technical issues associated with this technique include sticking, adhesion, and material 
transport during imprinting [206] . 
Electrospinning Electrospinning is a process that uses electrostatic force to 
produce nanofi bers from a charged polymer. An electrode is placed into a spinning 
polymer solution/polymer melt and the other electrode is attached to a collector 
plate. A high - intensity electric fi eld is created by applying a high voltage such that 
the polymer solution is discharged as a jet, and during travel of this charged polymer 
jet toward the grounded collector plate, solvent evaporation leaves a charged 
polymer fi ber which deposits on the collector plate [207] . These fi bers have high 
specifi c surface areas and are highly fl exible, and applications include the preparation 
of controlled drug release membranes [208] . 
7.3.6 CHARACTERIZATION TECHNIQUES 
7.3.6.1 Nanoparticle Characterization Methods and Tools 
A summary of some key properties that may be assessed as part of a characterization 
schedule for nanoparticles and nanostructures and a comprehensive but not 
exhaustive list of tools and techniques that may be used are presented in Table 5 
[209 – 223] . The degree of characterization and method used will be determined by 
the intended application of the nanomaterials. 
Characterization of micellar and supramolecular structures and their counterparts 
often require different or additional tools and techniques [224 – 232] and a 
summary is provided in Table 6 of various characterization parameters for micellar 
and supramolecular structures and components and analytical tools that may be 
applied. 
7.3.6.2 Scanning Probe Technologies 
Scanning probe microscopy has almost become synonymous with nanomaterial 
characterization [233] . This is a family of techniques that have evolved from the use 
of a sharp proximal probe to scan a surface in order to ascertain its properties down 
to atomic - scale resolution based on tip – surface interaction. There are two main SPM 
techniques, scanning tunneling microscopy (STM) [234, 235] and AFM [236] . Near - 
fi eld scanning optical microscopy (NSOM) [237 – 239] also falls within the SPM 
family of techniques; however, this uses a subwavelength near - fi eld light source as 
the scanning probe, achieving resolutions down to 50 nm, and is not discussed further 
here. 
A host of techniques have evolved from STM and AFM, primarily involving 
adaptations to instrumentation depending on the material and parameter under 

TABLE 5 Characterization Parameters and Tools for Nanoparticles and Nanostructures 
Characterization Parameter Analytical Tool 
Composition Liquid chromatography, e.g., high - performance liquid 
chromatography (HPLC), size exclusion chromatography 
(SLC) – HPLC 
Field fl ow fractionation (FFF) 
UV – visible spectrophotometry 
Refractive Index 
Inductively coupled plasma – optical emission spectrometry 
(ICP – OES) 
Fourier transform infrared spectrometry 
Mass spectrometry 
X - ray fl uorescence 
Extended X - ray absorption fi ne structure (EXAFS) 
spectroscopy 
X - ray absorption near edge (XANES) spectroscopy 
Particle diameter Static and dynamic laser light scattering 
Scanning probe technologies 
Size distribution Static and dynamic laser light scattering 
Photon correlation spectroscopy 
Surface area BET method (Brunauer, Emmett, and Teller method) 
Porosity (pore size, volume, 
and distribution) 
Physical gas sorption 
Chemical gas sorption 
Helium picnometry 
Mercury intrusion porometry (MIP) 
Core – shell thickness Small - angle scattering of polarized neutrons (SANSPOL) 
Surface structure and 
morphology 
Small - angle neutron scattering (SANA) 
Proton nuclear magnetic resonance ( 1 H NMR) spectroscopy 
Scanning electron microscoscopy (SEM), atomic force 
microscopy (AFM), energy dispersive X - ray (EDXA), 
transmission electron microscopy (TEM), scanning probe 
microscopy (SPM), auger electron spectroscopy (AES), 
X - ray diffraction (XRD), X - ray photoelectron 
microscopy (APS), X - ray photoelectron spectroscopy 
(XPS), Vertical scanning phase shifting interferometry 
Surface charge density Zeta potential using Electrostatic light scattering (ELS) 
Zeta potential using multifrequencyelectro acoustics 
Zeta potential using phase analysis light scattering (PALS) 
Shape Electron microscopy 
Scanning probe technologies 
Concentration distribution Energy dispersive X - ray spectrometry (EDS) combined 
with SEM or scanning 
Crystallinity, Bulk X - ray diffraction 
Crystallinity, Local TEM/ – selected area diffraction (SAD) 
Differential scanning calorimetry (DSC) 
Magnetic properties Scanning probe technologies 
Electrical properties Scanning probe technologies 
Optical properties UV – visible Spectroscopy 
CHARACTERIZATION TECHNIQUES 1305

1306 PHARMACEUTICAL NANOSYSTEMS 
TABLE 6 Characterization Parameters and Analytical Tools for Micellar and 
Supramolecular Structures 
Characterization Parameter Analytical Tool 
Critical micelle concentration Flurimetric methods 
Static light scattering 
Dynamic Light Scattering 
Aggregation number Fluorescence correlation spectroscopy 
Radius of gyration, R G Small - angle X - ray scattering 
Hydrodynamic radius, R H Photon correlation spectroscopy 
Core/corona size, micelle structure, overall 
micelle size 
Small - angle X - ray scattering 
Overall shape, cross section Small - angle neutron scattering 
Size, shape, and internal structure Transmission electron microscopy 
Scanning probe technologies 
Average molecular weight Membrane and vapor pressure osmometry 
Monitor equilibrium state, stability monitoring Light - scattering methods 
Structure elucidation, polymer architecture, 
polymer interactions 
Nuclear magnetic resonance 
investigation. It should be noted that technological advances continue unabated and 
new techniques are constantly being developed within the scanning probe family to 
cater for the characterization of new and novel materials and nanoscopic constructs. 
Table 7 gives a current synopsis of these techniques. 
Scanning Tunneling Microscopy The scanning tunneling microscope was fi rst 
described by Nobel Prize winners Binnig and Rohrer in 1982 [249] and consists of 
an atomically sharpened tip usually composed of tungsten, gold, or platinum – irridium. 
The tip is scanned within atomic distance (about 6 – 10 A ) of the sample under 
study under very high vacuum, and a bias voltage is applied between the sample 
and the scanning probe tip, resulting in a quantum mechanical tunneling current 
across the gap. The magnitude of the tunnelling current is related exponentially to 
the distance of separation and the local density of states (i.e., electron density in a 
localized region of a material) [250, 251] . 
The relationship between tunneling current and separation is given as: 
I C ed = ..t s 
0 5 . 
where I = tunnelling current 
C = constant (linear function of voltage) 
. t = tip electron density 
. s = sample electron density 
e d 0.5 = separation (governed by exponential term) 
The tip is scanned across the sample surface using a piezoelectric transducer in one 
of two modes, topographic mode or current mode. In the topographic mode a con

TABLE 7 Summary of Scanning Probe Technologies 
SPM Technique Property Measured 
Atomic force microscopy Visualisation and measurement of surface features 
Noncontact AFM [240] Insulating substrates, atomic resolution 
Molecular systems, atomic resolution 
Biocluster and biomolecular imaging 
Imaging and spectroscopic data in liquid environments 
Nanoscale charge measurement 
Nanoscale magnetic properties 
Contact AFM [241, 242] Topographic imaging of solid substrates 
Mechanical properties 
Local adhesive properties 
Piezoresponse Characterization and domain engineering of ferroelectric 
materials 
Lateral force Fine structural detail 
Transitions between components on surface, e.g., polymer 
composites 
Scanning thermal Defects in sample based on thermal differences 
Intermittent AFM (tapping 
mode) [243] 
Biological systems: DNA/RNA analysis, protein – nucleic acid 
complexes, molecular crystals, biopolymers, ligand – 
receptor binding 
Phase imaging AFM Two phase polymer blends 
Surface contaminants 
Biological samples 
Lift mode AFM techniques 
[244 – 246] 
Topography 
Magnetic Force Magnetic properties/regions 
Electrical Force Electrical properties 
Surface Potential Surface potential 
Scanning Capacitance Material capacitance 
Force modulation Elasticity 
Scanning tunnelling 
microscopy [247] 
Surface imaging 
Three - dimensional profi ling with vertical resolutions to 0.1 A 
Measurement of electronic and magnetic properties 
Surface electronic state 
Spin - polarized STM/STS 
[248] 
Mapping surface magnetism at atomic scale 
stant distance is maintained between the tip and sample surface using a feedback 
loop operated with the scanner. In the current mode, variations in current with 
changes in surface topography are monitored by switching off the feedback loop, 
thus providing a 3D image of the surface under study. The key features and limitation 
of STM are as follows: 
Features of STM [252, 253] 
Can undertake topographical imaging of surfaces with atomic - scale lateral resolution, 
down to 1 A 
There - dimensional profi ling possible with vertical resolutions down to 0.1 A 
Wide range of materials can be analyzed 
CHARACTERIZATION TECHNIQUES 1307

1308 PHARMACEUTICAL NANOSYSTEMS 
Surface electronic properties may be measured 
Large fi eld of view, from 1 A to 100 . m 
Vibrational isolation allows highly sensitive measurements to be undertaken 
Ultrahigh vacuum (in the range 10 . 11 torr) minimizes sample contamination and 
reduces oxide layer growth, thus allowing for high sensitivity measurements 
Limitations of STM 
Can be diffi cult to differentiate between a composite of materials on the 
surface 
Tip - induced desorption of surface molecules may occur 
Ultrahigh vacuum requirements 
Vibrational isolation requirements lead to increased installation costs 
Low scanning speed 
Atomic Force Microscopy Atomic force microscopy is a direct descendant of STM 
and was fi rst described in 1986 [254] . The basic principle behind AFM is straightforward. 
An atomically sharp tip extending down from the end of a cantilever is 
scanned over the sample surface using a piezoelectric scanner. Built - in feedback 
mechanisms enable the tip to be maintained above the sample surface either at 
constant force (which allows height information to be obtained) or at constant 
height (to enable force information to be obtained). The detection system is usually 
optical whereby the upper surface of the cantilever is refl ective, upon which a laser 
is focused which then refl ects off into a dual - element photodiode, according to the 
motion of the cantilever as the tip is scanned across the sample surface. The tip is 
usually constructed from silicon or silicon nitride, and more recently carbon nanotubes 
have been used as very effective and highly sensitive tips. 
In noncontact - mode AFM the cantilever is oscillated slightly above its resonant 
frequency and the tip does not make contact with the sample surface but instead 
oscillates just above the adsorbed fl uid layer on the surface, maintaining a constant 
oscillation. The resonant frequency of the cantilever decreases due to van der Waals 
forces extending from the adsorbed fl uid layer. This changes the amplitude of oscillation, 
the variations of which are detected using sensitive alternating current (AC) 
phase - sensitive devices, providing topographical information. In contact mode, AFM 
the tip remains in contact with the sample surface, and the feedback loop maps the 
vertical vibrational changes. In tapping mode, the cantilever is oscillated above the 
sample surface such that it intermittently contacts the sample surface. The key features 
and limitations of AFM are a follows: 
Features of AFM [255 – 257] 
High scan speeds 
Atomic - scale resolution possible 
Rough sample surfaces can be analyzed 
High lateral resolutions possible 
Soft samples (e.g., biological tissue) can be measured 

Limitations of AFM [255 – 257] 
Potential for image distortion due to lateral shear forces (in contact mode) 
May be reduced spatial resolution due to sample scraping (in contact mode) 
Tapping mode has lower scan speed compared to contact mode, though there is 
less susceptibility for sample damage and image distortion 
7.3.7 TOXICOLOGY CONSIDERATIONS 
Nanomaterials may in their own right possess novel and useful properties or as a 
composite of the same or different materials to form larger useful structures. Safety 
consideration is therefore of paramount importance since completely inert materials 
have the ability to exhibit toxic effects by virtue of a reduction in their size and 
associated increase in surface area – mass ratio, let alone materials manipulated 
specifi cally to impart novel properties. 
Two obvious routes of human contact with nanoparticulates are the skin and via 
inhalation. Given the size of the particles, there may be a propensity for absorption 
into the systemic circulation. In some cases the nanosystems are engineered to 
achieve enhanced systemic absorption. The established methodology for toxicological 
assessment of new materials should be adhered to, and the discussion below is 
intended only to touch upon some of the immediate safety concerns that should be 
understood and addressed when dealing with nanomaterials. 
7.3.7.1 Lung Toxicity 
The safety of ultrafi ne particles remains to be clearly elucidated and requires the 
collaborative input of toxicologists (animal, cellular, molecular), epidemiologists, 
clinicians (pulmonary, cardiovascular, neurological), and atmospheric scientists. 
There are several published studies to indicate that ultrafi ne particles pose a higher 
toxicity risk compared with their larger counterparts [258 – 261] . Figure 1 outlines 
the potential effects of ultrafi ne particles on respiratory mucosa, the cardiovascular 
system, and central and peripheral nervous systems, upon inhalation. 
Hohfeld et al. [262] hypothesized the toxic effects of ultrafi ne particles to be 
attributable to the following: 
• High effi ciency of deposition in the alveolar region due to particle size 
• Large surface area 
• Decreased phagocytosis leading to interaction of the particles with the epithelium, 
resulting in the development of conditions such as chronic diffuse interstitial 
fi bronodular lung disease 
• Dislocation from the alveolar space, leading to potential systemic effects 
The hypothesis affi rms the need to characterize the material ’ s physical and 
chemical properties, including morphological analysis. The latter has signifi cant 
ramifi cations on the aerodynamics of the particulate matter and hence its ultimate 
disposition. 
TOXICOLOGY CONSIDERATIONS 1309

1310 PHARMACEUTICAL NANOSYSTEMS 
Carbon nanotubes are a class of materials fi nding increasingly widespread applications 
in drug discovery and development and may be classed as a form of ultrafi ne 
material. It has been shown that single - wall carbon nanotubes do not produce any 
signifi cant respirable aerosol levels due to agglomeration resulting from the very 
high surface area – volume ratio and associated electrostatic interaction between the 
nanotubes [263] . A good deal of research has focused on developing methods for 
the dispersion of nanotubes for further downstream processing for conversion to 
useful applications. The liquid dispersions do not pose the same level of hazards 
posed by the dry powder material, and most of the work in the pharmaceutical 
industry with carbon nanotubes is focused on liquid dispersions whereby nanotubes 
are being functionalized and conjugated with drugs and possibly other carriers for 
therapeutic intent [264] . 
However, it has at the same time been shown that dry powder carbon nanotubes 
can persist in the lungs and have the potential to induce infl ammatory and fi brotic 
reactions, evident in the form of collagen - rich granulomas in the bronchi and interstitium 
[265] . This emphasizes the need for caution and further work to establish 
the exact cause of these effects given the propensity of nanotubes to agglomerate. 
7.3.7.2 Systemic Uptake 
Nanoparticles, by virtue of size, have a tendency to evade phagocytosis. Uptake into 
the systemic circulation is thus thought to be through diffusion and via the endo- 
FIGURE 1 Potential mechanisms of effects of inhaled ultrafi ne particles. ( Reproduced with 
permission from G. Oberd o rster, Inhaled nano - sized particles: Potential effects and mechanisms, 
paper presented at the Symposium, Health Implications of Nanomaterials, October 
2004 .) 
Particle translocation 
Mediators Inhalation 
Neurons 
Interstitium 
Respiratory tract deposition 
Lung 
inflammation 
Blood vessel 
dysfunction Systemic 
inflammation 
Heart effects 
Modifying factors: Age, gender, underlying disease, copollutants 
CNS 
(effects ?) 
Autonomic nervous system 
Circulation 
Extrapulmonary 
organs 
Liver Heart 
White blood 
cell activation

REFERENCES 1311 
thelial cells, the epithelium, interstitium, and blood vessels. Translocation into the 
blood is thought to be through enhanced epithelial or endothelial permeability 
imparted by infl ammatory mediators. Systemic hypercoagulation may be triggered 
by the infl ammatory mediators in response to the diffusion of the nanoparticles 
through endothelium and vasculature [260] . 
7.3.7.3 Skin Permeation of Nanoparticles 
The stratum corneum provides a formidable barrier to the entry of chemical and 
particulate matter into human tissue and systemic circulation. It provides a fi rst - line 
defence to the ingress of foreign agents. However, there are indications that particles 
up to 1 . m are able to penetrate the skin ’ s barrier and deposit in the epidermis 
where the antigen - presenting cells reside [266] . It follows therefore that submicrometer 
particles in the nanometer range have the potential to cross the stratum 
corneum and illicit an infl ammatory response. Once again, however, the tendency 
of fi ne particles to agglomerate will to some extent inhibit penetration into the skin, 
in particular where the agglomerates are macroscopic. A correlation must however 
be drawn to establish any potential link between nanoparticle affi nity for skin penetration 
and particle physical and morphological characteristics or indeed whether 
the novel and unique properties of the engineered particle in any way impart 
enhanced skin permeation properties and, if so, their nature and mechanisms. 
REFERENCES 
1. FDA and Nanotechnology Products . U.S. Food and Drug Administration (FDA), available: 
http://www.fda.gov/nanotechnology/faqs.html , accessed June 2006 . 
2. Freitas , R. A. , Jr. ( 2005 ), What is nanomedicine? Nanomed. Nanotechnol. Biol. Med. , 1 , 
2 – 9 . 
3. Yang , L. , et al. ( 2000 ), Physicochemical aspects of drug delivery and release from 
polymer - based colloids , Curr. Opin. Coll. Interf. Sci. , 5 ( 1 – 2 ), 132 – 143 . 
4. Bontha , S. , et al. ( 2006 ), Polymer micelles with cross - linked ionic cores for delivery of 
anticancer drugs, J. Controlled Release , 114 ( 2 ), 163 – 174 . 
5. Sphurti , V. , et al. ( 2006 ), Nanofi bers and spheres by polymerization of cyanoacrylate 
monomer , Polymer , 47 ( 12 ), 4328 – 4332 . 
6. Qian , F. , et al. ( 2006 ), Preparation, characterization and enzyme inhibition of methylmethacrylate 
copolymer nanoparticles with different hydrophilic polymeric chains , 
Eur. Polym. J. , 44 ( 7 ), 1653 – 1661 . 
7. Choi , C. , et al. ( 2006 ), Thermosensitive poly( N - isopropylacrylamide) - b - poly( . - caprolactone) 
nanoparticles for effi cient drug delivery system , Polymer , 47 ( 13 ), 4571 – 4580 . 
8. Gross , M. , and Maskos , M. ( 2005 ), Dye loading of unimolecular, amphiphilic polymeric 
nanocontainers , Polymer , 46 ( 10 ), 3329 – 3336 . 
9. Bro z , P. , et al. ( 2005 ), Cell targeting by a generic receptor - targeted polymer nanocontainer 
platform , J. Controlled Release , 102 ( 2 ), 475 – 488 . 
10. Santoso , S. S. , et al. ( 2002), Structures, function and applications of amphiphilic peptides , 
Curr. Opin. Coll. Interf. Sci. , 7 ( 5 – 6 ), 262 – 266 . 
11. Heyes , J. , et al. ( 2006 ), Synthesis and characterization of novel poly(ethylene glycol) - 
lipid conjugates suitable for use in drug delivery , J. Controlled Release , 112 ( 2 ), 
280 – 290 . 

1312 PHARMACEUTICAL NANOSYSTEMS 
12. Seki , J. , et al. ( 2004 ), A nanometer lipid emulsion, lipid nano - sphere (LNS ® ), as a parenteral 
drug carrier for passive drug targeting , Int. J. Pharm. , 273 ( 1 – 2 ), 75 – 83 . 
13. Carlson , J. C. T. , and Jena , S. S. ( 2006 ), Chemically controlled self - assembly of protein 
nanorings , J. Am. Chem. Soc. , 128 ( 23 ), 7630 – 7638 . 
14. Perkins , W. R. , et al. ( 2000 ), Novel therapeutic nano - particles (lipocores): Trapping 
poorly water soluble compounds , Int. J. Pharm. , 200 ( 1 ), 27 – 39 . 
15. Fukui , H. , et al. ( 2003 ), A novel delivery system for amphotericin B with lipid nano - 
sphere (LNS ® ) , Int. J. Pharm. , 265 ( 1 – 2 ), 37 – 45 . 
16. Muller , R. H. , et al. (2006), Oral bioavailability of cyclosporine: Solid lipid nanoparticles 
(SLN ® ) versus drug nanocrystals , Int. J. Pharm. , 317 ( 1 ), 82 – 89 . 
17. Casadei , M. A. , et al. ( 2006 ), Solid lipid nanoparticles incorporated in dextran hydrogels: 
A new drug delivery system for oral formulations, Int. J. Pharm , 325 ( 1 – 2 ), 140 – 146 . 
18. Masuda , M. , and Shimizu , T. ( 2004 ), Lipid nanotubes and microtubes: experimental 
evidence for unsymmetrical monolayer membrane formation from unsymmetrical 
bolaamphiphiles , Langmuir , 20 ( 14 ), 5969 – 5977 . 
19. Gupta , K. , et al. ( 2006 ), Nanoparticle formation from poly(acrylic acid) and oppositely 
charged peptides , Biophys. Chem. , 119 ( 3 ), 303 – 306 . 
20. Costantino , L. , et al. ( 2005 ), Peptide - derivatized biodegradable nanoparticles able to 
cross the blood – brain barrier , J. Controlled Release , 108 ( 1 ), 84 – 96 . 
21. Cortez - Retamozo , V. , and Backmann , N. ( 2004 ), Effi cient cancer therapy with a 
nanobody - based conjugate , Cancer Res. , 64 , 2853 – 2857 . 
22. Parekh , H. S. , et al. ( 2006 ), Synthesis of a library of polycationic lipid core dendrimers 
and their evaluation in the delivery of an oligonucleotide with hVEGF inhibition , 
Bioorg. Med. Chem. , 14 ( 14 ), 4775 – 4780 . 
23. Gupta , U. , et al. ( 2006 ), A review of in vitro – in vivo investigations on dendrimers: The 
novel nanoscopic drug carriers , Nanomed. Nanotechnol. Biol. Med. , 2 ( 2 ), 66 – 73 . 
24. Najlah , M. , et al. ( 2006 ), Synthesis, characterization and stability of dendrimer prodrugs , 
Int. J. Pharm. , 308 ( 1 – 2 ), 175 – 182 . 
25. Koga , T. , et al. ( 2006 ), Fabrication of a switchable nano - surface composed of acidic 
and basic block - polypeptides , Coll. Surf. Physicochem. Eng. Aspects , 284 – 285 , 521 – 
527 . 
26. Yin , A. , and Xu , A. ( 2006 ), Fabrication of highly - ordered and densely - packed silicon 
nano - needle arrays for bio - sensing applications , Mater. Res. Soc. Symp. Proc. , 900E . 
27. Vaccari , L. , et al. ( 2006 ), Porous silicon as drug carrier for controlled delivery of doxorubicin 
anticancer agent , Microelectron. Eng. , 83 ( 4 – 9 ), 1598 – 1601 . 
28. Venkatesan , N. , et al. ( 2005 ), Liquid fi lled nanoparticles as a drug delivery tool for 
protein therapeutics , Biomaterials , 26 ( 34 ), 7154 – 7163 . 
29. Xu , Z. P. , et al. ( 2006 ), Inorganic nanoparticles as carriers for effi cient cellular delivery , 
Chem. Eng. Sci. , 61 ( 3 ), 1027 – 1040 . 
30. Neuberger , T. , et al. ( 2005 ), Superparamagnetic nanoparticles for biomedical applications: 
Possibilities and limitations of a new drug delivery system , J. Magnet. Magnetic 
Mater. , 293 ( 1 ), 483 – 496 . 
31. Kim , J - H. , and Lee , T. R. ( 2004 ), Discrete thermally responsive hydrogel - coated gold 
nanoparticles for use as drug - delivery vehicles , Chem. Mater , 16 , 3647 – 3651 . 
32. Gao , X. , et al. ( 2004 ), In vivo cancer targeting and imaging with semiconductor quantum 
dots , Nat. Biotechnol. , 22 , 8 . 
33. Wu , C. , et al. ( 2005 ), Metal nanoshells as a contrast agent in near - infrared diffuse optical 
tomography , Opt. Commun. , 253 ( 1 – 3 ), 214 – 221 . 

REFERENCES 1313 
34. Ostrander , K. D. , et al. (1999), An in-vitro assessment of a NanoCrystal™ beclomethasone 
dipropionate colloidal dispersion via ultrasonic nebulization , Eur. J. Pharm. Biopharm. 
, 48 ( 3 ), 207 – 215 . 
35. Muller , R. H. , and Keck , C. M. ( 2004 ), Challenges and solutions for the delivery of 
biotech drugs — A review of drug nanocrystal technology and lipid nanoparticles , 
J. Biotechnol. , 113 ( 1 – 3 ), 151 – 170 . 
36. Cui , F. , et al. ( 2006 ), Preparation and characterization of mucoadhesive polymer - coated 
nanoparticles , Int. J. Pharm. , 316 ( 1 – 2 ), 154 – 161 . 
37. Goodson , T. , et al. ( 2004 ), Optical properties and applications of dendrimer - metal nanocomposites 
, Int. Rev. Phys. Chem. , 23 , 1 . 
38. Panchapakesan , B. ( 2005 ), Tiny technology — Tremendous therapeutic potential, in 
Oncology Issues, Nanotechnology: Part 2 . 
39. Lin , C - R. , et al. ( 2006 ), Magnetic behavior of core – shell particles , J. Magnet. Magnetic 
Mater. , 304 ( 1 ), 34 – 36 . 
40. Layre , A. , et al. ( 2006 ). Novel composite core - shell nanoparticles as busulfan carriers , J. 
Controlled Release , 111 ( 3 ), 271 – 280 . 
41. Foley , S. , et al. ( 2002 ), Cellular localisation of a water - soluble fullerene derivative , 
Biochem. Biophys. Res. Commun. , 294 ( 1 ), 116 – 119 . 
42. Friedman , S. H. , et al. ( 2002 ), Optimizing the binding of fullerence inhibitors of the 
HIV - 1 protease through predicted increases in hydrophobic desolvation , J. Med. Chem. , 
41 , 2424 – 2429 . 
43. Zhu , Z. , et al. ( 2003 ), Molecular dynamics study of the connection between fl ap closing 
and binding of fullerene - based inhibitors of the HIV - 1 protease , Biochemistry , 42 , 
1326 – 1337 . 
44. Smart , S. K. , et al. ( 2006 ), The biocompatibility of carbon nanotubes , Carbon , 44 ( 6 ), 
1034 – 1047 . 
45. Bianco , A. , et al. ( 2005 ), Applications of carbon nanotubes in drug delivery , Curr. Opin. 
Chem. Biol. , 9 ( 6 ), 674 – 679 . 
46. Luan , B. , and Pan , C - Y. ( 2006 ), Synthesis and characterizations of well - defi ned 
dendrimer - like copolymers with the second and third generation based on polystyrene 
and poly( l - lactide) , Eur. Polym. J. , 42 , 1467 – 1478 . 
47. Tomalia , D. A. , et al. ( 1990 ), Starburst dendrimers: Control of size, shape, surface chemistry, 
topology and fl exibility , Angew. Chem., Int. Ed. Engl. , 29 , 138 . 
48. Petkov , V. , et al. ( 2005 ), 3D structure of dendritic and hyper - branched macromolecules 
by X - ray diffraction , Solid State Commun. , 134 ( 10 ), 671 – 675 . 
49. Tomalia , D. A. , et al. ( 1985 ), A new class of polymers: Starburst - dendritic macromolecules 
, Polym. J. , 17 , 117 – 132 . 
50. Buhleier , E. , et al. (1978), Cascade and nonskid-chain-like syntheses of molecular cavity 
topologies , Synthesis , 155 – l58 . 
51. Tomalia , D. A. , et al. ( 1991 ), Comb - burst dendrimer topology. New macromolecular 
architecture derived from dendritic grafting , Macromolecules , 24 , 1435 – 1438 . 
52. Hawker , C. J. , and Frechet , J. M. J. ( 1990 ), Preparation of polymers with controlled 
molecular architecture. A new convergent approach to dendritic macromolecules , J. Am. 
Chem. Soc. , 112 , 7638 – 7647 . 
53. Miller , T. M. , and Neenan , T. X. ( 1990 ), Convergent synthesis of monodisperse dendrimers 
based upon 1,3,5 trisubstituted benzenes , Chem. Mater. , 2 , 346 – 349 . 
54. Hedrick , J. L. , et al. ( 1998 ), Dendrimer - like star block and amphiphilic copolymers by 
combination of ring - opening and atom transfer radical polymerization , Macromolecules , 
31 , 8691 – 8705 . 

1314 PHARMACEUTICAL NANOSYSTEMS 
55. Hou , S. J. , et al. ( 2003 ), Synthesis of water - soluble star - block and dendrimer - like 
copolymers based on poly(ethylene oxide) and poly(acrylic acid) , Macromolecules , 36 , 
3874 – 3881 . 
56. Angot , S. , et al. ( 2000 ), Amphiphilic stars and dendrimer - like architectures based on 
poly(ethylene oxide) and polystyrene , Macromolecules , 33 , 5418 – 5426 . 
57. Reuter , J. D. , et al. ( 1999 ), Inhibition of viral adhesion by sialic - acid - conjugated dendritic 
polymers , Bioconjug. Chem. , 10 , 271 – 278 . 
58. Na , M. , et al. ( 2006 ), Dendrimers as potential drug carriers. Part II. Prolonged delivery 
of ketoprofen by in vitro and in vivo studies , Eur. J. Med. Chem. , 41 ( 5 ), 670 – 674 . 
59. Vandamme , Th. F. , and Brobeck , L. ( 2005 ), Poly(amidoamine) dendrimers as ophthalmic 
vehicles for ocular delivery of pilocarpine nitrate and tropicamide , J. Controlled Release , 
102 ( 1 ), 23 – 38 . 
60. Zeng , H. , et al. (2002), Exchange-coupled nanocomposite magnets via nanoparticle self- 
assembly , Nature , 420 , 395 – 398 . 
61. Chan , W. C. W. , et al. ( 2002 ), Luminescent quantum dots for multiplexed biological 
detection and imaging , Curr. Opin. Biotechnol. , 13 , 40 – 46 . 
62. Qu , L. H. , and Peng , X. G. ( 2002 ), Control of photoluminescence properties of CdSe 
nanocrystals in growth , J. Am. Chem. Soc. , 124 , 2049 – 2055 . 
63. Murphy , C. J. ( 2002 ), Optical sensing with quantum dots , Anal. Chem. , 74 , 520 – 526 . 
64. Parak , W. J. , et al. ( 2003 ), Biological applications of colloidal nanocrystals , Nanotechnology 
, 14 , 15 – 27 . 
65. Gao , X. , et al. ( 2005 ), In vivo molecular and cellular imaging with quantum dots , Curr. 
Opin. Biotechnol. , 16 , 63 – 72 . 
66. Leatherdale , C. A. , et al. ( 2002 ), On the absorption cross section of CdSe nanocrystal 
quantum dots , J. Phys. Chem. B , 106 , 7619 – 7622 . 
67. Gao , X. H. , et al. ( 2004 ), In vivo cancer targeting and imaging with semiconductor 
quantum dots , Nat. Biotechnol. , 22 , 969 – 976 . 
68. Xiao , Y. , et al. ( 2003 ), Plugging into enzymes: Nanowiring of redox enzymes by a gold 
nanoparticle , Science , 299 ( 5614 ), 1877 – 1881 . 
69. Liedberg , B. , et al. ( 1995 ), Biosensing with surface plasmon resonance — How it all 
started , Biosens. Bioelectron. , 10 , 1 – 4 . 
70. Chen , W. P. , and Chen , J. M. ( 1981 ), Use of surface plasma waves for determination 
of the thickness and optical constants of thin metallic fi lms , J. Opt. Soc. Am. , 71 , 189 – 
191 . 
71. Link , S. , and El - Sayed , M. A. ( 1999 ), Spectral properties and relaxation dynamics of 
surface plasmon electronic oscillations in gold and silver nanodots and nanorods , 
J. Phys. Chem. , 103 , 8410 . 
72. Jensen , T. R. , et al. ( 2000 ), Nanosphere lithography: Tunable localized surface plasmon 
resonance spectra of silver nanoparticles , J. Phys. Chem. , 104 , 10549 . 
73. Salmon , Z. , et al. ( 1997 ), Surface plasmon resonance spectroscopy as a tool for investigating 
the biochemical and biophysical properties of membrane protein system. II. 
Application to biological system , Biochim. Biophys. Acta , 1331 , 131 – 152 . 
74. Homola , J. , et al. ( 2001 ), A novel multichannel surface plasmon resonance biosensor , 
Sens. Actuators Chem. , 76 , 403 – 410 . 
75. Oldenburg , S. J. ( 1998 ), Nanoengineering of optical resonances , Chem. Phys. Lett. , 288 , 
243 – 247 . 
76. Sershen , S. , et al. ( 2000 ), Temperature - sensitive polymer - nanoshell composites for photothermally 
modulated drug delivery, J. Biomed. Mater. Res. , 51 ( 03 ), 293 – 298 . 

REFERENCES 1315 
77. Muangsiri , W. , and Kirsch , L. E. ( 2006 ), The protein - binding and drug release properties 
of macromolecular conjugates containing daptomycin and dextran , Int. J. Pharm. , 
315 ( 1 – 2 ), 30 – 43 . 
78. Rothenhausler , B. , and Knoll , W. ( 1988 ), Surface - plasmon microscopy , Nature , 332 , 
615 – 617 . 
79. Piliarik , M. , et al. ( 2005 ), A new surface plasmon resonance sensor for high - throughput 
screening applications , Biosens. Bioelectron. , 20 , 2104 – 2110 . 
80. Averitt , R. D. ( 1997 ), Plasmon resonance shifts of Au - coated Au 2 S nanoshells: Insight 
into multicomponent nanoparticle growth , Phys. Rev. Lett. , 78 , 4217 – 4220 . 
81. McDonnell , J. M. ( 2001 ), Surface plasmon resonance: Towards an understanding of the 
mechanisms of biological molecular recognition , Curr. Opin. Chem. Biol. , 5 , 572 – 577 . 
82. Baek , S. H. , et al. ( 2004 ), Surface plasmon resonance imaging analysis of hexahistidine - 
tagged protein on the gold thin fi lm coated with a calix crown derivative , Biotechnol. 
Bioprocess. Eng. , 9 , 143 – 146 . 
83. Oh , B. K. , et al. ( 2003 ), Immunosensor for detection of Legionella pneumophila using 
surface plasmon resonance , Biosens. Bioelectron. , 18 , 605 – 611 . 
84. Oh , B. K. , et al. ( 2004 ), Surface plasmon resonance immunosensor for the detection of 
Salmonella typhimurium , Biosens. Bioelectron. , 19 , 1497 – 1504 . 
85. Lehn , J. M. ( 1993 ), Supramolecular chemistry , Science , 260 , 1762 – 1763 . 
86. Tanford , C. ( 1974 ), Theory of micelle formation in aqueous solutions , J. Phys. Chem. , 78 , 
24 . 
87. Desai , H. , et al. ( 2006 ), Micellar characteristics of diblock polyacrylate – polyethylene 
oxide copolymers in aqueous media , Eur. Polym. J. , 42 ( 3 ), 593 – 601 . 
88. Yoshii , N. , and Okazaki , S. ( 2006 ), A molecular dynamics study of structural stability of 
spherical SDS micelle as a function of its size , Chem. Phys. Lett. , 425 ( 1 – 3 ), 58 – 61 . 
89. Gochman - Hecht , H. , and Bianco - Peled , H. ( 2006 ), Structure modifi cations of AOT 
reverse micelles due to protein incorporation , J. Coll. Interf. Sci. , 297 ( 1 ), 276 – 283 . 
90. Luisi , P. L. , et al. ( 1998 ), Reverse micelles as hosts for proteins and small molecules , 
Biochim. Biophys. Acta , 947 ( 1 ), 209 – 246 . 
91. Munch , M. R. , and Gast , A. P. ( 1998 ), Block copolymers at interfaces. 1. Micelle formation 
, Macromolecules , 21 , 1360 – 1366 . 
92. Siegel , D. P. ( 1986 ), Inverted micellar intermediates and the transitions between lamellar, 
cubic, and inverted hexagonal lipid phases. II. Implications for membrane - membrane 
interactions and membrane fusion , Biophys. J. , 49 ( 6 ), 1171 – 1183 . 
93. Sokolova , I. , and Kievskya , Y. ( 2005 ), 3D Design of self - assembled nanoporous colloids , 
Stud. Surf. Sci. Catal. , 56 , 433 – 442 . 
94. Zhang , Y. , et al. ( 2004 ), Hollow spheres from shell cross - linked, noncovalently connected 
micelles of carboxyl - terminated polybutadiene and poly(vinyl alcohol) in water , 
Macromolecules , 37 , 1537 – 1543 . 
95. Venema , L. C. , et al. ( 2000 ), Atomic structure of carbon nanotubes from scanning tunneling 
microscopy , Phys. Rev. B , 61 , 2991 – 2996 . 
96. Iijima , S. , and Ichihashi , T. ( 1993 ), Single - shell carbon nanotubes of 1 - nm diameter , 
Nature , 363 , 603 – 605 . 
97. Vinciguerra , V. , et al. ( 2003 ), Growth mechanisms in chemical vapour deposited carbon 
nanotubes , Nanotechnology , 14 ( 6 ), 655 – 660 . 
98. Sonoyama , N. , et al. ( 2006 ), Synthesis of carbon nanotubes on carbon fi bers by means 
of two - step thermochemical vapor deposition , Carbon , 44 , 1754 – 1761 . 
99. Bondi , S. N. , et al. ( 2006 ), Laser assisted chemical vapor deposition synthesis of carbon 
nanotubes and their characterization , Carbon , 44 , 1393 – 1403 . 

1316 PHARMACEUTICAL NANOSYSTEMS 
100. Gavillet , J. , et al. ( 2002 ), Microscopic mechanisms for the catalyst assisted growth of 
single - wall carbon nanotubes , Carbon , 40 ( 10 ), 1649 – 1663 . 
101. Lee , C. J. , et al. ( 2002 ), Large - scale production of aligned carbon nanotubes by the vapor 
phase growth method , Chem. Phys. Lett. , 359 ( 1 – 2 ), 109 – 114 . 
102. Endo , M. , et al. (2005), “Buckypaper” from coaxial nanotubes , Nature , 433 , 476 . 
103. Hata , K. , et al. ( 2004 ), Water - assisted highly effi cient synthesis of impurity - free single - 
walled carbon nanotubes , Science , 306 ( 5700 ), 1362 – 1364 . 
104. Pantarotto , D. , et al. ( 2003 ), Synthesis, structural characterization, and immunological 
properties of carbon nanotubes functionalized with peptides , J. Am. Chem. Soc. , 125 ( 20 ), 
6160 – 6164 . 
105. Kam , N. W. S. , and Dai , H. ( 2005 ), Carbon nanotubes as intracellular protein transporters: 
Generality and biological functionality , J. Am. Chem. Soc. , 127 ( 16 ), 6021 – 6026 . 
106. Kroto , H. W. , et al. (1985), C60: Buckminsterfullerene , Nature , 318 , 162 – 163 . 
107. Goel , A. , et al. ( 2002 ), Combustion synthesis of fullerenes and fullerenic nanostructures , 
Carbon , 40 , 177 – 182 . 
108. Kreatschmer , W. , et al. ( 1990 ), Solid C60: New form of carbon , Nature , 347 , 354 – 358 . 
109. Baierl , T. , et al. ( 1996 ), Comparison of immunological effects of fullerene C60 and raw 
soot from fullerene production on alveolar macrophages and macrophage like cells in 
vitro , Exp. Toxicol. Pathol. , 48 , 508 – 511 . 
110. Satoh , M. , et al. ( 1997 ), Inhibitory effects of a fullerene derivative, dimalonic acid 
C60, on nitric oxide - induced relaxation of rabbit aorta , Eur. J. Pharmacol. , 327 , 175 – 
181 . 
111. Gharbi , N. , et al. ( 2005 ), Fullerene is a powerful antioxidant in vivo with no acute or 
subacute toxicity , NanoLetters , 5 , 2578 – 2585 . 
112. Tabata , Y. , et al. ( 1997 ), Antitumor effect of poly(ethylene glycol) - modifi ed fullerene , 
Fullerene Sci. Technol. , 5 , 989 – 1007 . 
113. Signori , F. , et al. ( 2005 ), New self - assembling biocompatible – biodegradable amphiphilic 
block copolymers , Polymer , 46 ( 23 ), 9642 – 9652 . 
114. Lecommandoux , S. , et al. ( 2006 ), Smart hybrid magnetic self - assembled micelles and 
hollow capsules , Prog. Solid State Chem. , 34 ( 2 – 4 ), 171 – 179 . 
115. McNally , H. , et al. ( 2003 ), Self - assembly of micro - and nano - scale particles using bio - 
inspired events , Appl. Surf. Sci. , 214 ( 1 – 4 ), 109 – 119 . 
116. Bashir , R. ( 2001 ), Invited Review: DNA - mediated artifi cial nanobiostructures: State of 
the art and future directions , Superlatt. Microstruct. , 29 ( 1 ), 1 – 16 . 
117. H a nel , K. , et al. ( 2006 ), Manipulation of organic “ needles ” using an STM operated under 
SEM control , Surf. Sci. , 600 ( 12 ), 2411 – 2416 . 
118. Guthold , M. , et al. ( 1999 ), Investigation and modifi cation of molecular structures with 
the nanoManipulator , J. Mol. Graphics Modell. , 17 ( 3 – 4 ), 187 – 197 . 
119. Rogers , J. A. , and Nuzzo , R. G. ( 2005 ), Recent progress in soft lithography , Mater. Today , 
8 ( 2 ), 50 – 56 . 
120. Choi , D - G. , et al. ( 2004 ), 2D nano/micro hybrid patterning using soft/block copolymer 
lithography , Mater. Sci. Eng. , 24 ( 1 – 2 ), 213 – 216 . 
121. Sellergren , B. , and Allender , C. J. ( 2005 ), Molecularly imprinted polymers: A bridge to 
advanced drug delivery , Adv. Drug Deliv. Rev. , 57 ( 12 ), 1733 – 1741 . 
122. van Nostrum , C. F. ( 2005 ), Molecular imprinting: A new tool for drug innovation , Drug 
Discov. Today Technol. , 2 ( 1 ), 119 – 124 . 
123. Caruso , F. , et al. ( 2000 ), Enzyme encapsulation in layer - by - layer engineered polymer 
multilayer capsules , Langmuir , 16 , 1485 – 1488 . 

REFERENCES 1317 
124. Caruso , F. , et al. ( 1998 ), Nanoengineering of inorganic and hybrid hollow spheres by 
colloidal templating , Science , 282 , 1111 – 1114 . 
125. Deotare , P. B. , and Kameoka , J. ( 2006 ), Fabrication of silica nanocomposite - cups using 
electrospraying , Nanotechnology , 17 , 1380 – 1383 . 
126. Lu , Y. , and Chen , S. C. ( 2004 ), Micro and nano - fabrication of biodegradable polymers 
for drug delivery , Adv. Drug Deliv. Rev. , 56 , 1621 – 1633 . 
127. Lu , Y. , et al. ( 2005 ), Shaping biodegradable polymers as nanostructures: Fabrication and 
applications , Drug Discov. Today Techno. , 2 ( 1 ), 97 – 102 . 
128. Michel , R. , et al. ( 2002 ), A novel approach to produce biologically relevant chemical 
patterns at the nanometer scale: Selective molecular assembly patterning combined with 
colloidal lithography , Langmuir , 18 , 8580 – 8586 . 
129. Hanarp , P. , et al. ( 1999 ), Nanostructured model biomaterial surfaces prepared by colloidal 
lithography , Nanostruct. Mater. , 12 ( 1 ), 429 – 432 . 
130. Di Fabrizio , E. , et al. ( 2004 ), X - ray lithography for micro - and nano - fabrications at 
ELETTRA for interdisciplinary applications , J. Phys. Condens. Matter , 16 , 3517 – 3535 . 
131. Couvreur , P. , et al. ( 2002 ), Nanocapsule technology: A review , Crit. Rev. Ther. Drug 
Carrier Syst. , 19 ( 2 ), 99 – 134 . 
132. Watnasirichaikul , S. , et al. ( 2000 ), Preparation of biodegradable insulin nanocapsules 
from biocompatible microemulsions , Pharm. Res. , 17 ( 6 ), 684 – 689 . 
133. Hitt , J. , et al. ( 2002 ), Nanoparticles of poorly water soluble drugs made via a continuous 
precipitation process, in Proceedings, American Association of Pharmaceutical Scientists , 
annual meeting, Toronto. 
134. Wang , Y. , et al. ( 2004 ), Polymer coating/encapsulation of nanoparticles using a supercritical 
anti - solvent process , J. Supercrit. Fluids , 28 , 85 – 99 . 
135. Perez , C. , et al. ( 2001 ), Poly(lactic acid) - poly(ethylene glycol) nanoparticles as new carriers 
for the delivery of plasmid DNA , J. Controlled Release , 75 ( 1 – 2 ), 211 – 224 . 
136. Dickert , F. L. ( 2003 ), Nano - and micro - structuring of sensor materials — From molecule 
to cell detection , Synthetic Metals , 138 ( 1 – 2 ), 65 – 69 . 
137. Zeng , J. , et al. ( 2003 ), Biodegradable electrospun fi bers for drug delivery , J. Controlled 
Release , 92 ( 3 ), 227 – 231 . 
138. Xu , X. , et al. ( 2005 ), Ultrafi ne medicated fi bers electrospun from W/O emulsions , 
J. Controlled Release , 108 ( 1 ), 33 – 42 . 
139. Nicolau , D. V. , et al. ( 1997 ), Bionanostructures built on e - beam - assisted functionalized 
polymer surfaces , Proc. SPIE , 3241 . 
140. Sinha , P. M. , et al. ( 2004 ), Nanoengineered device for drug delivery application , Nanotechnology 
, 15 , 585 – 589 . 
141. Maloney , J. M. , et al. ( 2005 ), In vivo biostability of CVD silicon oxide and silicon nitride 
fi lms , Mater. Res. Soc. Symp. Proc. , 872 . 
142. Sugai , T. , and Yoshida , H. ( 2003 ), New synthesis of high - quality double - walled carbon 
nanotubes by high - temperature pulsed arc discharge , NanoLetters , 3 ( 6 ), 769 – 773 . 
143. Du , F. , et al. ( 2006 ), The synthesis of single - walled carbon nanotubes with controlled 
length and bundle size using the electric arc method , Carbon , 44 ( 7 ), 1327 – 1330 . 
144. Tamir , S. , and Drezner , Y. ( 2006 ), New aspects on pulsed laser deposition of aligned 
carbon nanotubes , Appl. Surf. Sci. , 252 ( 13 ), 4819 – 4823 . 
145. Aratono , Y. , et al. ( 2005 ), Formation of fullerene(C 60 ) by laser ablation in superfl uid 
helium at 1.5 K , Chem. Phys. Lett. , 408 ( 4 – 6 ), 247 – 251 . 
146. He , C. , et al. ( 2006 ), A practical method for the production of hollow carbon onion particles, 
J. Alloys Compounds , 425 ( 1 – 2 ), 329 – 333 . 

1318 PHARMACEUTICAL NANOSYSTEMS 
147. Liu , T. - C. , et al. ( 2006 ), Synthesis of carbon nanocapsules and carbon nanotubes by an 
acetylene fl ame method , Carbon , 44 ( 10 ), 2045 – 2050 . 
148. Rodriguez-Hernandez, J. , et al. (2005), Toward “smart” nano-objects by self-assembly of 
block copolymers in solution , Prog. Polym. Sci. , 30 , 691 – 724 . 
149. Birac , J. J. ( 2006 ), Geometry based design strategy for DNA nanostructures , J. Mol. 
Graphics Modell. , 25 , 470 – 480 . 
150. Samori , B. , and Zuccheri , G. ( 2004 ), DNA codes for nanoscience , Angew. Chem. Int. Ed. 
Engl. , 44 , 1166 – 1181 . 
151. Seeman , N. C. ( 1998 ), DNA nanotechnology: Novel DNA constructions , Annu. Rev. 
Biophys. Biomol. Struct. , 27 , 225 – 248 . 
152. Seeman, N. C. (1997), DNA components for molecular architecture , Acc. Chem. Res. , 30 , 
347 – 391 . 
152a. Rothemund , P. W. K. ( 2006 ), Folding DNA to create nanoscale shapes and patterns , 
Nature , 440 , 297 – 302 . 
153. Williams , K. A. , et al. ( 2002 ), Nanotechnology: Carbon nanotubes with DNA recognition , 
Nature , 420 , 761 . 
154. Niemeyer , C. M. ( 2004 ), Semi - synthetic DNA – protein conjugates: Novel tools in analytics 
and nanobiotechnology , Biochem. Soc.Trans. , 32 , 51 – 53 . 
155. Asakawa, T. , et al. (2006), Build-to-order nanostructures using DNA self-assembly , Thin 
Solid Films , 509 ( 1 – 2 ), 85 – 93 . 
156. H o gberg , B. , et al. ( 2006 ), Study of DNA coated nanoparticles as possible programmable 
self - assembly building blocks , Appl. Surf. Sci. , 252 ( 15 ), 5538 – 5541 . 
157. Seeman , N. C. ( 1982 ), Nucleic acid junctions and lattices , J. Theor. Biol. , 99 , 237 – 247 . 
158. Chen , J. , and Seeman , N. C. ( 1991 ), Synthesis from DNA of a molecule with the connectivity 
of a cube , Nature , 350 , 631 – 633 . 
159. Li , X. J. , et al. ( 1996 ), Antiparallel DNA double crossover molecules as components for 
nanoconstruction , J. Am. Chem. Soc. , 118 , 6131 – 6140 . 
160. Fu , T. J. , and Seeman , N. C. ( 1993 ), DNA double crossover structures , Biochemistry , 32 , 
3211 – 3220 . 
161. Zhang , Y. , and Seeman , N. C. ( 1992 ), A solid - support methodology for the construction 
of geometrical objects from DNA , J. Am. Chem. Soc. , 114 , 2656 – 2663 . 
162. Niemeyer , C. M. , et al. ( 1999 ), DNA - directed immobilization: Effi cient, reversible and 
site - selective surface binding of proteins by means of covalent DNA - streptavidin conjugates 
, Anal. Biochem. , 268 , 5463 . 
163. Kallenbach , N. R. , et al. ( 1983 ), An immobile nucleic aid junction constructed from oligonucleotides 
, Nature , 305 , 829 – 831 . 
164. Ma , R. I. , et al. ( 1986 ), Three - arm nucleic acid junctions are fl exible , Nucl. Acids Res. , 
14 , 9745 – 9753 . 
165. Brucale , M. , et al. ( 2006 ), Mastering the complexity of DNA nanostructures , Trends 
Biotechnol. , 24 ( 5 ), 235 – 243 . 
166. Nakayama , Y. ( 2002 ), Scanning probe microscopy installed with nanotube probes and 
nanotube tweezers , Ultramicroscopy , 91 ( 1 – 4 ), 49 – 56 . 
167. L u , J. H. ( 2004 ), Nanomanipulation of extended single - DNA molecules on modifi ed 
mica surfaces using the atomic force microscopy , Coll. Surf. B Biointerf. , 39 ( 4 ), 
177 – 180 . 
168. Decossas , S. , et al. ( 2003 ), Nanomanipulation by atomic force microscopy of carbon 
nanotubes on a nanostructured surface , Surf. Sci. , 543 ( 1 – 3 ), 57 – 62 . 
169. Ishii , Y. , et al. ( 2001 ), Single molecule nanomanipulation of biomolecules , Trends Biotechnol. 
, 19 ( 6 ), 211 – 216 . 

REFERENCES 1319 
170. Rogers , J. A. , et al. ( 1997 ), Using an elastomeric phase mask for sub - 100 nm photolithography 
in the optical near fi eld , Appl. Phys. Lett. , 70 , 2658 – 2660 . 
171. Rogers , J. A. , et al. ( 1998 ), Generating similar to 90 nanometer features using near - fi eld 
contact - mode photolithography with an elastomeric phase mask , J. Vac. Sci. Technol. , 16 , 
59 – 68 . 
172. Wulff , G. , et al. ( 1991 ), Racemic resolution of free sugars with macroporous polymers 
prepared by molecular imprinting. Selective dependence on the arrangement of functional 
groups versus spatial requirements , J. Org. Chem. , 56 ( 1 ), 395 – 400 . 
173. Sellergren , B. , et al. ( 1988 ), Highly enantioselective and substrate - selective polymers 
obtained by molecular imprinting utilizing noncovalent interactions — NMR and 
chromatrographic studies on the nature of recognition , J. Am. Chem. Soc. , 110 , 
5853 – 5860 . 
174. Lubke , C. , et al. ( 2000 ), Imprinted polymers prepared with stoichiometric template - 
monomer complexes: Effi cient binding of ampicillin from aqueous solutions , Macromolecules 
, 33 , 5098 – 5105 . 
175. Tskruk , V. V. , et al. ( 1997 ), Self - assembled multilayer fi lms from dendrimers , Langmuir , 
13 ( 8 ), 2171 – 2175 . 
176. Schulze , K. , and Kirstein , S. ( 2005 ), Layer - by - layer deposition of TiO 2 nanoparticles , 
Appl. Surf. Sci. , 246 ( 4 ), 415 – 419 . 
177. Ji , J. , and Shen , J. ( 2005 ), Electrostatic self - assembly and nanomedicine. Engineering in 
medicine and biology society, in IEEE - EMBS 27th Annual International Conference 
Proc ., Shanghai , pp. 720 – 722 . 
178. Beers , A. M. , et al. ( 1983 ), CVD silicon structures formed by amorphous and crystalline 
growth , J. Crystal Growth , 64 ( 3 ), 563 – 557 . 
179. Kyung , S. , et al. ( 2006 ), Deposition of carbon nanotubes by capillary - type atmospheric 
pressure PECVD , Thin Solid Films , 506 – 507 , 268 – 273 . 
180. Goodman, C. , and Pessa, M. (1986), Atomic layer epitaxy , J. Appl. Phys. , 60 ( 3 ), 65 – 81 . 
181. Suntola, T. (1992), Atomic layer epitaxy , Thin Solid Films , 216 ( 1 ), 84 – 89 . 
182. Xia , C. , et al. ( 1998 ), Metal - organic chemical vapor deposition of Sr - Co - Fe - O fi lms on 
porous substrates , J. Mater. Res. , 13 ( 1 ), 173 . 
183. Choy , K. L. ( 2003 ), Chemical vapour deposition of coatings , Prog. Mater. Sci. , 48 ( 2 ), 
57 – 170 . 
184. Kaul , A. R. , and Seleznev , B. V. ( 1993 ), New principle of feeding for fl ash evaporation 
MOCVD devices , J. Phys. , 3 , 375 – 378 . 
185. Douard , A. , and Maury , F. ( 2006 ), Nanocrystalline chromium - based coatings deposited 
by DLI - MOCVD under atmospheric pressure from Cr(CO)6 , Surf. Coat. Technol. , 200 , 
6267 – 6271 . 
186. Lau , K. K. S. ( 2001 ), Hot - wire chemical vapor deposition (HWCVD) of fl uorocarbon 
and organosilicon thin fi lms , Thin Solid Films , 395 ( 1 – 2 ), 288 – 291 . 
187. Ren , Z. F. , et al. ( 1998 ), Synthesis of large arrays of well - aligned carbon nanotubes on 
glass , Science , 282 ( 5391 ), 1105 – 1107 . 
188. Han , B. W. , et al. ( 2001 ), Growth of in situ CoSi 2 layer by metalorganic chemical vapor 
deposition on Si tips and its fi eld - emission properties , J. Vac. Sci. Technol. Microelectron. 
Nanometer Struct. , 19 ( 2 ), 533 – 536 . 
189. Kuo , T. - F. , et al. ( 2001 ), Microwave - assisted chemical vapor deposition process for synthesizing 
carbon nanotubes , J. Vac. Sci. Technol. Microelectron. Nanometer Struct. , 19 ( 3 ), 
1030 – 1033 . 
190. McSporran , N. , et al. ( 2002 ), Preliminary studies of atmospheric pressure plasma 
enhanced CVD (AP - PECVD) of thin oxide fi lms , Phys. IV France , 12 , 4 – 17 . 

1320 PHARMACEUTICAL NANOSYSTEMS 
191. Chang , J. P. , et al. ( 2001 ), Rapid thermal chemical vapor deposition of zirconium oxide 
for metal - oxide - semiconductor fi eld effect transistor application , J. Vac. Sci. Technol. B 
Microelectron. Nanometer Struct. , 19 ( 5 ), 1782 – 1787 . 
192. Huang , Z. P. , et al. ( 1998 ), Growth of highly oriented carbon nanotubes by plasma - 
enhanced hot fi lament chemical vapor deposition , Appl. Phys. Lett. , 73 ( 26 ), 3845 – 3847 . 
193. Huczko , A. ( 2002 ), Synthesis of aligned carbon nanotubes , Appl. Phys. A Mater. Sci. 
Process. , 74 ( 5 ), 617 – 638 . 
194. Lim , C. - Y. , et al. ( 2004 ), Development of an electrodeposited nanomold from compositionally 
modulated alloys , J. Appl. Electrochem. , 34 , 857 – 866 . 
195. Micheletto , R. , et al. ( 1995 ), A simple method for the production of a two - dimensional, 
ordered array of small latex particles , Langmuir , 11 , 3333 – 3336 . 
196. Boneberg , J. , et al. ( 1997 ), The formation of nano - dot and nano - ring structures in colloidal 
monolayer lithography , Langmuir , 13 , 7080 – 7084 . 
197. Blanco , A. , et al. ( 2000 ), Large - scale synthesis of a silicon photonic crystal with a complete 
three - dimensional bandgap near 1.5 micrometres , Nature , 405 , 437 – 440 . 
198. Denis , F. A. , et al. ( 2004 ), Nanoscale chemical patterns fabricated by using colloidal 
lithography and self - assembled monolayers , Langmuir , 20 , 9335 – 9339 . 
199. Cadotte , J. E. ( 1981 ), U.S. Patent 4,277,344 . Interfacially synthesized reverse osmosis 
membrane . 
200. Krauel , K. , et al. ( 2005 ), Using different structure types of microemulsions for the preparation 
of poly(alkylcyanoacrylate) nanoparticles by interfacial polymerization , J. 
Controlled Release , 106 ( 1 – 2 ), 76 – 87 . 
201. Govender , T. , et al. ( 1999 ), PLGA nanoparticles prepared by nanoprecipitation: Drug 
loading and release studies of a water soluble drug , J. Controlled Release , 57 ( 2 ), 
171 – 185 . 
202. Chorny , M. , et al. ( 2002 ), Lipophilic drug loaded nanospheres prepared by nanoprecipitation: 
Effect of formulation variables on size, drug recovery and release kinetics , 
J. Controlled Release , 83 ( 3 ), 389 – 400 . 
203. Desgouilles , S. , et al. ( 2003 ), The design of nanoparticles obtained by solvent evaporation: 
A comprehensive study , Langmuir , 19 ( 22 ), 9504 – 9510 . 
204. Zambaux , M. F. ( 1998 ), Infl uence of experimental parameters on the characteristics of 
poly(lactic acid) nanoparticles prepared by a double emulsion method , J. Controlled 
Release , 50 ( 1 – 3 ), 31 – 40 . 
205. Cao , H. , et al. ( 2002 ), Fabrication of 10 nm enclosed nanofl uidic channels , Appl. Phys. 
Lett. , 81 ( 1 ), 174 – 176 . 
206. Scheer , H. - C. , et al. ( 1998 ), Problems of the nanoimprinting technique for nanometer 
scale pattern defi nition , J. Vac. Sci. Technol. B Microelectron. Nanometer Struct. , 16 ( 6 ), 
3917 – 3921 . 
207. Kim , J - S. , and Reneker , D. H. ( 1999 ), Polybenzimidazole nanofi ber produced by electrospinning 
, Polym. Eng. Sci. , 39 ( 5 ), 849 – 854 . 
208. Abidian , M. R. , et al. ( 2006 ), Conducting - polymer nanotubes for controlled drug release , 
Adv. Mater. , 18 , 405 – 409 . 
209. Arias , J. L. , et al. ( 2006 ), Preparation and characterization of carbonyl iron/ 
poly(butylcyanoacrylate) core/shell nanoparticles , J. Collo. Interf. Sci. , 299 ( 2 ), 599 – 607 . 
210. Cui , F. , et al. ( 2006 ), Preparation and characterization of mucoadhesive polymer - coated 
nanoparticles , Int. J. Pharm. , 316 ( 1 – 2 ), 154 – 161 . 
211. Huang , C. - Y. , and Lee , Y. - D. ( 2006 ), Core - shell type of nanoparticles composed of 
poly[(n - butyl cyanoacrylate) - co - (2 - octyl cyanoacrylate)] copolymers for drug delivery 

REFERENCES 1321 
application: Synthesis, characterization and in - vitro degradation, Int. J. Pharm. , 325 ( 1 – 2 ), 
132 – 139 . 
212. Hirsj a rvi , S. , et al. ( 2006 ), Layer - by - layer polyelectrolyte coating of low molecular weight 
poly(lactic acid) nanoparticles , Coll. Surf. B Biointerf. , 49 ( 1 ), 93 – 99 . 
213. Schubert , M. A. , and M u ller - Goymann , C. C. ( 2005 ), Characterisation of surface - 
modifi ed solid lipid nanoparticles (SLN): Infl uence of lecithin and nonionic emulsifi er , 
Eur. J. Pharm. Biopharm. , 61 ( 1 – 2 ), 77 – 86 . 
214. Lo , C. - L. ( 2005 ), Preparation and characterization of intelligent core - shell nanoparticles 
based on poly( d , l - lactide) - g - poly( N - isopropyl acrylamide -co - methacrylic acid) , J. Controlled 
Release , 104 ( 3 ), 477 – 488 . 
215. Garcia - Fuentes , M. ( 2005 ), Design and characterization of a new drug nanocarrier made 
from solid – liquid lipid mixtures , J. Coll. Interf. Sci. , 285 ( 2 ), 590 – 598 . 
216. Lochmann , D. , et al. ( 2005 ), Albumin – protamine – oligonucleotide nanoparticles as a new 
antisense delivery system. Part 1: Physicochemical characterization , Eur. J. Pharm. Biopharm. 
, 59 ( 3 ), 419 – 429 . 
217. M u ller - Goymann , C. C. ( 2004 ), Physicochemical characterization of colloidal drug delivery 
systems such as reverse micelles, vesicles, liquid crystals and nanoparticles for topical 
administration , Eur. J. Pharm. Biopharm. , 58 ( 2 ), 343 – 356 . 
218. Weyermann , J. , et al. ( 2004 ), Physicochemical characterisation of cationic polybutylcyanoacrylat 
— Nanoparticles by fl uorescence correlation spectroscopy , Eur. J. Pharm. 
Biopharm. , 58 ( 1 ), 25 – 35 . 
219. Bootz , A. , et al. ( 2004 ), Comparison of scanning electron microscopy, dynamic light 
scattering and analytical ultracentrifugation for the sizing of poly(butyl cyanoacrylate) 
nanoparticles , Eur. J. Pharm. Biopharm. , 57 ( 2 ), 369 – 375 . 
220. Merroun , M. , et al. ( 2007 ), Spectroscopic characterization of gold nanoparticles formed 
by cells and S - layer protein of Bacillus sphaericus JG - A12, Mater. Sci. Eng. , 27 ( 1 ), 
188 – 192 . 
221. Lu , Q. H. , et al. ( 2006 ), Synthesis and characterization of composite nanoparticles 
comprised of gold shell and magnetic core/cores , J. Magnet. Magnetic Mater. , 301 ( 1 ), 
44 – 49 . 
222. Vdovenkova , T. , et al. ( 2000 ), Silicon nanoparticles characterization by Auger electron 
spectroscopy , Surf. Sci. , 454 – 456 , 952 – 956 . 
223. Bonini , M. , et al. ( 2006 ), Synthesis and characterization of magnetic nanoparticles coated 
with a uniform silica shell , Mater. Sci. Eng. , 26 ( 5 – 7 ), 745 – 750 . 
224. Pilon , L. N. , et al. ( 2006 ), Synthesis and characterisation of new shell cross - linked micelles 
with amine - functional coronas , Eur. Polym. J. , 42 ( 7 ), 1487 – 1498 . 
225. Castro , E. , et al. ( 2006 ), Characterization of triblock copolymer E 67 S 15 E 67 — Surfactant 
interactions , Chem. Phys. , 325 ( 2 – 3 ), 492 – 498 . 
226. Tao , L. , and Uhrich , K. E. ( 2006 ), Novel amphiphilic macromolecules and their in vitro 
characterization as stabilized micellar drug delivery systems , J. Coll. Interf. Sci. , 298 ( 1 ), 
102 – 110 . 
227. Gaucher , G. , et al. ( 2005 ), Block copolymer micelles: Preparation, characterization and 
application in drug delivery , J. Controlled Release , 109 ( 1 – 3 ), 169 – 188 . 
228. Yu , L. , et al. ( 2006 ), Determination of critical micelle concentrations and aggregation 
numbers by fl uorescence correlation spectroscopy: Aggregation of a lipopolysaccharide , 
Anal. Chim. Acta , 556 ( 1 ), 216 – 225 . 
229. Kataoka , K. , et al. ( 2001 ), Block copolymer micelles for drug delivery: Design, characterization 
and biological signifi cance , Adv. Drug Deliv. Rev. , 47 ( 1 ), 113 – 131 . 

1322 PHARMACEUTICAL NANOSYSTEMS 
230. Hagan , S. A. , et al. ( 1996 ), Polylactide - poly(ethylene glycol) copolymers as drug delivery 
systems. 1. characterization of water dispersible micelle - forming systems , Langmuir , 
12 ( 9 ), 2153 – 2161 . 
231. Shin , I. G. , et al. ( 1998 ), Methoxy poly(ethylene glycol)/epsilon - caprolactone amphiphilic 
block copolymeric micelle containing indomethacin. I. Preparation and characterization , 
J. Controlled Release , 51 ( 1 ), 1 – 11 . 
232. Morishima , Y. , et al. ( 1995 ), Characterization of unimolecular micelles of random 
copolymers of sodium 2 - (acrylamido) - 2 - methylpropanesulfonate and methacrylamides 
bearing bulky hydrophobic substituents , Macromolecules , 28 , 2874 – 2881 . 
233. Vansteenkiste , S. O. , and Davies , M. C. ( 1998 ), Scanning probe microscopy of biomedical 
interfaces , Prog. Surf. Sci. , 57 ( 2 ), 95 – 136 . 
234. Binnig , G. , and Rohrer , H. ( 2000 ), Scanning tunneling microscopy , IBM J. Res. Dev. , 
44 ( 1 – 2 ), 279 . 
235. Chen , C. J. , and Smith , W. F. ( 1994 ), Introduction to scanning tunneling microscopy , 
Am. J. Phys. , 62 ( 6 ), 573 – 574 . 
236. Giessibl, F. J. (2005), AFM’ s path to atomic resolution, Mater. Today , 8 ( 5 ), 32 – 41 . 
237. Durig , U. , et al. ( 1986 ), Near - fi eld optical - scanning microscopy , J. Appl. Phys. , 59 ( 10 ), 
3318 – 3327 . 
238. Ash , E. A. , and Nichols , G. ( 1972 ), Super - resolution aperture scanning microscope , 
Nature , 237 , 510 . 
239. Merritt , G. ( 1998 ), A compact near - fi eld scanning optical microscope , Ultramicroscopy , 
71 ( 1 – 4 ), 183 – 189 . 
240. Albrecht , T. R. , et al. ( 1991 ), Frequency modulation detection using high -Q cantilevers 
for enhanced force microscope sensitivity , J. Appl. Phys. , 69 , 668 – 673 . 
241. Ohta , M. , et al. ( 1995 ), Atomically resolved image of cleaved surfaces of compound 
semiconductors observed with an ultrahigh vacuum atomic force microscope , J. Vac. Sci. 
Technol. , 13 ( 3 ), 1265 – 1267 . 
242. Binnig , G. , et al. (1986), Atomic force microscope , Phys. Rev. Lett. , 56 ( 9 ), 930 – 933 . 
243. Zhong , Q. , et al. ( 1993 ), Fractured polymer/silica fi ber surface studied by tapping mode 
atomic force microscopy , Surf. Sci. , 290 ( 1 – 2 ), 688 – 692 . 
244. Stark , R. W. , and Drobek , T. ( 1998 ), Determination of elastic properties of single aerogel 
powder particles with the AFM , Ultramicroscopy , 75 ( 3 ), 161 – 169 . 
245. M u ller , F. , et al. ( 1997 ), Applications of scanning electrical force microscopy , Microelectron. 
Reliabil. , 37 ( 10 – 11 ), 1631 – 1634 . 
246. Cappella , B. , et al. ( 1997 ), Improvements in AFM imaging of the spatial variation of 
force – distance curves: On - line images , Nanotechnology , 8 , 82 – 87 . 
247. Hamers , R. J. ( 1989 ), Atomic - resolution surface spectroscopy with the scanning tunneling 
microscope , Annu. Rev. Phys. Chem. , 40 , 531 – 559 . 
248. Laiho , R. , et al. ( 1997 ), Spin - polarized scanning tunnelling microscopy with detection of 
polarized luminescence emerging from a semiconductor tip , J. Phys. Condens. Matter , 9 , 
5697 – 5707 . 
249. Binnig , G. , et al. ( 1982 ), Surface studies by scanning tunneling microscopy , Phys. Rev. 
Lett. , 49 , 57 – 61 . 
250. Ciraci , S. , and Tekman , E. ( 1989 ), Theory of transition from the tunneling regime to point 
contact in scanning tunneling microscopy , Phys. Rev. , 40 , 11969 – 11972 . 
251. Doyen , G. , et al. ( 1993 ), Green - function theory of scanning tunneling microscopy: Tunnel 
current and current density for clean metal surfaces , Phys. Rev. , 47 , 9778 – 9790 . 

252. Fontaine , P. A. , et al. ( 1998 ), Characterization of scanning tunneling microscopy and 
atomic force microscopy - based techniques for nanolithography on hydrogen - passivated 
silicon , J. Appl. Phys. , 84 ( 1 ), 1776 – 1781 . 
253. Wiesendanger , R. ( 1994 ), Contributions of scanning probe microscopy and spectroscopy 
to the investigation and fabrication of nanometer - scale structures , J. Vac. Sci. Technol. 
Microelectron. Nanometer Struct. , 12 ( 2 ), 515 – 529 . 
254. Binnig , G. , et al. (1986), Atomic force microscopy , Phys. Rev. Lett. , 56 , 930 – 933 . 
255. de Souza Pereira , R. ( 2001 ), Atomic force microscopy as a novel pharmacological tool , 
Biochem. Pharmacol. , 62 ( 8 ), 975 – 983 . 
256. Rajagopalan , R. ( 2000 ), Atomic force and optical force microscopy: Applications to 
interfacial microhydrodynamics , Coll. Surf. Physicochem. Eng. Aspects , 174 ( 1 – 2 ), 253 – 
267 . 
257. Santos , N. C. , and Castanho , M. A. R. B. ( 2004 ), An overview of the biophysical applications 
of atomic force microscopy , Biophys. Chem. , 107 ( 2 ), 133 – 149 . 
258. Ferin , J. , et al. ( 1992 ), Pulmonary retention of ultrafi ne and fi ne particles in rats , Am. J. 
Respir. Cell Mol. Biol. , 6 , 535 – 542 . 
259. Li , X. Y. , et al. ( 1999 ), Short term infl ammatory responses following intratracheal 
instillation of fi ne and ultrafi ne carbon black in rats , Inhal. Toxicol. , 11 , 709 – 
731 . 
260. Oberdorster , G. , et al. ( 1995 ), Association of particulate air pollution and acute mortality: 
Involvement of ultrafi ne particles? Inhal. Toxicol. , 7 , 111 – 124 . 
261. Li , X. Y. , et al. ( 1997 ), In vivo and in vitro pro - infl ammatory effects of particulate air 
pollution (PM10) , Environ. Health Perspect. , 105 , 1279 – 1283 . 
262. Br u ske - Hohlfeld , I. , et al. ( 2004 ), Epidemiology of nanoparticles, in Proc. of First International 
Symposium on Occupational Health Implications of Nanomaterials . 
263. Warheit , D. B. , et al. ( 2004 ), Pulmonary bioassay toxicity study in rats with single wall 
carbon nanotubes, in Proc. of First International Symposium on Occupational Health 
Implications of Nanomaterials . 
264. Klumpp , C. , et al. (2006), Functionalized carbon nanotubes as emerging nanovectors for 
the delivery of therapeutics , Biochim. Biophys. Acta , 1758 , 404 – 412 . 
265. Muller , J. ( 2004 ), Respiratory toxicity of carbon nanotubes, in Proc. of First International 
Symposium on Occupational Health Implications of Nanomaterials . 
266. Tinkle , S. ( 2004 ), Dermal penetration of nanoparticles, in Proc. of First International 
Symposium on Occupational Health Implications of Nanomaterials . 
SUGGESTED READING 
General 
Dai , L. ( 2004 ), Intelligent Macromolecules for Smart Devices: From Materials Synthesis to 
Device Applications , Springer . 
Di Ventra , M. , Evoy , S. , and Hefl in , J. R. ( 2004 ), Introduction to Nanoscale Science and Technology 
, Springer . 
Lyshevski , S. E. ( 2005 ), Nano - and Mirco - Electromechnical Systems: Fundamentals of Nano 
and Microengineering , CRC Press , Boca Raton, FL . 
Roco , M. C. , and Bainbridge , W. S. ( 2003 ), Converging Technologies for Improving Human 
Performance: Nanotechnology, Biotechnology , Springer . 
SUGGESTED READING 1323

1324 PHARMACEUTICAL NANOSYSTEMS 
Nano – Pharmaceutical Materials and Structures 
Hirsch , A. , and Brettreich , M. ( 2004 ), Fullerenes: Chemistry and Reactions , Wiley , 
Hoboken, NJ . 
Cao , G. ( 2004 ), Nanostructures & Nanomaterials: Synthesis, Properties & Applications , 
Imperial College Press . 
Haley , M. M. , and Tykwinski , R. R. ( 2006 ), Carbon - Rich Compounds: From Molecules to 
Materials , Wiley , Hoboken, NJ . 
Harris , P. J. ( 2001 ), Carbon Nanotubes and Related Structures , Cambridge University Press . 
Kumar , C. S. S. R. ( 2005 ), Biofunctionalization of Nanomaterials , Wiley , Hoboken, NJ . 
Kumar , C. S. S. R. ( 2006a ), Nanomaterials for Cancer Therapy , Wiley , Hoboken, NJ . 
Kumar , C. S. S. R. ( 2006b ), Biological and Pharmaceutical Nanomaterials , Wiley , Hoboken, 
NJ . 
Lazzari , M. , Liu , G. , and Lecommandoux , S. ( 2006 ), Block Copolymers in Nanoscience , Wiley , 
Hoboken, NJ . 
Schmid , G. ( 2004 ), Nanoparticles: From Theory to Application , Wiley , Hoboken, NJ . 
Tadros , T. F. ( 2005 ), Applied Surfactants: Principles and Applications , Wiley , Hoboken, NJ . 
Vogtle , F. ( 2000 ), Dendrimers III: Design, Dimension, Function , Springer . 
Zhang , J. , Zhang , J. , Wang , Z. , Liu , J. , and Chen , S. ( 2002 ), Self - Assembled Nanostructures , 
Springer . 
Manufacturing Technologies 
Butt , H. - J. , Graf , K. , and Kappl , M. ( 2006 ), Physics and Chemistry of Interfaces , Wiley , 
Hoboken, NJ . 
Champion , Y. , and Fecht , H. ( 2004 ), Nano - Architectured and Nanostructured Materials: 
Fabrication, Control and Properties , Wiley , Hoboken, NJ . 
Elimelech , M. , Jia , X. , Gregory , J. , and Williams , R. ( 1998 ), Particle Deposition & Aggregation: 
Measurement, Modelling and Simulation , Elsevier , New York . 
Hoch , H. C. , Jelinski , L. W. , and Craighead , H. G. ( 1996 ), Nanofabrication and Biosystems , 
Cambridge University Press . 
K o hler , M. , and Fritzsche , W. ( 2004 ), Nanotechnology: An Introduction to Nanostructuring 
Techniques , Wiley , Hoboken, NJ . 
Komiyama , M. , Takeuchi , T. , Mukawa , T. , and Asanuma , H. ( 2003 ), Molecular Imprinting: 
From Fundamentals to Applications , Wiley , Hoboken, NJ . 
Kroschwitz , J. I. ( 1989 ), Polymers: Biomaterials and Medical Applications , Wiley , New York . 
Kumar , C. S. S. R. , Hormes , J. , and Leuschner , C. ( 2005 ), Nanofabrication Towards Biomedical 
Applications: Techniques, Tools, Applications, and Impact , Wiley , Hoboken, NJ . 
Osada , Y. , and Nakagawa , T. ( 1992 ), Membrane Science and Technology , Marcel Dekker , 
New York . 
Pierson , H. ( 1999 ), Handbook of Chemical Vapor Deposition: Principles, Technologies and 
Applications , William Andrew . 
Ramakrishna , S. , Fujihara , K. , Teo , W. , Lim , T. , and Ma , Z. ( 2005 ), An Introduction to Electrospinning 
and Nanofi bers , World Scientifi c . 
Rotello , V. M. ( 2004 ), Nanoparticles: Building Blocks for Nanotechnology , Springer . 
Sotomayor Torres , C. M. ( 2004 ), Alternate Lithography: Unleashing the Potentials of Nanotechnology 
, Springer . 
Wise , D. L. ( 2000 ), Handbook of Pharmaceutical Controlled Release Technology , Marcel 
Dekker , New York . 

Characterization Methods 
Binks , B. P. ( 1999 ), Modern Characterization Methods of Surfactant Systems , Marcel Dekker , 
New York . 
Brittain , H. G. ( 1995 ), Physical Characterization of Pharmaceutical Solids , Marcel Dekker , 
New York . 
Brown , W. , Ed. ( 1996 ), Light Scattering: Principles and Development , Oxford University 
Press . 
Buyana , T. ( 1997 ), Molecular Physics , World Scientifi c. 
Chung , F. H. , and Smith , D. K. , Eds. ( 1999 ), Industrial Applications of X - Ray Diffraction , 
Marcel Dekker , New York . 
Jena , P. B. , and Hoerber , J. K. H. , Eds. ( 2006 ), Force Microscopy: Applications in Biology and 
Medicine , Wiley , Hoboken, NJ . 
Jenkins , R. , and Jenkins , J. ( 1995 ), Quantitative X - Ray Spectrometry , Marcel Dekker , 
New York . 
Meyer , E. , Hug , H. J. , and Bennewitz , R. ( 2003 ), Scanning Probe Microscopy , Springer . 
Pethrick , R. A. , and Viney , C. , Eds. ( 2003 ), Techniques for Polymer Organisation and Morphology 
Characterisation , Wiley , Hoboken, NJ . 
Rosoff , M. ( 2002 ), Nano - Surface Chemistry , Marcel Dekker , New York . 
Sharma , A. , and Schulman , S. G. ( 1999 ), Introduction to Fluorescence Spectroscopy , Wiley , 
New York . 
Sibilia , J. P. ( 1988 ), A Guide to Materials Characterization and Chemical Analysis , Wiley , 
New York . 
Watts , J. F. , and Wolstenholme , J. ( 2004 ), An Introduction to Surface Analysis by XPS and 
AES , Wiley , Hoboken, NJ . 
Wyckoff , R. W. G. (1971), Crystal Structures , Wiley , New York . 
Toxicology 
Fan , A. M. ( 1996 ), Toxicology and Risk Assessment: Principles, Methods & Applications , 
Marcel Dekker , New York . 
Gad , S. C. ( 2004 ), Safety Pharmacology in Pharmaceutical Development and Approval , CRC 
Press , Boca Raton, FL . 
Gardner , D. E. ( 2005 ), Toxicology of the Lung , CRC Press , Boca Raton, FL . 
Kumar , C. S. S. R. ( 2006 ), Nanomaterials: Toxicity, Health and Environmental Issues , Wiley , 
Hoboken, NJ . 
Meeks , R. G. ( 1991 ), Dermal and Ocular Toxicology: Fundamentals and Methods , CRC Press , 
Boca Raton, FL . 
SUGGESTED READING 1325


1327 
7.4 
OIL - IN - WATER NANOSIZED 
EMULSIONS: MEDICAL APPLICATIONS 
Shunmugaperumal Tamilvanan * 
University of Antwerp, Antwerp, Belgium 
Contents 
7.4.1 Introduction 
7.4.2 Generations of Oil - in - Water Nanosized Emulsions 
7.4.2.1 First - Generation Emulsion 
7.4.2.2 Second - Generation Emulsion 
7.4.2.3 Third - Generation Emulsion 
7.4.2.4 Unique Property of Third - Generation Emulsion 
7.4.3 Preparation Methods for Drug - Free/Loaded Oil - in - Water Nanosized Emulsions 
7.4.4 Excipient Inclusion: Oil - in - Water Nanosized Emulsions 
7.4.5 Medical Applications of Oil - in - Water Nanosized Emulsions 
7.4.5.1 Parenteral Routes 
7.4.5.2 Ocular Routes 
7.4.5.3 Nasal Route 
7.4.5.4 Topical Route 
7.4.6 Future Perspective 
References 
7.4.1 INTRODUCTION 
It is estimated that 40% or more of bioactive substances being identifi ed through 
combinatorial screening programs are poorly soluble in water [1, 2] . Consequently, 
the drug molecules belonging to these categories cannot be easily incorporated into 
aqueous - cored/based dosage forms at adequate concentrations, and thus the clinical 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc. 
* Current address: Department of Pharmaceutics, Arulmigu Kalasalingam College of Pharmacy, Anand 
Nagar, Krishnankoil, India

1328 OIL-IN-WATER NANOSIZED EMULSIONS 
effi cacy of highly lipophilic drugs is being impeded. Furthermore, it is well established 
that the pharmaceutical industry will face more diffi culties in formulating and 
developing novel dosage forms of new chemical entities since 50 – 60% of these 
molecules are lipophilic in nature and often exhibit hydrophobic character. Among 
the different innovative formulation approaches that have been suggested for 
enhancing lipophilic drug absorption and then clinical effi cacy, lipid - based colloidal 
drug delivery systems such as nanosized emulsions recognized particularly for overcoming 
the formulation and bioavailability - related problems of such drug molecules. 
Other nomenclatures are also being utilized often in the medical literature to refer 
to nanosized emulsions, including miniemulsions [3] , ultrafi ne emulsions [4] , and 
submicrometer emulsions [5, 6] . The term nanosized emulsion [7] is preferred 
because in addition to giving an idea of the nanoscale size range of the dispersed 
droplets, it is concise and avoids misinterpretation with the term microemulsion 
(which refers to thermodynamically stable systems). Hence, nanosized emulsions 
are heterogenous dispersions of two immiscible liquids [oil - in - water (o/w) or water - 
in - oil (w/o)], and they are subjected to various instability processes such as aggregation, 
fl occulation, coalescence, and therefore eventual phase separation according 
to the second law of thermodynamics. However, the physical stability of nanosized 
emulsions can substantially be improved with the help of suitable emulsifi ers that 
are capable of forming a mono - or multilayer coating around the dispersed liquid 
droplets in such a way as to reduce interfacial tension or increase droplet – droplet 
repulsion. Depending on the concentrations of these three components (oil – water – 
emulsifi er) and the effi ciency of the emulsifi cation equipment/techniques used to 
reduce droplet size, the fi nal nanosized emulsion may be in the form of o/w, w/o, 
macroemulsion, micrometer emulsion, submicrometer emulsion, and double or multiple 
emulsions (o/w/o and w/o/w). Preparation know - how, potential application, and 
other information pertinent to w/o emulsions [8] , macroemulsions [9 – 11] , microemulsions 
[12, 13] , and multiple emulsions [14] are thoroughly covered elsewhere. 
In addition, some studies have compared the performance of different emulsifi ed 
systems (macroemulsions, microemulsion, multiple emulsions, and gel emulsions) 
prepared with similar oils and surfactants for applications such as controlled drug 
release [15] or drug protection [16] . Similarly the state of the art of so - called oxygen 
carriers or perfl uorocarbon emulsions, dispersions containing submicrometer/nanosized 
fl uoroorganic particles in water, is also thoroughly covered in the literature 
[17 – 19] and readers can refer to these complete and interesting articles. 
Possible usefulness as carriers stems from the nanosized emulsion ’ s ability to 
solubilize substantial amounts of hydrophilic/hydrophobic drug either at the innermost 
(oil or water) phase or at the o/w or w/o interfaces. While hydrophilic drugs 
are contained in the aqueous phase of a w/o - type emulsion or at the w/o interface 
of the system, hydrophobic drugs could be incorporated within the inner oil phase 
of an o/w - type emulsion or at the o/w interface of the system. It appears that the 
choice of the type of emulsion to be used therefore depends, to a large extent, upon 
the physicochemical properties of the drug. Between w/o and o/w types, the o/w 
type of nanosized emulsions would be preferred in order to successfully exploit the 
advantages of an emulsion carrier system. Additionally, within the o/w type, simple 
modifi cations on surface/interface structures of emulsions can be made. For instance, 
incorporating an emulsifi er molecule alone or in a specifi c combination that is 
capable of producing either positive or negative charges over the emulsifi ed droplets 
surface will lead to the formation of surface - modifi ed emulsions. Based on these 

GENERATIONS OF OIL-IN-WATER NANOSIZED EMULSIONS 1329 
surface modifi cations, the o/w - type nanosized emulsions can be divided into cationic 
and anionic emulsions. 
The o/w nanosized emulsions have many appealing properties as drug carriers. 
They are biocompatible, biodegradable, physically stable, and relatively easy to 
produce on a large scale using proven technology [20] . Due to their subcellular and 
submicrometer size, emulsions are expected to penetrate deep into tissues through 
fi ne capillaries and even cross the fenestration present in the epithelial lining in 
liver. This allows effi cient delivery of therapeutic agents to target sites in the body. 
Not only considered as delivery carriers for lipophilic and hydrophobic drugs, nanosized 
emulsions can also be viewed nowadays as adjuvants to enhance the potency 
of deoxyribonucleic acid (DNA) vaccine. For instance, Ott et al. [21] prepared a 
cationic o/w emulsion based on MF59 (commercially termed Fluad), a potent squalene 
in water and a cationic lipid, 1,2 - dioleoyl - sn - glycero - 3 - trimethylammonium 
propane (DOTAP). It is shown that an interaction of cationic emulsion droplets 
with DNA and the formed DNA - adsorbed emulsion had a higher antibody response 
in mice in vivo while maintaining the cellular responses equivalent to that seen with 
naked DNA at the same doses. Another example of o/w emulsion - based adjuvants 
resulting from U.S. patent literature is the Ribi adjuvant system (RAS) [22 – 24] . 
Depending on the animal species used, RAS can be classifi ed into two types: one 
for use in mice, termed monophosphoryl - lipid A + trehalose dicorynomycolate 
emulsion (MPL + TDM emulsion), and another for use in rabbits, goats, and larger 
animals, called monophosphoryl - lipid A + trehalose dicorynomycolate + cell wall 
skeleton emulsion (MPL + TDM + CWS emulsion). Strikingly, the MPL + TDM 
and MPL + TDM + CWS emulsions are prepared based on 2% oil (squalene) – 
Tween 80 – water. These adjuvants are derived from bacterial and mycobacterial cell 
wall components that have been prepared to reduce the undesirable side effects of 
toxicity and allergenicity but still provide potent stimulus to the immune system. 
Another example is the syntex adjuvant formulation (SAF) that contains a preformed 
o/w emulsion stabilized by Tween 80 and Pluronic L121 [25] . 
Keeping in mind the potential of o/w nanosized emulsions, the purpose of this 
chapter is to classify the emulsions and provide a short overview on the preparation 
of the new - (second - and third - ) generation emulsions followed by a description of 
the unique property of the third - generation emulsion and examples of selected 
excipients used for emulsion preparation. Given a specifi c interest especially on the 
parenteral route and then on the ocular topical route, o/w nanosized emulsions for 
both routes share a common platform on strict criteria concerning the maximum 
globule size and requirement of sterility in the fi nal emulsions. Nasal and topical 
routes are also covered based on published research works with nanosized emulsions. 
It is emphasized that the chapter focuses only on preformed nanosized emulsions 
(having size distribution ranging between 50 and 1000 nm with a mean droplet 
size of about 250 nm), which should not be confused with self - microemulsifying drug 
delivery systems or preformed microemulsions that are transparent, thermodynamically 
stable dosage forms. 
7.4.2 GENERATIONS OF OIL - IN - WATER NANOSIZED EMULSIONS 
In this chapter, the o/w nanosized emulsions are classifi ed into three generations 
(see Figure 1 ) based on their development to ultimately make a better colloidal 

1330 OIL-IN-WATER NANOSIZED EMULSIONS 
carrier for a target site within the organs/parts of the body and eye, thus allowing 
site - specifi c drug delivery and/or enhanced drug absorption. 
7.4.2.1 First - Generation Emulsion 
To be healthy with a quality life style is every human ’ s desire. According to documented 
Indian scriptures dating back to 5000 b . c ., nutritional status has always been 
associated with health [26] . Because nutritional depletion due to either changes in 
the quality or amount of dietary fat intake or abnormalities in lipid metabolism 
results in immunosuppression and therefore host defense impairment, favoring 
increased infection and mortality rates. 
Traditionally depletion in dietary fats in malnourished or hypercatabolic patients 
is compensated through intravenous feeding using a solution containing amino 
acids, glucose, electrolytes, and vitamins as well as nanosized emulsions. Structurally, 
an o/w - type emulsion is triglyceride (TG) droplets enveloped with a stabilizing 
superfi cial layer of phospholipids [27] . Emulsions for parenteral use are complex 
nutrient sources composed not only of fatty acids but also substances other than 
TG, such as phosphatidylcholine, glycerol, and . - tocopherol in variable amounts. 
The emulsions also had a complex inner structure and consisted of particles with 
different structures, namely, oil droplets covered by an emulsifi er monolayer, oil 
droplets covered by emulsifi er oligolayers, double - emulsion droplets, and possibly 
small unilamellar vesicles. Commercially available nanosized emulsions used as 
intravenous high - calorie nutrient fl uids have particle size normally around 160 – 
400 nm in diameter and, typically, their surfaces are normally negatively charged. 
Larger droplets can also be detected in commercially available emulsions [28] . Lutz 
et al. [29] reported that the mean diameter of particles in the 20% emulsions is 
larger than in the 10% emulsions. 
The TG in nanosized emulsions may be presented structurally in long or medium 
chains respectively, named LCT and MCT. The mean diameter of particles in LCT 
emulsions is greater than that in MCT emulsions [29] . LCT contains fatty acid chains 
with 14, 16, 18, 20, and 22 carbon atoms and sometimes with double bonds. The 
number of double bonds present defi nes the fatty acids in LCT as saturated, monounsaturad, 
or polyunsaturated. If the fi rst double bond is on carbon number 3, 6, or 
9 from the methyl end of the carbon chain, then the fatty acid is n - 3, n - 6, or n - 9, 
respectively. Purifi ed soybean or saffl ower oil contains LCT with a high proportion 
FIGURE 1 Flow chart of three generations of emulsion. 
Nanosized emulsion, NE 
First-generation ne (phospholipid based) 
Second-generation NE, with PEGylation on droplet surface 
Third-generation NE, with PEGylation and positive charge on droplet surface

GENERATIONS OF OIL-IN-WATER NANOSIZED EMULSIONS 1331 
of n - 6 polyunsaturated fatty acids whereas olive oil has LCT with n - 9 monounsaturated 
fatty acids. Fish oil includes LCT with 20 or more carbon atoms where the 
fi rst double bond is located between the third and fourth carbons from the methyl 
terminal of the fatty acids chain (omega - 3 or n - 3). On the other hand, MCT is 
derived from coconut oil and has saturated fatty acids with chains containing carbon 
atoms at the 6, 8, 10, or 12 positions. Both LCT and MCT either alone or MCT in 
combination with LCT are known for their long - term commercial acceptability in 
parenteral emulsions and are found in several U.S. Food and Drug Administration 
(FDA) – approved products. Also in Europe emulsion containing LCT/MCT enriched 
with fi sh oil became available for research. With MCT/LCT combinations in a specifi 
c ratio, nanosized emulsions appear to provide a more readily metabolizable 
source of energy [30] . However, LCT emulsion has been used in clinical practice for 
over 40 years. But for drug solubilization purpose, MCT is reported to be 100 times 
more soluble in water than LCT and thus to have an escalated solubilizing 
capability. 
7.4.2.2 Second - Generation Emulsion 
An easy and substantial association of lipophilic bioactive compound with the MCT 
or other vegetable oil – based emulsions, however, allows the emulsions to be used 
as vehicles/carriers for the formulation and delivery of drugs with a broad range of 
applications. These applications extend from enhanced solubilization or stabilization 
of the entrapped drug to sustained release and site - specifi c delivery. Hence the 
emulsions used for these applications are termed second - generation emulsions. Fittingly 
the o/w - type nanosized emulsions containing either positive or negative 
charge can be administered by almost all available routes, that is, topical, parenteral, 
oral, nasal, and even aerosolization into the lungs [31] . Despite differences in 
routes of administration, examples of commercially available emulsion - based formulations 
utilized for medical and nonmedical applications purposes are given in 
Table 1 . 
The lipid - induced enhancement in oral bioavailability of many drugs having poor 
water solubility is a well - known documented fact when the drugs are incorporated 
into emulsions [32, 33] . However, direct intravascularly or locally administered 
conventional fi rst - and second - generation emulsions could be taken up rapidly by 
the circulating monocytes for clearance by reticuloendothelial cells (through organs 
such as the liver, spleen, and bone marrow) [34] . Furthermore, the extent of clearance 
is enhanced by the adsorption of opsonic plasma proteins onto emulsion surfaces. 
However, the oily hydrophobic particles of the emulsions can also be taken 
up by macrophages without the necessity of opsonization provided the oil phase is 
liberated from the emulsion through the destabilization process occurring inside the 
blood compartment immediately after emulsion administration intravascularly or 
locally. Although the core of o/w emulsions is indeed hydrophobic, the emulsion 
envelope is not. The exposure of the hydrophobic part to the aqueous medium will 
therefore destabilize the emulsion. Moreover, Sasaki et al. [35] have assumed that 
when the castor oil – based emulsions interact with the tears in the eye, the electrolytes 
in the tears elicit a physical emulsion instability resulting in some release of 
the oil. The electrolytes present in blood or cellular fl uids can also cause a similar 
type of emulsion destabilization process, resulting in separation of the oil and water 

TABLE 1 Selected Marketed Medical and Nonmedical Emulsions 
Parenteral Fat Emulsions (o/w Type) for 
Nutrition Registered Emulsions (o/w Type) Containing Drugs 
Product Producer Product Drug Producer Application 
Abbolipid/Liposyn Abbott Diazepam - Lipuro Diazepam Braun Melsungen Intravenous 
Intralipid Pharmacia - Upjohn Diprivan Propofol AstraZeneca Intravenous 
Lipofundin N or Endolipide B. Braun Etomidat - Lipuro Etomidate Braun Melsungen Intravenous 
Lipofundin MCT/LCT 
— Lipotalon (Limethason) Dexamethasone palmitate Merckle Intra - arthr. 
Medialipide/Vasolipid B. 
Braun Stesolid Diazepam Dumex Intravenous 
Medianut B. 
Braun Sandimmune Cyclosporin A Novartis Oral 
Lipovenos Fresenius Neoral Cyclosporin A Novartis Oral 
Ivelip/Salvilipid Clintec/Baxter Gengraf Cyclosporin A Abbott Oral 
Clinoleic Clintec/Baxter Norvir Ritonavir Abbott Oral 
Intralipos Green Gross Restasis Cyclosporin A Allergan Ocular topical 
Kabimix Pharmacia - Upjohn Refresh Endura Drug free Allergan Ocular topical 
Triv e 1000 Baxter SA Fluad (MF59) Adjuvant Chiron Parenteral 
Perfl 
uorocarbon Emulsions (Fluorocarbon - in - Water Emulsions) 
Selected Topical Formulations Based on o/w or 
w/o Emulsions 
Product Producer Application Product Producer 
Fluosol DA 
Green Gross, 
Osaka Blood supplement or O 2 
carrier Daivonex cream and ointment 
Laboratoire Leo 
Imagent Alliance Pharmaceutical Contrast agent to image heart 
Voltaren emulgel Ciba - Geigy 
Oxygent Alliance Pharmaceutical Blood supplement or O 2 
carrier EMLA cream 
Astra, 
Swedan 
1332

GENERATIONS OF OIL-IN-WATER NANOSIZED EMULSIONS 1333 
phases from the parenterally administered emulsions. It is thus reasonable to say 
that the resultant oily hydrophobic particles of the emulsions would also be taken 
up by macrophages independent of an opsonization process. An opsonization 
process is the adsorption of protein entities capable of interacting with specifi c 
plasma membrane receptors on monocytes and various subsets of tissue macrophages 
(see Figure 2 ), thus promoting particle recognition by these cells. Classical 
examples of opsonic molecules include various subclasses of immunoglobulins [36, 
37] , complement proteins such as C1q and generated C3 fragments (C3b, iC3b) [38] , 
apolipoproteins [36, 37] , von Willebrand factor, thrombospondin, fi bronectin [39] , 
and mannose - binding protein. When given by other parentral routes, for example, 
intraperitoneally, subcutaneously, or intramuscularly, the majority of emulsion droplets 
enter the lymphatic system and eventually the blood circulation where particles 
behave as if given intravenously. Liver, spleen, and bone marrow uptake is signifi - 
cantly lower. Indeed, relative to the emulsion droplet size, lymph nodes take up a 
much greater (over 100 - fold) proportion than any other reticuloendothelial system 
(RES) tissue. 
There is increasing interest in developing injectable emulsions that are not cleared 
quickly from the circulation when they are designed to reach non - RES tissues in 
the vascular system or extravascular sites of action or to act as circulating drug 
reservoirs. Earlier approaches for making long - circulating emulsions concentrate 
mainly on changes in the oil phase of the emulsion such as MCT versus LCT [40] , 
use of structured lipids (SLS) having medium - chain (C 8 – C 10 ) fatty acids (SLM) and 
short - chain (C 4 ) fatty acids (SLS) [41] , addition of sphingomyelin [42 – 45] and cholesterol 
[46] to the emulsion, and use of hydrogenated castor oil (HCO) with at least 
20 oxyethylene units (HCO20) [47 – 52] . Using the further established formulation 
approaches by which the emulsion droplet surfaces could be altered might, however, 
be more realistic and even more useful for a wide array of drug - targeting purposes. 
Steric barrier or enhanced hydrophilicity effect exerted by a polyoxyethylene (POE) 
chain having surfactants when added as coemulsifi er into the phospholipid - 
stabilized fi rst - generation emulsion allows, to some extent, the passive/inverse drug 
targeting to the lung, kidneys, and areas of infl ammation [53, 54] . Addition of POE - 
based surfactants into the otherwise amphipathic phospholipid - stabilized emulsion 
FIGURE 2 Mononuclear phagocyte system. 
Promonocyte (bone marrow) 
Monocyte (blood) 
Macrophages (tissues) highly phagocytic 
Connective tissue (histiocyte) 
Liver (Kupffer cell) 
Lung (alveolar macrophage) 
Spleen (free and fixed macrophages, sinusoidal lining cell) 
Lymph node (free and fixed macrophage) 
Bone marrow (macrophages, sinusoidal lining cell) 
Serous cavity (peritoneal macrophage) 
Bone tissue (osteoclast) 
Nervous system (microglia)

1334 OIL-IN-WATER NANOSIZED EMULSIONS 
is particularly effective against plasma protein adsorption onto emulsion surfaces 
because of the hydrophilicity and unique solution properties of POE - based surfactants, 
including minimal interfacial free energy with water, high aqueous solubility, 
high mobility, and large exclusion volume [54] . In addition, colloidal particles presenting 
hydrophilic surfaces with a low contact angle will be almost ignored by 
phagocytic cells [55] , although emulsion particles are not supposed to be recognized 
as foreign by the body to some extent. Examples of POE chains containing surfactants 
employed so far in emulsions are Tween 80, Span 80, Brij, and Poloxamer 188 
(commercially named Pluronic F - 68 or Lutrol F - 68). The effectiveness of these 
polymeric surfactant molecules to intercalate at the oil – water interface with strong 
bonding to the phospholipid molecules could also be checked/judged through an in 
vitro monolayer experiment [56] . 
In general, the modifi cation of particulate carriers using amphipathic polyethylene 
glycol (PEG) – containing molecules results in a prolongation of their blood 
circulation time [57, 58] . A phosphatidylethanolamine derivative with polyethylene 
glycol (PEG – PE) is widely used to increase the plasma retention of particulate carriers 
such as liposomes [59 – 61] , polystyrene microspheres [62] , and nanospheres 
[63] . Therefore, similar to POE, the PEG – PE is also incorporated as a coemulsifi er 
into emulsions (termed PEGylated emulsion) to augment its circulation half - life 
[64] . Liu and Liu [53] studied the biodistribution of emulsion particles coated with 
phosphatidylethanolamine derivatives with three different molecular weight PEGs 
(MW 1000, 2000, and 5000). Among them, PEG - 2000 was able to prolong the circulation 
time of emulsion probably due to the increased hydrophilicity of the 
droplet surface and/or the formation of a steric barrier. However, Tirosh et al. [65] 
assumed, while characterizing the PEG - 2000 - grafted liposome by differential scanning 
calorimetry, densitometry, and ultrasound velocity and absorption measurements, 
that the steric stabilization is much more than increasing hydrophilicity. In 
addition, PEG - containing compounds also decrease the lipolysis of emulsion particles 
[47] and prevent the uptake by the mononuclear phagocytes [66] . 
A dipalmitoyl phosphatidylcholine (DPPC) – stabilized emulsion was prepared by 
Lundberg et al. [67] and the effect of addition of PEG – PE, polysorbate 80, or 
Pluronic F - 68 on the metabolism of DPPC - stabilized emulsion was studied. Two 
different radioactive markers were used to investigate the fate of emulsion particles 
following injection into the tail vein of female BALB/c inbred mice. While 14 C - triolein 
(TO) is susceptible to the action of lipoprotein lipase (LPL), 3 H - cholesteryl 
oleate ether (CO ether) is not. Hence the removal of 14 C - TO represents the triglyceride 
metabolism, whereas the other one is a core marker to represent whole particle 
removal by RES organ uptake. The emulsions with DPPC as sole emulsifi er 
were rapidly cleared from the blood with only 10 – 11% of CO or TO left in circulation 
after 1 h. However, addition of PEG – PE gave a prolonged clearance rate, 
especially during fi rst 3 h. A further addition of cosurfactant polysorbate 80 or 
Pluronic F - 68 resulted in a marked extension of the circulation lifetime during the 
fi rst 6 h. The notable effects of polysorbate 80 and Pluronic F - 68 can apparently be 
attributed mainly to the decrease in droplet size, although an additional infl uence 
due to the increased hydrophilicity may not be ruled out. 
The in vivo disposition of emulsions administered as nutrients (surface - 
unmodifi ed fi rst - generation emulsion) as well as administered as drug carriers 
(surface - modifi ed second - generation emulsion) would depend on the particle prop

GENERATIONS OF OIL-IN-WATER NANOSIZED EMULSIONS 1335 
erties, such as the size [68 – 71] , zeta potential (see Sections 7.4.2.3 and 7.4.5 ), and 
compositions of phospholipids and oil phase (see the above paragraphs), which may 
vary among different products and the batches of each product. The size of particulate 
carriers such as liposomes is known to infl uence both the phagocytic uptake by 
the mononuclear phagocyte system (MPS) [68 – 70] and the binding of apolipoprotein 
(apo) to emulsions [71] . Furthermore, the particle size is a major determinant 
of the transfer to extravascular spaces from the blood compartment. The capillaries 
of the vascular system can be classifi ed into three categories: continuous, fenestrated, 
and discontinuous (sinusoidal) [72] . Particulate carriers including nanosized 
emulsions are considered to pass through capillaries and reach extravascular cells 
only in organs having discontinuous capillaries such as liver, spleen, and bone 
marrow. In such tissues, the extravasation of particles should be regulated by their 
size since the largest pores in the capillary endothelium is reported to be about 
100 nm [73] . In addition, tumor capillaries have unique characteristics in their structures 
and functions in comparison with normal tissues such as muscle [74, 75] , which 
results in the enhanced distribution of particulate carriers to tumor tissues [76 – 78] . 
The distribution of emulsions within a tumor tissue was also regulated by the size 
of particulate carriers [79] . Obviously, because of the submicrometer size range 
(175 – 400 nm in diameter) of the emulsions, the more they circulate, the greater their 
chance of reaching respective targets. More specifi cally, growing solid tumors as well 
as regions of infection and infl ammation have capillaries with increased permeability 
as a result of the disease process (e.g., tumor angiogenesis [74] ). Pore diameters 
in these capillaries can range from 100 to 800 nm. Thus, drug - containing emulsion 
particles are small enough to extravasate from the blood into the tumor interstitial 
space through these pores [80] . Normal tissues, by and large, contain capillaries with 
tight junctions that are impermeable to emulsions and other particles of this diameter. 
This differential accumulation of emulsion - laden drug in tumor tissues relative 
to normal cells is the basis for the increased tumor specifi city for the emulsion - laden 
drug relative to free (nonemulsion) drug. In addition, tumors lack lymphatic drainage 
and therefore there is low clearance of the extravasated emulsion from tumors. 
Thus, long - circulating lipid carriers, such as POE/PEG - coated nanosized emulsions, 
tend to accumulate in tumors as a result of increased microvascular permeability 
and defective lymphatic drainage, a process also referred to as the enhanced permeability 
and retention (EPR) effect [81] . Table 2 lists various formulation factors 
affecting the metabolism as lipoproteins, the recognition by the MPS, and the elimination 
from the blood circulation of both second - and third - generation nanosized 
emulsions after parenteral administration. 
On the other hand, essential requirements of this “ active ” targeting approach 
include identifi cation of recognition features (receptors) on the surface of the target 
and the corresponding molecules (ligands) that can recognize the surface. Indeed, 
emulsions with appropriate ligands anchored on their surface must be able to access 
the target, bind to its receptors, and, if needed, enter it. Furthermore, in order to 
bring the colloidal carrier closer to otherwise inaccessible pathological target tissues, 
homing devices/ligands such as antibodies and cell recognition proteins are usually 
linked somehow onto the particle surfaces. Various methods have been employed 
to couple ligands to the surface of the nanosized lipidic and polymeric carriers with 
reactive groups. These can be divided into covalent and noncovalent couplings. 
Noncovalent binding by simple physical association of targeting ligands to the 

1336 OIL-IN-WATER NANOSIZED EMULSIONS 
nanocarrier surface has the advantage of eliminating the use of rigorous, destructive 
reaction agents. Common covalent coupling methods involve formation of a disul- 
fi de bond, cross - linking between two primary amines, reaction between a carboxylic 
acid group and primary amine, reaction between maleimide and thiol, reaction 
between hydrazide and aldehyde, and reaction between a primary amine and free 
aldehyde [82] . For antibody - conjugated second - generation anionic emulsions, the 
reaction of the carboxyl derivative of the coemulsifi er molecule with free amine 
groups of the antibody and disulfi de bond formation between coemulsifi er derivative 
and reduced antibody were the two reported conjugation techniques so far 
[83 – 85] . However, by the formation of a thio - ether bond between the free maleimide 
reactive group already localized at the o/w interface of the emulsion oil droplets 
and a reduced monoclonal antibody, the antibody - tethered cationic emulsion was 
developed for active targeting to tumor cells [86] . It should be added that the 
cationic emulsion investigated for tumor - targeting purpose belongs to the third - 
generation emulsion category (Section 7.4.2.3 ). 
Apart from non - RES - related disease treatment through target - specifi c ligand 
conjugation, the second - generation emulsions may also be useful for RES - related 
disease treatment. Certain lipoprotein or polysaccharide moiety inclusion into the 
emulsions would help to achieve this concept. In general, uptake of small colloidal 
drug carriers by the phagocytotic mononuclear cells of RES in the liver can be 
exploited to improve the treatment of parasitic, fungal, viral and bacterial diseases 
such as, for example, leishmaniasis, acquired immunodefi ciency syndrome (AIDS), 
and hepatitis B. The approach to use emulsions as a drug carrier against microbial 
TABLE 2 Formulation Factors Affecting Metabolism as Lipoproteins, Recognition by 
Mononuclear Phagocyte System (MPS), and Elimination from Blood Circulation of Second - and 
Third - Generation Nanosized Emulsions after Parenteral Administration 
Factor 
Metabolism as 
Lipoproteins Recognition by MPS 
Elimination from 
Blood Circulation 
Poor Extensive Poor Extensive Slow Rapid 
Particle size Large Small Small Large Small Large 
Emulsifi er DPPC EYPC DPPC DSPC DPPC EYPC 
DSPC — SM — SM DSPC 
SM — — — — — 
Coemulsifi er Poloxamers — Poloxamers — Poloxamers — 
HCO - 60 — HCO - 60 — HCO - 60 — 
PEG – PE — PEG – PE — PEG – PE — 
Polysorbates — Polysorbates — Polysorbates — 
Solutol — Solutol — Solutol — 
Cationic lipid SA/OA — SA/OA — SA/OA — 
Oil phase LCT MCT — — LCT MCT 
— — — — SLS SLM 
Note: DPPC, dipalmitoylphosphatidylcholine; DSPC, distearoylphosphatidylcholine; SM, sphingomyelin; 
EYPC, egg yolk phosphatidylcholine; HCO - 60, polyoxyethylene - (60) - hydrogenated castor oil; PEG – PE, phosphotidylethanolamine 
derivative with polyethylene glycol; SA, stearylamine; OA, oleylamine; LCT, long - chain 
triglyceride; MCT, medium - chain triglyceride; SLS, structured lipid with short - chain fatty acids, C 8 – C 10 ; SLM, 
structured lipid with medium - chain fatty acids, C 4 . 

GENERATIONS OF OIL-IN-WATER NANOSIZED EMULSIONS 1337 
diseases is superior to free antimicrobial agents in terms of both distribution to the 
relevant intracellular sites and treating disseminated disease states effectively. As 
already discussed, conventional emulsion particles are capable of localizing in liver 
and spleen, where many pathogenic microorganisms reside. 
Rensen et al. [87] demonstrated the active/selective liver targeting of an antiviral 
prodrug (nucleoside analogue, iododeoxyuridine) incorporated in an emulsion 
complexed with ligands such as recombinant apolipoprotein E (apoE) using the 
Wistar rat as animal model because its apoE – receptor system is comparable to that 
of humans [88] . Whereas the parent drug did not show any affi nity for emulsion 
due to its hydrophilic property, derivatization with hydrophobic anchors allowed 
incorporation of at least 130 prodrug molecules per emulsion particle without 
imparting any effect on the emulsion structure and apoE association to emulsion 
droplets. The authors did not describe where the 130 prodrug molecules reside in 
the emulsion and what is the emulsion/medium partition coeffi cient of the prodrug. 
The prodrug molecules might have reasonably higher solubility in the oil or o/w 
interface of the emulsion possibly due to a high partition coeffi cient value. Plausibly, 
this high partition value for prodrug molecules will determine the kinetic parameter 
koff (desorption rate of an emulsion component from the assembly), as suggested 
by Barenholz and Cohen [89] for liposomal technology. Furthermore, without being 
bound by theory, the apoE component helps to disguise the emulsion particles so 
that the body does not immediately recognize it as foreign but may allow the body 
to perceive it as native chylomicrons or very low density lipoproteins (VLDL). The 
small size and the approximately spherical shape allow the emulsion particles to 
exhibit similar physicochemical properties to native chylomicrons or VLDL (hydrolyzed 
by LPL) whereas the incorporated prodrug remained associated with the 
emulsion remnant particles following injection into the blood circulation of the rat 
[87] . Because the carrier particles are not recognized as foreign, the systemic circulation 
of the drug increases, thus increasing the likelihood of drug delivery to the 
target tissues (up to 700 n M drug concentration in liver parenchymal cells). Additionally, 
the clearance rate of the drug decreases, thereby reducing the likelihood 
of toxic effects of the drug on clearance tissues since accumulation of the drug in 
another part of the clearance tissues is reduced. Thus, specifi c organs may be targeted 
by using nanosized emulsion particles as described above due to target cells 
comprising high levels of specifi c receptors, for example, but not limited to apoE 
receptors. 
To address this issue, the saccharide moieties of glycolipids and glycoproteins on 
the cell surface are considered to play an important role in various intercellular 
recognition processes. For instance, Iwamoto et al. [90] investigated the infl uence of 
coating the oil droplets in emulsion with cell - specifi c cholesterol bearing polysaccharide, 
such as mannan, amylopectin, or pullulan, on the target ability of those 
formulations. They observed a higher accumulation of mannan - coated emulsion in 
the lung in guinea pigs. Thus selective drug targeting through emulsion - bearing 
ligands not only leads to an improved drug effectiveness and a reduction in adverse 
reactions but also offers the possibility of applying highly potent drugs. Hence, the 
composition of the emulsion plays an important role concerning intercellular cell 
recognition processes and, indeed, cell recognizability is also being improved by 
incorporation of apoproteins or galactoproteins onto the emulsion particles to 
enhance their specifi city [91] . 

1338 OIL-IN-WATER NANOSIZED EMULSIONS 
Overall, although second - generation emulsion is usually used as a means of 
administering aqueous insoluble drugs by dissolution of the drugs within the oil 
phase of the emulsion, employing surface modifi cation/PEGylation by the attachment 
of targeting ligands (apoE, polysaccharide, and antibody) onto the droplet 
surface of emulsions may be useful for both passive and active drug - targeting purposes. 
Thus receptor - mediated drug targeting using ligands attached to emulsions 
seems to hold a promising future to the achievement of cell - specifi c delivery of 
multiple classes of therapeutic cargoes, and this approach will certainly make a 
major contribution in treating many life - threatening diseases with a minimum of 
systemic side effects. 
7.4.2.3 Third - Generation Emulsion 
In order to increase cellular uptake, a cationization strategy is applied particularly 
on the surfaces of nonviral, colloidal carrier systems such as liposomes, nano - and 
microparticulates, and nanocapsules [92] . To make the surface of these lipidic and 
polymeric carrier systems a cationic property, some cationic lipids/polymers are 
usually added into these systems during/after preparation. But, adding only the 
cationic substances in phospholipids - stabilized fi rst - generation emulsions does not 
help to obtain a physically stabilized emulsion for a prolonged storage period. 
However, using different cationic lipids as emulsifi er and additional helper lipids 
as coemulsifi er, for example, DOTAP, 1,2 - dioleoyl - sn - glycero - 3 - phosphoethanolamine 
(DOPE), and 1 - palmitoyl - 2 - oleoyl - sn - glycero - 3 - phosphoethanolamine - N - 
[poly(ethylene glycol) 2000 ] (PEG 2000 PE), reports are available to prepare emulsions 
with positive charges on their droplet surfaces [93, 94] . Alternatively, inclusion of 
cation - forming substances such as lipids (stearyl or oleyl chain having primary 
amines) [95, 96] , polymers (chitosan) [97, 98] , and surfactants (cetyltrimethylammonium 
bromide) [99] during the preparation of second - generation emulsion 
allows the formation of a stabilized system with positive charges over on it. 
Further, the positive charge caused by stearylamine was also confi rmed by a 
selective adsorption of thiocyanate. Its adsorption was correlated with increasing 
stearylamine concentration [95] . So, nanosized emulsion consisting of complex 
emulsifi ers, that is, phospholipid – polyoxyethylene surfactant - cationized primary 
amine or a polymer combination, can conveniently be termed third - generation 
emulsion. 
The extemporaneous addition of the solid drug or drug previously solubilized in 
another solvent or oil to the preformed fi rst - and second - generation emulsions is 
not a favored approach as it might compromise the integrity of the emulsion. Since 
therapeutic DNA and single - stranded oligonucleotides or small interfering ribonucleic 
acid (siRNA) are water soluble due to their polyanionic character, the aqueous 
solution of these compounds need to be added directly to the preformed third - generation 
emulsion in order to interact electrostatically with the cationic emulsion 
droplets and thus associate/link superfi cially at the oil – water interface of the emulsion 
[100, 101] . During in vivo conditions when administered via parenteral, nasal, 
and ocular routes, the release of the DNA and oligonucleotides from the associated 
emulsion droplet surfaces should therefore initially be dependent solely on the 
affi nity between the physiological anions of the biological fl uid and the cationic 
surface of the emulsion droplets. For instance, the mono - and divalent anions con

GENERATIONS OF OIL-IN-WATER NANOSIZED EMULSIONS 1339 
taining biological fl uid available in the parenteral route is plasma and in ocular 
topical route is tear fl uid, aqueous humor, and vitreous humor. Moreover, these 
biofl uids contain a multitude of macromolecules and nucleases. There is a possibility 
that endogenous negatively charged biofl uid components could dissociate the DNA 
and oligonucleotides from cationic emulsion. It is noteworthy to conduct, during the 
preformulation development stages, an in vitro release study for therapeutic DNA 
and oligonucleotide - containing emulsion in these biological fl uids, and this type of 
study could be considered an indicator of the strength of the interaction that 
occurred between DNA or oligonucleotide and the emulsion particles. However, it 
is interesting to see what could happen when the third - generation emulsion is 
applied to in vitro cell culture models in the presence of serum. The serum stability 
of the emulsion – DNA complex was reported [102, 103] . Further interesting investigations 
using third - generation emulsions in gene delivery purposes are briefl y summarized 
in a review article [104] . 
7.4.2.4 Unique Property of Third - Generation Emulsion 
To enhance the drug - targeting effi cacy of colloidal carriers such as nanospheres and 
liposomes, a PEGylation/cationization strategy is traditionally made over the surface 
of these carriers. While surface PEGylated colloidal carriers exhibit a prolonged 
plasma residence time through an escaping tendency from RES uptake following 
parenteral administration, surface - cationized colloidal carriers can facilitate the 
penetration of therapeutic agents into the cell surface possibly via an endocytotic 
mechanism. These two facts are proved in both liposomes and nanospheres when 
they possess separately the cationic and PEGylatic surface modifi cations on them. 
However, a cationic emulsion colloidal carrier system developed in Simon Benita ’ s 
laboratory at Hebrew University of Jerusalem, Israel, differs signifi cantly in such a 
way that it holds a combination of cationic and PEGylatic surface properties on it. 
Benita ’ s group have prepared a novel cationic emulsion vehicle using a combination 
of emulsifi ers consisting of Lipoid E 80, Poloxamer 188, and stearylamine and have 
found the formulation suitable for parenteral use, ocular application, nasal drug 
delivery, and topical delivery [105] . 
It has been reported in an ocular pharmacokinetic study of cyclosporin A incorporated 
in deoxycholic acid – based anionic and stearylamine - based cationic emulsions 
in rabbit that, when compared to anionic emulsion, the cationic emulsion 
showed a signifi cant drug reservoir effect of more than 8 h in corneal and conjunctival 
tissues of the rabbit eye following topical application [106] . Since cornea and 
conjunctiva are of anionic nature at physiological pH [107] , the cationic emulsion 
would interact with these tissues electrostatically to implicate the observed cyclosporin 
A reservoir effect. This hypothesis is supported, in principle, by an ex vivo 
study which showed that cationic emulsion carrier exhibited better wettability properties 
on rabbit cornea than either saline or anionic emulsion carrier [108] . 
Studies [109, 110] have shown that small changes in physical properties of emulsions 
can infl uence the elimination rate of these formulations from the blood. 
Indeed, an organ distribution study of stearylamine - based cationic or deoxycholic 
acid – based anionic nanosized emulsions and Intralipid, a well - known commercial 
anionic emulsion, containing 14 C - CO was carried out following injection into the tail 
vein of male BALB/c mice (20 – 26 g) at a volume of 5 mL/kg [111, 112] . Since CO 

1340 OIL-IN-WATER NANOSIZED EMULSIONS 
(cholesteryl oleate) is one of the most lipophilic compounds used in biopharmacy 
and is not prone to degradation in the body (which remains within particles even 
after lipolysis of emulsion), its in vivo behavior can be regarded as refl ecting that 
of the injected nanosized emulsion in the early phase [42, 113] . Following intravenous 
administration of the various emulsions having 14 C - CO to BALB/c mice, the 
14 C - CO was found to accumulate in organs such as lung and liver. Furthermore, it 
was observed that the concentration of 14 C - CO in the lung decreased but was again 
elevated over time for both the developed cationic and anionic emulsion formulations, 
with a concomitant decrease in the concentration of the radiolabeled compound 
in the liver. However, within the various emulsion distribution patterns 
observed in liver, a lower 14 C - CO concentration was observed for stearylamine - 
based cationic emulsion when compared to Intralipid while for deoxycholic acid – 
based anionic emulsion the observed concentration of 14 C - CO was relatively very 
low when compared to cationic emulsion and Intralipid. In addition, in comparison 
to both anionic emulsions, the stearylamine - based cationic emulsion elucidated a 
much longer retention time of 14 C - CO in the plasma, clearly indicating a long circulating 
half - life for cationic emulsion in the blood. Thus, the cationic nanosized 
emulsion can be considered a stealth long - circulating emulsion. 
The above two studies clearly described the unique characteristics of third - 
generation emulsion in enhancing ocular drug bioavailability; on the other hand, 
the same emulsion has the property of circulating for a longer time in blood following 
parenteral administration. Excess positive charge at the oil – water interface in 
conjunction with the projection of highly hydrophilic POE chain (due to the presence 
of Poloxamer 188) toward the aqueous phase of the o/w - type nanosized emulsion 
is the main reason behind the emulsion attaining its unique property, which is 
absent in fi rst - and second - generation emulsions. However, a better understanding 
of the structure of the third - generation emulsion in terms of forces involved in its 
formation and stabilization must ultimately be obtained in an effort to provide a 
clearly understood physical basis for the uniqueness in its biological effi cacy following 
parenteral and ocular administration. 
It should be added that the use of stearylamine in intravenous administered 
emulsion might be problematic. Stearylamine is a single - chain amphiphile having 
relatively high critical micellar concentration, although the concentration used in 
the studied emulsion is much higher than the critical micellar concentration. Therefore, 
due to the dilution in plasma as well as plasma lipoproteins and blood cells, 
there is a high probability that the emulsion will lose its stearylamine almost instantaneously. 
To substantiate indirectly this issue, Klang et al. [114] showed the lack of 
potential induced toxicity of stearylamine - based cationic emulsion in animal models 
in vivo and Korner et al. [115] investigated the surface properties of mixed phospholipid 
– stearylamine monolayers and their interaction with a nonionic surfactant 
(poloxamer) in vitro. Despite the presence of the stearylamine, which may be suspected 
of being an irritant in pure form, in the emulsifi er combination, the hourly 
instillation of stearylamine - based cationic emulsion vehicle into rabbit eye was well 
tolerated without any evidence of any toxic or infl ammatory response to the ocular 
surface during the 5 days of the study (40 single - drop instillations between 8 AM 
and 4 PM each day) [114] . Following 0.2 - , 0.4 - , and 0.6 - mL single - bolus injections 
of the same emulsion vehicle, representing a huge single administered dose of 
30 mL/kg, no animal deaths were noted over a period of 30 days, apparently indicat

ing the absence of marked acute toxicity [114] . Furthermore, the same stearylamine - 
based cationic emulsion vehicle did not cause acute neurotoxicity in rats when a 
continuous intravenous infusion (3.3 mL) for 2 h at a rate of 27.4 . L/min was administered 
through the jugular vein [114] . An another study from Benita ’ s laboratory 
suggests that long - term subchronic toxicity examination of the rabbit eye (healthy) 
following thrice - daily single - drop topical instillation of the stearylamine - based 
emulsion elicited an almost similar nonirritating effect to eye tissues in comparison 
to the thrice - daily single - drop topical instillation of the normal saline – treated 
control rabbit eyes (unpublished data). Thus, overall results clearly indicated that 
the stearylamine was strongly bound at a molecular level to the mixed interfacial 
fi lm formed by Lipoid E 80 and poloxamer 188 at the oil – water interface system 
[115] . Such an intercalation between the emulsifi ers is responsible for emulsifi ed oil 
droplet stability and, in fact, prevented the stearylamine from leaking and exerting 
any intrinsic possible local or systemic adverse effects in model animals. 
7.4.3 PREPARATION METHODS FOR DRUG - FREE/LOADED 
OIL - IN - WATER NANOSIZED EMULSIONS 
To get a better idea of how to formulate the nanosized emulsion delivery systems 
suitable for parenteral, ocular, percutaneous, and nasal uses, the reader is referred 
to more detailed descriptions of methods of nanosized emulsion preparation [6, 
116] . A hot - stage high - pressure homogenization technique or combined emulsifi cation 
technique (de novo production) is frequently employed in order to prepare 
nanosized emulsions with desired stability even after subjection to autoclave sterilization. 
Therefore, the steps involved in this technique in making blank anionic and 
cationic emulsions were arranged in the following order: 
1. Weigh the oil - and water - soluble ingredients in separate beakers. 
2. Heat both oil and water phases separately to 70 ° C. 
3. Add the oil phase to the water phase and continue the heating up to 80 ° C 
with constant stirring to form a coarse emulsion. 
4. Mix at high shear to make a fi ne emulsion. 
5. Cool the fi ne emulsion formed in ice bath. 
6. Homogenize the fi ne emulsion. 
7. Cool the homogenized emulsion in ice bath. 
8. Filter the emulsion using a 0.5 . m membrane fi lter. 
9. Adjust the emulsion to 7 using 0.1 N hydrochloric acid or 0.1 N sodium 
hydroxide solution. 
10. Pass nitrogen gas into the vials containing the emulsion. 
11. Sterilize the emulsion using an autoclave. 
The traditional droplet size – reducing steps involved during the preparation include 
constant mild stirring using a magnetic stirrer when initially mixing oil and water 
phases, rapid Polytron mixing at high speed, and fi nal droplet size homogenization 
using a two - stage homogenizer valve assembly. The initial heating is vital for the 
DRUG-FREE/LOADED OIL-IN-WATER NANOSIZED EMULSIONS 1341

1342 OIL-IN-WATER NANOSIZED EMULSIONS 
effective solubilization of the respective oil and water phase components in their 
corresponding phases. Mixing the two phases with constant mild stirring and subsequently 
raising the temperature to 85 ° C are needed to form an initial coarse 
emulsion and to localize the surfactant molecules for better adsorption at the oil – 
water interface, respectively. A typical formula to make anionic and cationic nanosized 
emulsions is given in Table 3 . 
There are three different approaches to incorporate lipophilic drugs into the oil 
phase or at the o/w interface of the nanosized emulsions, namely, extemporaneous 
drug addition, de novo emulsion preparation, and an interfacial incorporation 
approach, which includes the recently developed SolEmul technology [117] . In 
principle, the lipophilic drug molecules should however be incorporated by a de 
novo process. Thus, the drug is initially solubilized or dispersed together with an 
emulsifi er in suitable single - oil or oil mixture by means of slight heating. The water 
phase containing the osmotic agent with or without an additional emulsifi er is also 
heated and mixed with the oil phase by means of high - speed mixers. Further homogenization 
takes place to obtain the needed small droplet size range of the emulsion. 
A terminal sterilization by fi ltration, steam, or autoclave then follows. The emulsion 
thus formed contains most of the drug molecules within its oil phase. This is a generally 
accepted and standard method to prepare lipophilic drug – loaded nanosized 
emulsions for parenteral, ocular, percutaneous, and nasal uses, as illustrated in 
Figure 3 . This process is normally carried out under aseptic conditions and nitrogen 
atmosphere to prevent both contamination and potential oxidation of sensitive 
excipients. 
7.4.4 EXCIPIENT INCLUSION: OIL - IN - WATER NANOSIZED 
EMULSIONS 
In general, nanosized emulsions should be formulated with compatible vehicles and 
additives. The components of the internal and external phases of emulsion should 
be chosen to confer enhanced solubility and/or stability to the incorporated biologically 
active lipophilic drug. In addition, it should also be designed to infl uence 
biofate or therapeutic index of the incorporated drug following administration via 
parenteral, ocular, percutaneous, and nasal routes. In this section, general considerations 
concerning excipient selection and optimum concentrations are comprehen- 
TABLE 3 Typical Formula to Make o/w Anionic and 
Cationic Nanosized Emulsions 
Oil Phase Water Phase 
Natural/semisynthetic oils Poloxamer 188 
Phospholipid mixture Glycerol 
Stearylamine/oleylamine a Double - distilled water 
Deoxycholic acid/oleic acid b 
Vitamin E 
a Necessary ingredient for cationic emulsions. 
b Necessary ingredient for anionic emulsions. 

sively presented mainly in their relation to the oil phase, the aqueous phase, and 
the emulsifi ers. 
Prior to the formulation design of the emulsions, data are needed concerning the 
drug solubility in the oil vehicle. Additionally, prerequisite information is needed 
about compatibility of the oil vehicle with other formulation additives and the 
established ocular/skin tissues – oil vehicle matching before the dosage form can be 
prepared. Table 4 lists the common emulsion excipients and the oils suitable for 
dissolving or dispersing lipophilic drugs of ocular/parenteral interests. Since oils are 
triglycerides, care must be taken to minimize or eliminate oxidation. Therefore, 
antioxidants such as . - tocopherol (0.001 – 0.002% w/w) should be included in a 
typical emulsion formulation for medical applications. The fi nal oil - phase concentration 
in emulsions meant for ocular use is now widely accepted to be at or below 
5% w/w taking into account that the emulsion must be kept in a low - viscosity range 
of between 2 and 3 centipoises, which also is the optimal viscosity for ocular preparations 
[118] . However, for all other medical uses, the amount of oil may be varied 
but generally is within 5 – 20% w/w. Sometimes, a mixture of oils rather than a single 
oil is employed since drug solubilization in the oil phase is a prerequisite to exploiting 
the emulsion advantages. Jumaa and M u ller [98, 119] reported the effect of 
mixing castor oil with MCT on the viscosity of castor oil. The oil combination at the 
ratio of 1 : 1 (w/w) led to a decrease in the viscosity of castor oil and simultaneously 
to a decrease in the interfacial tension of the oil phase. This was related to the free 
fatty acids contained in castor oil, which can act as a coemulsifi er resulting in lower 
interfacial tension and, simultaneously, in a more stable formulation in comparison 
with the other oil phases. In addition to the digestible oils from the family of triglycerides, 
including soybean oil, sesame seed oil, cottonseed oil, and saffl ower oil, 
which are routinely used for making medical emulsions, alternative biocompatible 
FIGURE 3 Preparation of o/w nanosized emulsion (de novo method). 
EXCIPIENT INCLUSION: OIL-IN-WATER NANOSIZED EMULSIONS 1343

1344 OIL-IN-WATER NANOSIZED EMULSIONS 
oils such as . - tocopherol and/or other tocols were also investigated for drug delivery 
purposes via o/w emulsions [120, 121] . But the emulsions formed from tocols are 
often considered as microemulsion systems with few exceptions being the nanosized 
emulsions [122, 123] . 
Unlike spontaneously forming thermodynamically stable microemulsion systems 
that require a high surfactant concentration (20% and higher), the kinetically stable 
nanosized emulsions can be prepared by using relatively lower surfactant concentrations. 
For example, a 20% o/w nanosized emulsion may only require a surfactant 
concentration of 5 – 10%. Traditionally, lecithins or phospholipids are the emulsifi ers 
of choice to produce nanosized emulsions. However, emulsifi ers of this kind are not 
suitable to produce submicrometer – sized emulsion droplets or to withstand the heat 
during steam sterilization. Therefore, additional emulsifi ers preferably dissolved in 
the aqueous phase are usually included in the emulsion composition. A typical 
example of the aqueous soluble emulsifi ers is nonionic surfactants (e.g., Tween 20) 
after taking into consideration their nonirritant nature when compared to ionic 
surfactants. The nonionic block copolymer of polyoxyethylene – polyoxypropylene, 
Pluronics F68 (Poloxamer 188), is included to stabilize the emulsion through strong 
steric repulsion. However, amphoteric surfactants Miranol MHT (lauroamphodiacetate 
and sodium tridecethsulfate) and Miranol C 2 M (cocoamphodiacetate) were 
also used in earlier ophthalmic emulsions [124] . It should be added that commercially 
available cyclosporin A – loaded anionic emulsion (Restasis) contains only 
polysorbate 80 and carbomer 1342 at alkali pH to stabilize the drug - loaded anionic 
emulsion. To prepare a cationic emulsion, cationic lipids (stearyl and oleylamine) 
or polysaccharides (chitosan) are added to the formulation. Strikingly, if chitosan is 
a choice of cation producing polysaccharide emulsifi er molecules, there is no need 
to add amphoteric or nonionic surfactants to the phospholipid or lecithin - stabilized 
TABLE 4 Excipients Used for Formulation of o/w Nanosized Emulsions 
Oils Emulsifi ers 
Cationic Lipids and 
Polysaccharide Miscellaneous 
Sesame oil Cholesterol Stearylamine . - Tocopherol 
Castor oil Phospholipids (Lipoid) Oleylamine Glycerin 
Soya oil Polysorbate 80 and 20 
(Tween 80 and 20) 
Chitosan Xylitol 
Paraffi n oil Transcutol P Sorbitol 
Paraffi n light Cremophor RH Thiomersal 
Lanolin Poloxamer 407 EDTA 
Vaseline Poloxamer 188 Methyl paraben 
Corn oil Miranol C 2 M and MHT Propyl paraben 
Glycerin monostearate Tyloxapol TPGS 
Medium - chain 
monoglycerides 
Medium - chain 
triglycerides 
Squalene 
Vitamin E 
Note: TPGS, . - tocopheryl - polyethylene glycol - 1000 - succinate; EDTA, ethylenediamine tetraacetic 
acid. 

emulsion [125] . Conversely, a cationic emulsion based on an association of poloxamer 
188 and chitosan without lecithin was prepared and also showed adequate 
physicochemical properties regarding stability and charge effects [97, 98] . Oil - in - 
water emulsion compositions based on a tocopherol (or a tocopherol derivative) as 
the disperse phase have been described in a patent granted to Dumex [126] . The 
emulsion is intended for use with compounds that are sparingly soluble in water. 
Interestingly, the emulsifying agent used to make tocol - based emulsions are restricted 
to vitamin E TPGS. 
Additives other than antioxidants such as preservatives (e.g., benzalkonium chloride, 
chlorocresol, parabens) are regularly included in emulsions to prevent microbial 
spoilage of multidose medical emulsions. . - Tocopherol is a good example of an 
antioxidant used to obtain a desirable stabilized emulsion under prolonged storage 
conditions. The presence of components of natural origin such as lecithin or oils 
with high calorifi c potential renders the emulsion a good medium to promote microbial 
growth when it is packed in multidose containers. Pharmaceutical products 
when distributed into multidose containers, especially for parenteral and ocular 
administrations, should be properly preserved against microbial contamination and 
proliferation during storage in normal conditions and proper use. Incorporation of 
preservatives in single - dose vials is also a common procedure if fi ltration is used as 
a sterilization method. Sznitowska et al. [127] studied the physicochemical compatibility 
between the lecithin - stabilized emulsion and 12 antimicrobial agents over two 
years of storage at room temperature. Preliminary physicochemical screening results 
indicate that addition of chlorocresol, phenol, benzyl alcohol, thiomersal, chlorhexidine 
gluconate, and bronopol should be avoided due to the occurrence of an unfavorable 
pH change followed by coalescence of lecithin - stabilized droplets of the 
emulsion. Furthermore, the effi cacy of antimicrobial preservation was assessed using 
the challenge test according to the method described by the European Pharmacopoeia. 
Despite good physicochemical compatibility, neither parabens nor benzalkonium 
chloride showed satisfying antibacterial effi cacy in the emulsion against the 
tested microorganisms and consequently did not pass the test. Therefore, higher 
concentrations of antimicrobial agents or their combination may be required for 
effi cient preservation of the lecithin - stabilized emulsion probably because of unfavorable 
phase partitioning of the added antimicrobials within the different internal 
structures of the emulsion. It is interesting to note that benzalkonium chloride, a 
highly aqueous soluble drug, did not pass the standard challenge test even when 
incorporated in a cationic emulsion, particularly the third - generation category 
(unpublished data). This fi nding clearly indicates that the possible electrostatic 
attraction between the negatively charged lipid moieties of the mixed emulsifying 
fi lm formed around the anionic emulsifi ed oil droplets [127] and the quaternary 
cationic ammonium groups of the preservative is not the plausible cause for the 
reduced activity of the benzalkonium chloride. Thus, the possible intercalation of 
this surfactant in either the cationic or anionic interfacial mixed emulsifying fi lm is 
likely to occur, preventing benzalkonium chloride from eliciting its adequate preservative 
action. Overall, it is preferable to formulate nanosized emulsions devoid 
of preservative agents and fi ll it in sterile single - dose packaging units to prevent 
potential contamination due to repeated use of multidose packaging. It should be 
pointed out that the two available ocular emulsion products (Refresh Endura and 
Restasis, Allergan, Irvine, CA) on the market are preservative free and packed in 
EXCIPIENT INCLUSION: OIL-IN-WATER NANOSIZED EMULSIONS 1345

1346 OIL-IN-WATER NANOSIZED EMULSIONS 
single - use vials only. Currently there is no commercial parenteral emulsion which 
contains preservatives and research concerning the problem of preservation of 
nanosized emulsion is very limited [128 – 131] . 
7.4.5 MEDICAL APPLICATIONS OF OIL - IN - WATER 
NANOSIZED EMULSIONS 
It has been shown in a number of studies that the incorporation of drug in o/w 
nanosized emulsions signifi cantly increased the absorption of the drug when compared 
with the equivalent aqueous solution administered orally [132 – 135] . However, 
the use of emulsions for oral application is limited since other attractive alternatives, 
such as self - emulsifying oil delivery systems, which are much less sensitive and easy 
to manufacture, are available [136, 137] . Thus the potential of nanosized emulsions 
after administration with parenteral and traditional nonparenteral topical routes 
such as ocular, percutaneous, and nasal is covered in this section. 
7.4.5.1 Parenteral Routes 
The o/w nanosized emulsion formulations of lipophilic drugs, such as propofol, 
etomidate, dexamethasone palmitate, and diazepam, were already developed and 
marketed (Table 1 ). Furthermore, various research groups across the world are currently 
undertaking projects to exploit the potential of o/w emulsions for parenteral 
delivery of a myriad of investigational drugs as well as other lipophilic drugs by 
receptor - mediated targeting to cancer cells. The important technical and clinical 
points to keep in mind before the use of the emulsion systems for this kind of work 
are given below. 
It has to be clear that, once diluted and injected (or administered in ocular and 
other routes), the emulsion stability and fate are determined by three measurable 
parameters. The fi rst is the partition coeffi cient of each emulsion component (including 
added drugs and agents) between the emulsion assembly and the medium. To 
some extent this partition coeffi cient is related to oil – water and/or octanol – water 
partition coeffi cients. For example, it was well demonstrated that per component of 
which log P is lower than 8, the stability upon intravenous (IV) injection is questionable 
[42, 138] . The other two parameters are koff , a kinetic parameter which describes 
the desorption rate of an emulsion component from the assembly, and kc , the rate 
of clearance of the emulsion from the site of administration. This approach is useful 
to decide if and what application a drug delivery system will have a chance to 
perform well [89] . 
Stability in plasma is an important requirement for IV emulsions as fl occulated 
droplets may result in lung embolism. It was found that tocol - based emulsions stabilized 
by sodium deoxycholate/lecithins fl occulated strongly when mixed with 
mouse, rat, and sheep plasma and serum, whereas soya oil – based emulsions with 
the same emulsifi ers did not [123] . It was hypothesized that this effect was caused 
by the adsorption of plasma proteins onto the tocol droplets (opsonization). Indeed, 
the steric stabilization of emulsions by incorporation of emulsifi ers like poloxamer 
188 or PEGylated phospholipids such as PEG 5000 PE proved to be effective in the 
stabilization of tocol - based emulsions in plasma. Conversely, in vitro studies were 

conducted on plasma protein adsorption onto the blank second - and third - 
generation emulsion droplets [37, 139] to mimic the in vivo opsonization phenomenon 
responsible for the rapid clearance of the emulsion droplets from the blood. 
According to these authors, the adsorption of many protein species such as apoAs, 
apoCs, apoE albumin, fi brinogen, and gamma globulin onto the emulsion droplet 
surfaces is detectable by two - dimensional polyacrylamide gel electrophoresis. 
7.4.5.2 Ocular Routes 
For the eye, the method of drug delivery is important. However, when nanosized 
emulsion is used as a vehicle for ocular drug delivery purposes, both topical/local and 
intraocular routes of administration can be possible (though no data concerning 
intraocular drug delivery through emulsion are currently available). The o/w nanosized 
emulsions having both anionic and cationic charges provide a liquid - retentive 
carrier for ocular active agents, particularly when topically instilled into the eye. It is 
interesting to add here that thermodynamically stable and optically isotropic colloidal 
systems such as the w/o microemulsion is also designed nowadays for ocular 
topical. As delivery the w/o microemulsion system comprises both aqueous and oily 
components into its multistructure, it has the ability to incorporate considerable 
amounts of both hydrophilic and lipophilic drugs [140] . In fact, in comparison to 
ocular inserts or implants and semisolid ocular preparations, the liquid - retentive 
nature gives impetus to investigating further the emulsion - based ophthalmic drug 
delivery as it has the benefi t of being comfortable to use for both ophthalmologists 
and patients. In addition, through topical instillation of emulsions possessing ocular 
active substances, the delivery of drug molecules even to the posterior portion of the 
eye might be of possible. In this context, the third - generation emulsion is being 
designed by adsorbing electrostatically the therapeutic oligonucleotides onto its 
surface for modulating functions of retinal pigment epithelium (RPE) cells effectively 
in order to treat blindness associated with age - related macular degeneration 
(AMD), proliferative vitreoretinopathy, retinal and choroidal neovascularization, 
and retinitis pigmentosa. To achieve this, it becomes necessary to know fi rst the ocular 
protective mechanisms and other concomitant factors to be faced by emulsion droplets 
following ocular topical application. This point is further developed below. 
Considerations of ocular drug delivery are not detailed in this section. Pertinent 
information concerning factors affecting drug permeation or retention as well as 
eye anatomy and physiology can be found in several reviews [141 – 146] . From a 
medical point of view, o/w nanosized emulsions for ophthalmic use aim at enhancing 
drug bioavailability either by providing prolonged delivery to the eye or by facilitating 
transcorneal/transconjunctival penetration. Drugs incorporated in o/w nanosized 
emulsions are lipophilic in nature, and depending on the extent of lipophilicity, 
either the corneal or the conjunctival/scleral route of penetration may be favored 
[147] . For the more lipophilic drugs the corneal route was shown to be the predominant 
pathway for delivering drugs to the iris, whereas the less lipophilic drugs 
underwent conjunctival/scleral penetration for delivery into the ciliary body [147] . 
Thus, transcorneal permeation has traditionally been the mechanism by which topically 
applied ophthalmic drugs are believed to gain access to the internal ocular 
structures. Relatively little attention has been given to alternate routes by which 
drugs may enter the eye. It was reported that drugs may be absorbed by the 
MEDICAL APPLICATIONS OF OIL-IN-WATER NANOSIZED EMULSIONS 1347

1348 OIL-IN-WATER NANOSIZED EMULSIONS 
noncorneal route and appeared to enter certain intraocular tissues through the 
conjunctiva/sclera [148 – 150] . Indeed when compared to the cornea, drug penetration 
through the conjunctiva has the advantage of a larger surface area and higher 
permeability, at least for drugs which are not highly lipophilic. Furthermore, the 
lasting presence of drug molecules in the lower conjunctival cul - de - sac of the eye 
could result in a reservoir effect. Nevertheless, the o/w nanosized emulsions more 
or less physically resemble a simple aqueous - based eye drop dosage form since 
more than 90% of the external phase is aqueous irrespective of the formulation 
composition. Hence, following topical administration, nanosized emulsions would 
probably face almost similar ocular protective events as encountered with conventional 
eye drops into the eye. The o/w nanosized emulsions are likely to be destabilized 
by the tear fl uid electrolytic and dynamic action. Because of constant eyelid 
movements, the basal tear fl ow rate (1.2 . L/min), and the refl ex secretion induced 
by instillation (up to 400 . L/min depending on the irritating power of the topical 
ocular solutions [35] ), topical eye drop dosage forms are known for being rapidly 
washed out from the eye. Therefore, the water phase of the emulsion is drained off 
while, probably, the oil phase of the emulsion remains in the cul - de - sac for a long 
period of time and functions as a drug reservoir [35] . If the volume of instilled emulsion 
in the eye exceeds the normal lachrymal volume of 7 – 10 . L, then the portion 
of the instilled emulsion (one or two drops, corresponding to 50 – 100 . L) that is not 
eliminated by spillage from the palpebral fi ssure of conjunctiva is drained quickly 
via the nasolacrimal system into the nasopharynx. In other words, the larger the 
instilled volume, the more rapidly the instilled emulsion is drained from the precorneal 
area. Hence the contact time of the emulsion with the absorbing surfaces 
(cornea and conjunctiva) is estimated to be a maximum of a few minutes, well 
beyond the short residence time of conventional eye drops. In order to verify the 
extension of the residence time of the emulsion in the conjunctival sac, Beilin et al. 
[151] added a fl uorescent marker to the formulations. One minute after the topical 
instillation into eye, 39.9 ± 10.2% of the fl uorescence was measured for the nanosized 
emulsions whereas only 6.8 ± 1.8% for regular eye drops. In addition a study 
was carried out in male albino rabbits to compare the corneal penetration of indomethacin 
from Indocollyre (a marketed hydro - PEG ocular solution) to that of 
negatively and positively charged emulsions [108] . By this comparison, it was 
intended to gain insightful mechanistic comprehension regarding the enhanced 
ocular penetration effect of the emulsion as a function of dosage form and surface 
charge. The contact angle of one droplet of the different dosage forms on the cornea 
was measured and found to be 70 ° for saline, 38 ° for the anionic emulsion, and 21.2 ° 
for the cationic emulsion. Respectively, the values of the calculated spreading coef- 
fi cient were . 47, . 8.6, and . 2.4 mN/m. It can clearly be deduced, owing to the 
marked low spreading coeffi cient values elicited by the emulsions, that both nanosized 
emulsions had better wettability properties on the cornea compared to saline. 
The emulsion may then prolong the residence time of the drop on the epithelial 
layer of the cornea, thereby enabling better drug penetration through the cornea to 
the internal tissues of the eye, as confi rmed by animal studies [108] . It is therefore 
believed that drug is not released from the oil droplet and equilibrates with the 
tears but rather partitions directly from the oil droplets to the cell membranes on 
the eye surface. Therefore, it is reasonable to consider that nanosized emulsions 
have a real advantage since they elicit an increased ocular residence time in com

parison to conventional eye drops and will signifi cantly improve the ocular drug 
bioavailability [152] . This is also confi rmed in numerous cited papers that are listed 
in Table 5 . 
In spite of a relatively rapid removal of conjunctivally absorbed emulsion from 
the eye by local circulation, direct transscleral access to some intraocular tissues 
cannot be excluded, especially if an electrostatic attraction does occur between the 
cationic oil droplets of emulsion and anionic membrane moieties of the sclera, as 
shown by some authors [108] . There is no doubt that the drug absorption from emulsion 
through the noncorneal route needs to be investigated further as it may elicit 
useful information on the potential of nanosized emulsions to promote drug penetration 
to the posterior segment through a mechanism which bypasses the anterior 
chamber. In addition to the above - described protective and elimination mechanisms 
of the eye, nanosized emulsions remaining in the precorneal area may be subject to 
protein binding and to metabolic degradation in the tear fi lm. In conjunction with 
blood plasma, although low, tear fi lm, aqueous humor, and vitreous humor also have 
varying amounts of relatively detectable proteins such as albumin, globulin, and 
immunoglobulins (e.g., IgA, IgG, IgM, IgE) and the enzyme lysozyme. Additional 
studies (at least in vitro) are necessary to understand clearly the nanosized emulsion 
interaction with the ocular fl uid components. Although it is unlikely to happen 
because of the low emulsion volume remaining in the conjunctival sac, the fl uid 
dynamics may be moderately altered by the physical and chemical properties of 
nanosized emulsions, which include tonicity, pH, refractive index, interfacial charge, 
viscosity, osmolality, and irritant ingredients. Thus, formulations of ophthalmic drug 
products must take into account not only the stability and compatibility of a drug in 
the emulsion but also the infl uence of the emulsion on precorneal fl uid dynamics. 
All of the concepts exposed in this section may ultimately result in transcorneal/conjunctival 
absorption of 1 – 2% or less of the drug applied topically through the emulsions. 
In summary, the rate of loss of drug/emulsion from the eye can be 500 – 700 
times greater than the rate of absorption into the eye. Thus, conventional topical 
delivery using emulsions cannot achieve adequate intracellular concentrations of 
drugs or other substances such as oligonucleotides or genes for the treatment of 
endophthalmitis or other sight - threatening intraocular diseases (e.g., AMD). 
TABLE 5 Selected List of o/w Nanosized Emulsions for Ocular Topical Drug Delivery 
Emulsion Type Drug Used Reference 
Anionic emulsion .8 - THC 154 
Pilocarpine base and indomethacin 163 
Adaprolol maleate 161, 162 
Indomethacin 159, 160 
Synthetic HU - 211 and pilocarpine base 124, 155, 156 
Pilocarpine base 157, 158, 164 
Cyclosporin A 168, 165 – 177 
Cationic emulsion Piroxicam 178 
Indomethacin 108 
Miconazole 112 
Cyclosporin A 106, 179 
Note: .8 - THC and synthetic HU - 211 are derivatives of Cannabis sativa. 
MEDICAL APPLICATIONS OF OIL-IN-WATER NANOSIZED EMULSIONS 1349

1350 OIL-IN-WATER NANOSIZED EMULSIONS 
In order to achieve a high concentration of drug within the eye using an emulsion 
delivery vehicle, an approach that bypasses physiological and anatomical barriers 
(e.g., blood – ocular) of the eye is a more viable and attractive option. One such 
approach is to administer emulsion through direct intraocular injections such as 
periocular (subconjunctival and sub - Tenon), intracameral, intravitreal, intracapsular, 
or subretinal. Moreover, it is likely that intraocularly administered emulsion is 
able to both signifi cantly increase drug half - life and minimize intraocular side effects 
that appear following intraocular injection of drug alone. In general, drugs encapsulated 
within emulsion are less toxic than their free counterparts. Additionally, 
there is a possibility of obtaining slow drug release from an intraocularly injected 
emulsion. Taking into account the nonphagocytic character of neural retinal cells 
and the ability of RPE cells to take up large molecules, including oligonucleotides, 
the third - generation emulsion for intravitreal or subretinal injections is more likely 
to be a successful approach in future. Moreover, intravitreally administered drug 
molecules are able to bypass the blood – ocular barrier to achieve constant therapeutic 
levels in the eye while minimizing systemic side effects. However, the hyalocytes, 
the main cellular components of the vitreous, have been classifi ed in at least 
one report [153] as macrophages and thus may play a role in the uptake of intravitreally 
injected emulsion. It should be added that no studies are focused so far on 
injecting emulsion intraocularly and signifi cant work should be devoted to generate 
this novel idea into a fruitful solution in ophthalmic drug delivery applications. 
Over the last decade, o/w nanosized emulsions containing either anionic or cationic 
droplets have been recognized as interesting and promising ocular topical 
delivery vehicles for lipophilic drugs. Complete details are available elsewhere [117] . 
As an overview of this topic, important results on emulsion - based ocular topical 
drug delivery are covered below and are listed in Table 5 . 
The in vivo data obtained from studies of early formulations confi rm that o/w 
nanosized anionic emulsions can be effective topical ophthalmic drug delivery 
systems [154] with a potential for sustained drug release [155] . Naveh and co - 
workers [156] have also noted that the intraocular pressure (IOP) – reducing effect 
of a single, topically administered dose of pilocarpine - loaded anionic emulsion 
lasted for more than 29 h in albino rabbits whereas that of the generic pilocarpine 
lasted only 5 h. Zurowska - Pryczkowska et al. [157] studied how nanosized emulsion 
as a vehicle infl uences the chemical stability of pilocarpine and the effect the drug 
has on the physical stability of nanosized emulsions. In a subsequent paper [158] 
from the same group on in vivo evaluation using normotensive rabbits, it was shown 
that the nanosized emulsion formulated with pilocarpine hydrochloride at pH 5.0 
could be indicated as a preparation offering prolonged pharmacological action 
(miotic effect) together with satisfactory chemical stability. However, the ocular 
bioavailability arising from such a formulation is not signifi cantly improved when 
compared to an aqueous solution. Calvo et al. [159, 160] observed an improvement 
in indomethacin ocular bioavailability when the drug was incorporated in an emulsion 
dosage form with respect to the commercial aqueous drops following topical 
application into rabbit eye. 
In order to verify the extension of the residence time of the emulsion in the 
conjunctival sac, Beilin et al. [151] added a fl uorescent marker to the formulations, 
as mentioned previously. From that observation, it is reasonable to consider that an 
emulsion formulation has the real advantage of increasing ocular residence time in 

comparison to eye drops. Anselem et al. [161] and Melamed et al. [162] prepared a 
nanosized emulsion containing adaprolol maleate, a novel soft . - blocking agent, and 
observed a delayed IOP depressant effect in human volunteers. A similar pharmacological 
effect was also observed in human volunteers by Aviv et al. [163] using 
pilocarpine base - loaded emulsion. Another randomized human trial conducted by 
Garty et al. [164] compared the activity of the pilocarpine base - laden nanosized 
emulsion instilled twice daily with a generic dosage form instilled four times a day 
to 40 hypertensive patients for seven days. No local side effects were observed. The 
IOP decreased 25% in both formulations during this time period. No signifi cant 
difference was noticed between the two treatments. These results proved that the 
anionic emulsion extended the action of the drug and two daily administrations have 
the same result as four instillations of regular eye drops. 
A novel nanosized anionic emulsion incorporating the immunomodulatory agent 
cyclosporin A was developed and the clinical effi cacy was investigated for the treatment 
of moderate to severe dry - eye disease in humans [165 – 167] . The novel cyclosporin 
A ophthalmic dosage form represents a real breakthrough in the formulation 
of a complex, highly lipophilic molecule such as cyclosporin A within an o/w nanosized 
emulsion. Following thorough validation of this formulation through several 
clinical studies in various countries [165 – 175] , an anionic o/w emulsion containing 
cyclosporin A 0.05% (Restasis, Allergan, Irvine, CA) was approved for the fi rst time 
by the FDA, on December 23, 2002. In addition, this anionic emulsion having cyclosporin 
A is now available at pharmacies in the United States for the treatment of 
chronic dry - eye disease (available at www.restasis.com and www.dryeye.com ). Furthermore, 
an over - the - counter (OTC) product that features an emulsion formula, 
Refresh Endura, is already launched in the U.S. market for eye - lubricating purposes 
in patients suffering from moderate to severe dry - eye syndrome. 
The effect of Restasis on contact lens comfort and reducing dry - eye symptoms 
in patients with contact lens intolerance was evaluated in comparison to rewetting 
drops (carboxymethylcellulose 0.5%, Refresh Contacts) [176] . Both formulations 
were applied twice per day before and after lens wear. Symptoms were assessed by 
lens wear time, use of rewetting drops during lens wear, subjective evaluation of 
dryness, and completion of the ocular surface disease index questionnaire. The 
results of this pilot study indicate that cyclosporin 0.05% is benefi cial for contact 
lens wearers with dry eye and reduces contact lens intolerance [176] . Furthermore, 
Sall et al. [177] have recently evaluated the effi cacy of marketed artifi cial tears 
(Systane and Restasis) in relieving the signs and symptoms of dry eye when used 
as supportive therapy to a cyclosporin - based ophthalmic emulsion (i.e., Restasis + 
Systane vs. Restasis + Refresh). Signifi cant differences were seen in favor of Restasis 
+ Systane versus Restasis + Refresh for less ocular burning, stinging, grittiness, and 
dryness. Systane was better than Restasis + Refresh for less burning, dryness, and 
scratchiness. Results indicate that the choice of concomitant therapy used with 
Restasis has signifi cant effects on outcome measures and both supportive therapies 
were compatible with Restasis [177] . 
When compared to either saline or anionic emulsions, the nanosized cationic 
emulsions were shown to enhance the ocular bioavailability of indomethacin [108] , 
piroxicam [178] , and cyclosporin A [106, 179] following one single - drop dose instillation 
into the rabbit eye (Figure 4 ). A signifi cant drug reservoir effect was noted 
in the cornea and conjunctiva even for more than 8 h following the instillation [106] . 
MEDICAL APPLICATIONS OF OIL-IN-WATER NANOSIZED EMULSIONS 1351

1352 OIL-IN-WATER NANOSIZED EMULSIONS 
This long residence time may help reduce evaporation of the limited volume of 
natural tears present in patients with dry eye. This was probably due to the adhesion 
of the positively charged oil droplets to the negatively charged corneal surface 
moieties as a result of electrostatic attraction. This hypothesis was supported by data 
from an ex vivo study which showed that cationic emulsion exhibited better wettability 
properties on albino rabbit eye cornea than either saline or anionic emulsion 
[108] . Associated with Poloxamer and phospholipids, a cationic primary amine, 
stearylamine, has been used to obtain the above - described third - generation cationic 
emulsions. Additionally, a cationic emulsion based on an association of Poloxamer 
188 and chitosan was prepared and also showed interesting physicochemical properties 
on stability and charge effects [97, 98] . Moreover, the stability and ocular tolerance 
following topical instillation into the eye of these cationic emulsion vehicles 
were investigated [98, 114] . The overall studies hence stress the effectiveness of 
nanosized cationic emulsion, which promotes ocular drug absorption via internalization 
possibly through an endocytic process [112] . 
7.4.5.3 Nasal Route 
The nasal route is still receiving great attention due to a number of advantages over 
parenteral and oral administration [180] , particularly when fi rst - pass metabolism 
makes the drug ineffective. The approach of an o/w emulsion formulation of the 
drug may increase absorption by solubilizing the drug in the inner phase of 
the emulsion and prolonging contact time between emulsion droplets and nasal 
mucosa. 
One of the fi rst examples for systemic delivery of peptides concerned a lipid - 
soluble rennin inhibitor [181] . The peptide was solubilized in the oil phase of an o/w 
emulsion containing membrane adjuvants such as oleic acid and mono - and diglyc- 
FIGURE 4 Infl uence of emulsion surface charges cyclosporin A (CsA) concentrations in 
ocular surface tissues (cornea and conjunctiva) following one single - dose (50 - . L) instillation 
of positively (cationic) and negatively (anionic) charged CsA - loaded nanosized emulsions 
into albino rabbit eye. 
0 
400 
800 
1200 
1600 
2000 
2400 
2800 
15 30 60 120 180 480 
Time (min) 
Cornea positive emulsion 
Cornea negative emulsion 
Conjunctiva positive emulsion 
Conjunctiva negative emulsion 
CsA concentration, ng/g

erides. Emulsion formulations have been proposed to simultaneously increase the 
solubility of the peptide and to enhance membrane permeability through interaction 
between the membrane and the oil components. Enhanced and prolonged in 
vivo nasal absorption of the rennin inhibitor was observed in emulsion formulation 
compared to aqueous suspension. From morphological studies, the emulsions did 
not provoke any signifi cant changes to the nasal mucosa [181] . Such a formulation 
approach was also used for the administration of a steroidal male sex hormone testosterone 
[182] . The steroid was solubilized in the oil phase of the o/w emulsion and 
the ionic composition of the aqueous phase was modifi ed in order to produce electrically 
positive, negative, and neutral droplets. Droplets with a surface charge led 
to better bioavailability than neutral droplets, but contrary to the above - described 
topical applicabilities of cationic emulsions over anionic emulsions, positively 
charged droplets did not provide the best results [182] . However, the emulsion 
approach was advantageous since it helped to overcome the solubility problem of 
the hydrophobic compounds. 
In another study which does not involve peptide drugs, various emulsion formulations 
were prepared in order to modulate the partitioning of the drug between the 
aqueous phase and the oil phase [183] . The disappearance of a drug from the nasal 
cavity was determined by an in situ perfusion technique. When the drug was solubilized 
in the aqueous phase, the formulation did not have a signifi cant effect on 
the drug disappearance rate. However, partitioning of the drug in the oil phase 
resulted in delaying absorption. It was suggested that oil droplets containing 
medium - chain triglycerides formed a pseudo – oily layer on the mucous membrane, 
which slowed down the drug disappearance from the nasal cavity [183] . Another 
interesting study reported nasal delivery of insulin formulated in both o/w and w/o 
emulsions [184] . As insulin partitions into the aqueous phase of the emulsion, the 
peptide is either incorporated within the continuous phase of the o/w emulsion or 
encapsulated in the aqueous droplets of the w/o emulsion. Following nasal perfusion 
experiments, plasma insulin concentration profi les showed enhanced insulin absorption 
when the peptide was formulated as an o/w emulsion compared to an aqueous 
solution. However, a w/o emulsion did not cause any signifi cant increase in plasma 
insulin concentration. Delivery of insulin by administration of nasal drops also 
revealed a large dose - dependent increase in plasma insulin concentration. It also 
needs to be pointed out that the emulsifi er mixture alone did not promote any 
absorption. It was suggested that insulin molecules probably were adsorbed at the 
surface of the oil droplets. Adhesion of the oil droplets on the mucosal membrane 
then induced a local increase in insulin concentration at the membrane surface. 
However, the number of droplets in contact with the surface had to be small enough. 
Otherwise, a stagnant oil layer is formed which acts as an additional barrier to the 
transport, as was observed with the w/o emulsion [183] . 
Other recent applications of emulsion formulation involve mucosal gene and 
vaccine delivery [185 – 187] and the preparation of polymeric microspheres by the 
w/o emulsifi cation solvent extraction technique [188] . 
7.4.5.4 Topical Route 
Many drugs exhibit low skin penetration, which results in poor effi cacy. As opposed 
to common chemical skin penetration enhancers, organic solvents, which are 
MEDICAL APPLICATIONS OF OIL-IN-WATER NANOSIZED EMULSIONS 1353

1354 OIL-IN-WATER NANOSIZED EMULSIONS 
generally associated to some degree with skin irritation, toxicity and sensitization, 
a solvent - free topical vehicle based on drug entrapment in o/w emulsion droplets 
of submicrometer size is more effi cacious in terms of percutaneous absorption and 
possibly devoid of adverse effects. In addition, the uniqueness of the large internal 
hydrophobic oil core of o/w nanosized emulsion droplets allows high solubilization 
capacity for water - insoluble topically active medicaments and also aids in carrying 
water, an excellent softener, to the skin. 
The concept of using anionic nanosized emulsion vehicles for enhanced percutaneous 
absorption of nonsteroidal anti - infl ammatory drugs (NSAIDs) and diazepam 
was clearly proven [189, 190] . NSAIDs and diazepam in a nanosized emulsion 
vehicle also demonstrated noticeable systemic activity. The o/w emulsion was tested 
for primary irritation in humans in a 48 - h trial. Low irritancy and excellent human 
acceptance were observed, subsequently making the further development of a nanosized 
emulsion vehicle very attractive. 
Even though emulsion vehicles increase dermal drug delivery of lipophilic drugs 
in humans, one of the problems for topical drug delivery is the diffi culty of applying 
these vehicles to the skin because of their fl uidity. Rheological properties are studied 
in transdermal formulations and different results are given. Realdaon and Ragazzi 
[191] have investigated different mechanical emulsifying conditions on o/w emulsion 
formulations containing methyl nicotinate. The infl uence of these procedures 
on rheological properties and in vivo availability of methyl nicotinate was evaluated. 
Even if various viscosities were obtained, differences between batches did not compromise 
drug availability. On the contrary, Welin - Berger and co - workers [192, 193] 
concluded in their study on nanosized emulsions containing model compounds that 
both release and permeation rates decrease when the apparent yield stress (i.e., the 
macroviscosity) increases by addition of gelling polymers. Because a topical anesthetic 
agent will induce a pain - suppressing anesthesia, the eutectic mixture of local 
anesthetics (EMLA) has proven to be a useful medication for children. It is an 
emulsion containing a mixture of lidocaine and prilocaine. This cream gives an effective 
deep sedation and can be applied half an hour to 1 h prior to the procedure. 
Local side effects with this emulsion are very mild [194, 195] . Systemic activity of 
nanosized emulsions containing diazepam was compared with regular creams or 
ointments by Schwarz et al. [190] . Their effi ciency was tested on protection against 
pentamethylenetetrazole, which induces convulsive effects in mice. Diazepam 
applied topically in emulsion creams was strongly dependent on oil droplet size. 
Furthermore, nanosized emulsions increased transdermal drug delivery and prolonged 
protective activity for up to 6 h. 
Many formulations for topical emulsions are available in the scientifi c literature, 
in patents, and on the market. Progresses made in the last years in this fi eld are 
concentrated on the various aspects of drug release and the infl uence of droplet 
size. 
Third - generation cationic emulsions were suggested as drug carriers for topical 
use in the skin. It was found that . - tocopherol - loaded cationic emulsion was able 
to prevent oxidative damage of cultured fi broblast cells [196] . In addition, the same 
cationic formulation was able to protect rat skin against oxidative stress induced by 
ultraviolet (UV) irradiation signifi cantly better than either the corresponding 
anionic emulsion or the cationic blank emulsion, as measured with a noninvasive 

evaluation of the lipid hydroperoxidation process of the rat skin. However, no difference 
was found between cationic or anionic nanosized emulsions of . - tocopherol 
as assessed by endogenous peroxyl radical scavenging ability. Taken together, these 
results suggest that the cationic emulsion allows the . - tocopherol to remain on the 
surface of the skin because of electrostatic interactions between the negatively 
charged sites of the superfi cial layers of the skin and the positively charged oil 
droplets. Although the extent of . - tocopherol incorporation into the skin is similar 
for both cationic and anionic emulsions, the prolonged skin surface residence time 
of the cationic emulsion allows an enhanced protective effect against oxidative 
stress. In contrast to these results, an in vitro percutaneous absorption study on 
hairless rat skin found that the antifungal drugs econazole and miconazole nitrate 
incorporated into a similar cationic emulsion formulation were more effective 
in terms of skin penetration than the corresponding anionic emulsion [197] . 
The enhanced rate of diffusion of these antifungal drugs through the skin by the 
cationic emulsion suggests a new approach for dermal drug penetration 
enhancement [197] . 
7.4.6 FUTURE PERSPECTIVE 
Based on the performances in previous and present decades, o/w - type nanosized 
emulsions can conveniently be classifi ed into three generations. First - generation 
emulsions are considered primarily as nutrient carriers to be administered via 
intravenous routes to bed - ridden patients. Second - generation emulsions start initially 
as drug carrier systems by solubilizing considerable amounts of lipophilic 
drugs at the oil phase or at the oil – water interface of the emulsion. This particular 
merit of emulsions is specifi cally exploited even commercially for both ocular and 
parenteral active drugs. Modifi cations made either in the oil phase or at the o/w 
interfacial fi lm forming emulsifi er molecules allow the emulsions to be able to 
escape from lipolysis by lipoprotein lipase, apo adsorption, and liver uptake. Such 
a surface - modifi ed emulsion would prolong the circulation time in plasma and 
thereby an alteration in in vivo disposition of incorporated drugs following parenteral 
administration. Attachment of homing devices such as antibody and apoE to 
the surfaces of emulsions makes the selective/active delivering of emulsion - 
incorporated drugs to target sites such as a tumorized organ or hepatic system. 
Active targeting increases the affi nity of the carrier system for the target site, while 
passive targeting minimizes the nonspecifi c interaction with nontargeted sites by 
the RES. Having together a positive charge and a steric stabilizing effect led to 
the development of third - generation emulsions that contain a unique property: 
plasma half - life prolongation and electrostatic adhesion to ocular surface tissues 
after topical instillation into eye. Furthermore, the third - generation emulsion shows 
a potential of carrying a wide range of lipophilic, amphiphilic, and polyanionic 
compounds, including DNA and oligonucleotides for transdermal and nasal routes. 
Accumulating knowledge thus suggests that constant progress in better understanding 
the principles and processes governing the various issues related to o/w 
nanosized emulsions has surely brought major improvements in the effi cacy of 
parenteral or nonparenteral drug delivery systems. 
FUTURE PERSPECTIVE 1355

1356 OIL-IN-WATER NANOSIZED EMULSIONS 
REFERENCES 
1. Lipinski , C. A , Lombardo , F. , Dominy , B. W. , and Feeney , P. J. ( 2001 ), Experimental and 
computational approaches to estimate solubility and permeability in drug discovery and 
development settings , Adv. Drug Deliv. Rev. , 46 , 3 – 26 . 
2. Lipinski , C. ( 2002 ), Poor aqueous solubility — An industry wide problem in drug discovery 
, Am. Pharm. Rev. , 5 , 82 – 85 . 
3. El - Aasser , M. S. , and Sudol , E. D. ( 2004 ), Miniemulsions: Overview of research and 
applications , JCT Res. , 1 , 21 – 31 . 
4. Nakajima , H. ( 1997 ), Microemulsions in cosmetics , in Solans , C. , and Kunieda , H. , Eds., 
Industrial Applications of Microemulsions , Marcel Dekker , New York , pp. 175 – 197 . 
5. Benita , S. ( 1998 ), Introduction and overview , in Benita , S. , Ed., Submicron Emulsion in 
Drug Targeting & Delivery , Harwood Academic , The Netherlands , pp. 1 – 5 . 
6. Klang , S. H. , Benita , S. ( 1998 ), Design and evaluation of submicron emulsions as colloidal 
drug carriers for intravenous administration , in Benita , S. , Ed., Submicron Emulsion in 
Drug Targeting & Delivery , Harwood Academic , The Netherlands , pp. 119 – 152 . 
7. Tadros , T. F. , Izquierdo , P. , Esquena , J. , and Solans , C. ( 2004 ), Formation and stability of 
nanoemulsions , Adv. Coll. Interf. Sci. , 108 – 109 , 303 – 318 . 
8. Solans , C. , Izquierdo , P. , Nolla , J. , Azemar , N. , and Garcia - Celma , M. J. ( 2005 ), Nanoemulsions 
, Curr. Opin. Coll. Interf. Sci. , 10 , 102 – 110 . 
9. Becher , P. , and Schick , M. J. ( 1987 ), Macroemulsions , in Schick , M. J. , Ed., Non - Ionic 
Surfactants , Surfactant Science Series Vol. 23., Marcel Dekker , Basel , pp. 435 – 493 . 
10. Kabalnov , A. ( 1998 ), Thermodynamic and theoretical aspects of emulsions and their 
stability , Curr. Opin. Coll. Interf. Sci. , 3 , 270 – 275 . 
11. Stefan , A. , Palazzo , G. , Ceglie , A. , Panzavolta , E. , and Hochkoeppler ,A. ( 2003 ), Water - 
in - oil macroemulsions sustain long - term viability of microbial cells in organic solvents , 
Biotechnol. Bioeng. , 81 , 323 – 328 . 
12. Ceglie , A. , Das , K. P. , and Lindman , B. ( 1987 ), Microemulsion structure in four component 
systems for different surfactants , Coll. Surf. , 28 , 29 – 40 . 
13. Attwood , D. ( 1994 ), Microemulsions , in Kreuter , J. , Ed., Colloidal Drug Delivery Systems , 
Marcel Dekker , New York , pp. 31 – 71 . 
14. Hino , T. , Kawashima , Y. , and Shimabayashi , S. ( 2000 ), Basic study for stabilization of 
w/o/w emulsion and its application to transcatheter arterial embolization therapy , 
Adv. Drug Deliv. Rev. , 45 , 27 – 45 . 
15. Gallarate , M. , Carlotti , M. E. , Trotta , M. , and Bovo , S. ( 1999 ), On the stability of 
ascorbic acid in emulsifi ed systems for topical and cosmetic use , Int. J. Pharm. , 188 , 
233 – 241 . 
16. Er A fnofas , I. , Csoka , I. , Csany , E. , Orosz , K. , and Makai , M. ( 1998 ), in Proceedings of 
2nd World Meeting APGI/APV , Paris, pp. 805 – 806 . 
17. Lowe , K. C. ( 1999 ), Perfl uorinated blood substitutes and artifi cial oxygen carriers , Blood 
Rev. , 13 , 171 – 184 . 
18. Spahn , D. R. ( 2000 ), Current status of artifi cial oxygen carriers , Adv. Drug Deliv. Rev. , 
40 , 143 – 151 . 
19. Krafft , M. P. ( 2001 ), Fluorocarbons and fl uorinated amphiphiles in drug delivery and 
biomedical research , Adv. Drug Deliv. Rev. , 47 , 209 – 228 . 
20. Fukushima , S. , Kishimoto , S. , Takeuchi , Y. , and Fukushima , M. ( 2000 ), Preparation and 
evaluation of o/w type emulsions containing antitumor prostaglandin , Adv. Drug Deliv. 
Rev. , 45 , 65 – 75 . 

21. Ott , G. , Singh , M. , Kazzaz , J. , Briones , M. , Soenawan , E. , Ugozzoli , M. , and O ’ Hagan , 
D. T. ( 2002 ), A cationic sub - micron emulsion (MF59/DOTAP) is an effective delivery 
system for DNA vaccines , J. Controlled Release , 79 , 1 – 5 . 
22. Ribi , E. E. ( 1984 ), Refi ned detoxifi ed endotoxin product, U.S. Patent 4,436,727 , 
March 13. 
23. Ribi , E. , Schwartzman , S. M. , and Cantrell , J. L. Refi ned detoxifi ed endotoxin product, 
( 1984 ), U.S. Patent 4,436,728 , March 13. 
24. Myers , K. R. , and Truchot , A. T. ( 1990 ), Modifi ed lipopolysaccharides and process of 
preparation, U.S. Patent 4,912,094 , March 27. 
25. Allison , A. C. , and Byars , N. E. ( 1986 ), An adjuvant formulation that selectively elicits 
the formation of antibodies of protective isotypes and of cell - mediated immunity , 
J. Immunol. Methods , 95 , 157 – 168 . 
26. Chandra , R. K. ( 1985 ), Grace A. Goldsmith Award lecture. Trace element regulation of 
immunity and infection , J. Am. Coll. Nutr. , 4 , 5 – 16 . 
27. Shils , M. E. ( 1998 ), Parentral nutrition , in Shils , M. E. , Olson , J. A. , Shike , M. E. , and 
Ross , O. , Eds., Modern Nutr. in Health and Disease , 9th ed., Williams & Wilkins , 
Baltimore, PA , pp. 415 – 428 . 
28. Koster , V. S. , Kuks , P. F. M., Langer , R. , and Talsma , H. ( 1996 ), Particle size in parenteral 
fat emulsions, what are the true limitations ? Int. J. Pharm. , 134 , 235 – 238 . 
29. Lutz , O. , Meraihi , Z. , Mura , J. L. , Frey , A. , Riess , G. H. , and Bach . A. C. ( 1989 ), Fat emulsion 
particle size: Infl uence on the clearance rate and the tissue lipolytic activity , Am. J. 
Clin. Nutr. , 50 , 1370 – 1381 . 
30. Rubin , M. , Harell , D. , Naor , N. , Moser , A. , Wielunsky , E. , Merlob , P. , and Lichtenberg , 
D. ( 1991 ), Lipid infusion with different triglyceride cores (long - chain vs medium - chain/ 
long - chain triglycerides): Effect on plasma lipids and bilirubin binding in premature 
infants , J. Parenteral Enteral Nutr. , 15 , 642 – 646 . 
31. Mizushima , Y. , Hoshi , K. , Aihara , H. , and Kurachi , M. ( 1983 ), Inhibition of bronchoconstriction 
by aerosol of a lipid containing prostaglandin E1 , J. Pharm. Pharmacol. , 35 , 
397 . 
32. Craig , D. Q. M. , Patel , M. , and Ashford , M. ( 2000 ), Administration of emulsions to the 
gastrointestinal tract , in Nielloud , F. , and Marti - Mestres , G. , Eds., Pharmaceutical Emulsions 
and Suspensions , Marcel Dekker , New York , pp. 323 – 360 . 
33. Hauss , D. J. ( 2002 ), Oral lipid - based drug delivery — a case of implementation failing to 
keep up with innovation? Am. Pharm. Rev. , 5 , 22 – 36 . 
34. Sakaeda , T. , and Hirano , K. ( 1998 ), Effect of composition on biological fate of oil particles 
after intravenous injection of O/W lipid emulsions , J. Drug Target. , 6 , 273 – 284 . 
35. Sasaki , H. , Yamamura , K. , Nishida , K. , Nakamura , J. , and Ichikawa , M. ( 1996 ), Delivery 
of drugs to the eye by topical application , Prog. Ret. Eye Res. , 15 , 583 – 620 . 
36. Harnisch , S. , and M u ller , R. H. ( 1998 ), Plasma protein adsorption patterns on emulsions 
for parenteral administration: Establishment of a protocol for two - dimensional polyacrylamide 
electrophoresis , Electrophoresis , 19 , 349 – 354 . 
37. Harnisch , S. , and M u ller , R. H. ( 2000 ), Adsorption kinetics of plasma proteins on oil - in - 
water emulsions for parenteral nutrition , Eur. J. Pharm. Biopharm. , 49 , 41 – 46 . 
38. Szebeni , J. ( 2001 ), Complement activation - related pseudoallergy caused by liposomes, 
micellar carriers of intravenous drugs, and radiocontrast agents , Crit. Rev. Ther. Drug 
Carrier Syst. , 18 , 567 – 606 . 
39. Price , M. E. , Cornelius , R. M. , and Brash , J. L. ( 2001 ), Protein adsorption to polyethylene 
glycol modifi ed liposomes from fi brinogen solution and from plasma , Biochim. Biophys. 
Acta , 1512 , 191 – 205 . 
REFERENCES 1357

1358 OIL-IN-WATER NANOSIZED EMULSIONS 
40. Deckelbaum , R. J. , Hamilton , J. A. , Moser , A. , Bengtsson - Olivecrona , G. , Butbul , E. , 
Carpentier , Y. A. , Gutman , A. , and Olivecrona , T. ( 1990 ), Medium - chain versus long - 
chain triacylglycerol emulsion hydrolysis by lipoprotein lipase and hepatic lipase: Implications 
for the mechanisms of lipase action , Biochemistry , 29 , 1136 – 1142 . 
41. Hedeman , H. , Br o ndsted , H. , M u llertz , A. , and Frokjaer , S. ( 1996 ), Fat emulsions based 
on structured lipids (1,3 - specifi c triglycerides): An investigation of the in vivo fate , 
Pharm. Res. , 13 , 725 – 728 . 
42. Takino , T. , Konishi , K. , Takakura , Y. , and Hashida , M. ( 1994 ), Long circulating emulsion 
carrier systems for highly lipophilic drugs , Biol. Pharm. Bull. , 17 , 121 – 125 . 
43. Takino , T. , Nakajima , C. , Takakura , Y. , Sezaki , H. , and Hashida , M. ( 1993 ), Controlled 
biodistribution of highly lipophilic drugs with various parenteral formulations , J. Drug 
Target. , 1 , 117 – 124 . 
44. Redgrave , T. G. , Rakic , V. , Mortimer , B. - C. , and Mamo , J. C. L. ( 1992 ), Effects of sphingomyelin 
and phosphatidylcholine acyl chains on the clearance of triacylglycerol - rich 
lipoproteins from plasma. Studies with lipid emulsions in rats , Biochim. Biophys. Acta , 
1126 , 65 – 72 . 
45. Arimoto , I. , Matsumoto , C. , Tanaka , M. , Okuhira , K. , Saito , H. , and Handa , T. ( 1998 ), 
Surface composition regulates clearance from plasma and triolein lipolysis of lipid emulsions 
, Lipids , 33 , 773 – 779 . 
46. Handa , T. , Eguchi , Y. , and Miyajima , K. ( 1994 ), Effects of cholesterol and cholesteryl 
oleate on lipolysis and liver uptake of triglyceride/phosphatidylcholine emulsions in rats , 
Pharm. Res. , 11 , 1283 – 1287 . 
47. Kurihara , A. , Shibayama , Y. , Mizota , A. , Yasuno , A. , Ikeda , M. , and Hisaoka , M. ( 1996 ), 
Pharmacokinetics of highly lipophilic antitumor agent palmitoyl rhizoxin incorporated 
in lipid emulsions in rats , Biol. Pharm. Bull. , 19 , 252 – 258 . 
48. Kurihara , A. , Shibayama , Y. , Yasuno , A. , Ikeda , M. , and Hisaoka , M. ( 1996 ), Lipid emulsions 
of palmitoylrhizoxin: Effects of particle size on blood dispositions of emulsion lipid 
and incorporated compound in rats , Biopharm. Drug Dispos. , 17 , 343 – 353 . 
49. Lin , S. Y. , Wu , W. H. , and Lui , W. Y. ( 1992 ), In vitro release, pharmacokinetic and tissue 
distribution studies of doxorubicin hydrochloride (Adriamycin HCl) encapsulated in 
lipiodolized w/o emulsions and w/o/w multiple emulsions , Die Pharmazie , 47 , 439 – 443 . 
50. Sakaeda , T. , Takahashi , K. , Nishihara , Y. , and Hirano , K. ( 1994 ), O/W lipid emulsions for 
parenteral drug delivery. I. Pharmacokinetics of the oil particles and incorporated 
sudan II , Biol. Pharm. Bull. , 17 , 1490 – 1495 . 
51. Yamaguchi , T. , Nishizaki , K. , Itai , S. , Hayashi , H. , and Ohshima , H. ( 1995 ), Physicochemical 
characterization of parenteral lipid emulsion: Infl uence of cosurfactants on fl occulation 
and coalescence , Pharm. Res. , 12 , 1273 – 1278 . 
52. Ueda , K. , Yamazaki , Y. , Noto , H. , Teshima , Y. , Yamashita , C. , Sakaeda , T. , and Iwakawa , 
S. ( 2003 ), Effect of oxyethylene moieties in hydrogenated castor oil on the pharmacokinetics 
of menatetrenone incorporated in O/W lipid emulsions prepared with hydrogenated 
castor oil and soybean oil in rats , J. Drug Target. , 11 , 37 – 43 . 
53. Liu , F. , and Liu , D. ( 1995 ), Long - circulating emulsions (oil - in - water) as carriers for lipophilic 
drugs , Pharm. Res. , 12 , 1060 – 1064 . 
54. Lee , J. H. , Lee , H. B. , and Andrade , J. D. ( 1995 ), Blood compatibility of polyethylene 
oxide surfaces , Prog. Polym. Sci. , 20 , 1043 – 1079 . 
55. Davis , S. S. , and Hansrani , P. ( 1985 ), The infl uence of emulsifying agents on the phagocytosis 
of lipid emulsions by macrophages , Int. J. Pharm. , 23 , 69 – 77 . 
56. Levy , M. Y. , Benita , S. , and Baszkin , A. ( 1991 ), Interactions of a non - ionic surfactant with 
mixed phospholipid - oleic acid monolayers. Studies under dynamic conditions , Coll. 
Surf. , 59 , 225 – 241 . 

57. Harris , J. M. , Martin , N. E. , and Modi , M. ( 2001 ), Pegylation: A novel process for modifying 
pharmacokinetics , Clin. Pharmacokinet. , 40 , 539 – 551 . 
58. Bhadra , D. , Bhadra , S. , Jain , P. , and Jain , N. K. ( 2002 ), Pegnology: A review of PEG - ylated 
systems , Die Pharm. , 57 , 5 – 29 . 
59. Klibanov , A. L. , Maruyama , K. , Torchilin , V. P. , and Huang , L. ( 1990 ), Amphipathic polyethyleneglycols 
effectively prolong the circulation time of liposomes , FEBS Lett. , 268 , 
235 – 237 . 
60. Allen , T. M. , Hansen , C. , Martin , F. , Redemann , C. , and Yau - Young , A. ( 1991 ), Liposomes 
containing synthetic lipid derivatives of poly(ethylene glycol) show prolonged circulation 
half - lives in vivo , Biochim. Biophys. Acta , 1066 , 29 – 36 . 
61. Woodle , M. C. , and Lasic , D. D. ( 1992 ), Sterically stabilized liposomes , Biochim. Biophys. 
Acta , 1113 , 171 – 199 . 
62. Dunn , S. E. , Brindley , A. , Davis , S. S. , Davies , M. C. , and Illum , L. ( 1994 ), Polystyrene - poly 
(ethylene glycol) (PS - PEG2000) particles as model systems for site specifi c drug delivery. 
2. The effect of PEG surface density on the in vitro cell interaction and in vivo biodistribution 
, Pharm. Res. , 11 , 1016 – 1022 . 
63. Gref , R. , Minamitake , Y. , Peracchia , M. T. , Trubetskoy , V. , Torchilin , V. , and Langer , R. 
( 1994 ), Biodegradable long - circulating polymeric nanospheres , Science , 263 , 1600 – 
1603 . 
64. Wheeler , J. J. , Wong , K. F. , Ansell , S. M. , Masin , D. , and Bally , M. B. ( 1994 ), Polyethylene 
glycol modifi ed phospholipids stabilize emulsions prepared from triacylglycerol , 
J. Pharm. Sci. , 83 , 1558 – 1564 . 
65. Tirosh , O. , Barenholz , Y. , Katzhendler , J. , and Priev , A. ( 1998 ), Hydration of polyethylene 
glycol - grafted liposomes , Biophys. J. , 74 , 1371 – 1379 . 
66. Papahadjopoulos , D. , Allen , T. M. , Gabizon , A. , Mayhew , E. , Matthay , K. , Huang , S. K. , 
Lee , K. - D. , Woodle , M. C. , Lasic , D. D. , Redemann , C. , and Martin , F. J. ( 1991 ), Sterically 
stabilized liposomes: Improvements in pharmacokinetics and antitumor therapeutic 
effi cacy , Proc. Nat. Acad. Sci. U. S. A. , 88 , 11460 – 11464 . 
67. Lundberg , B. B. , Mortimer , B. - C. , and Redgrave , T. G. ( 1996 ), Submicron lipid emulsions 
containing amphipathic polyethylene glycol for use as drug - carriers with prolonged circulation 
time , Int. J. Pharm. , 134 , 119 – 127 . 
68. Allen , T. M. , and Everest , J. M. ( 1983 ), Effect of liposome size and drug release properties 
on pharmacokinetics of encapsulated drug in rats , J. Pharmacol. Exp. Ther. , 226 , 
539 – 544 . 
69. Senior , J. , Crawley , J. C. W. , and Gregoriadis , G. ( 1985 ), Tissue distribution of liposomes 
exhibiting long half - lives in the circulation after intravenous injection , Biochim. Biophys. 
Acta , 839 , 1 – 8 . 
70. Liu , D. , Mori , A. , and Huang , L. ( 1991 ), Large liposomes containing ganglioside GM1 
accumulate effectively in spleen , Biochim. Biophys. Acta , 1066 , 159 – 165 . 
71. Connelly , P. W. , and Kuksis , A. ( 1981 ), Effect of core composition and particle size of 
lipid emulsions on apolipoprotein transfer of plasma lipoproteins in vivo , Biochim. 
Biophys. Acta , 666 , 80 – 89 . 
72. Bundgaard , M. ( 1980 ), Transport pathways in capillaries — In search of pores , Annu. Rev. 
Physiol. , 42 , 325 – 336 . 
73. Wisse , E. ( 1970 ), An electron microscopic study of the fenestrated endothelial lining of 
rat liver sinusoids , J. Ultrastruct. Res. , 31 , 125 – 150 . 
74. Jain , R. K. ( 1987 ), Transport of molecules across tumor vasculature , Cancer Metastasis 
Rev. , 6 , 559 – 593 . 
75. Takakura , Y. , and Hashida , M. ( 1995 ), Macromolecular drug carrier systems in 
cancer chemotherapy: Macromolecular prodrugs , Crit. Rev. Oncol./Hematol. , 18 , 
207 – 231 . 
REFERENCES 1359

1360 OIL-IN-WATER NANOSIZED EMULSIONS 
76. Gabizon , A. , Price , D. C. , Huberty , J. , Bresalier , R. S. , and Papahadjopoulos , D. ( 1990 ), 
Effect of liposome composition and other factors on the targeting of liposomes to 
experimental tumors: Biodistribution and imaging studies , Cancer Res. , 50 , 6371 – 6378 . 
77. Wu , N. Z. , Da , D. , Rudoll , T. L. , Needham , D. , Whorton , A. R. , and Dewhirst , M. W. 
( 1993 ), Increased microvascular permeability contributes to preferential accumulation 
of Stealth liposomes in tumor tissue , Cancer Res. , 53 , 3765 – 3770 . 
78. Yuan , F. , Dellian , M. , Fukumura , D. , Leunig , M. , Berk , D. A. , Torchilin , V. P. , and Jain , 
R. K. ( 1995 ), Vascular permeability in a human tumor xenograft: Molecular size 
dependence and cutoff size , Cancer Res. , 55 , 3752 – 3756 . 
79. Nomura , T. , Yamashita , F. , Takakura , Y. , and Hashida , M. ( 1995 ), Proc. Int. Symp. Control 
Release Bioactive Mater. , 22 , 420 – 421 . 
80. Yuan , F. , Leunig , M. , Huang , S. K. , Berk , D. A. , Papahadjopoulos , D. , and Jain , R. K. 
( 1994 ), Microvascular permeability and interstitial penetration of sterically stabilized 
(stealth) liposomes in a human tumor xenograft , Cancer Res. , 54 , 3352 – 3356 . 
81. Gabizon , A. , Shmeeda , H. , Horowitz , A. T. , and Zalipsky , S. ( 2004 ), Tumor cell targeting 
of liposome - entrapped drugs with phospholipid - anchored folic acid - PEG conjugates , 
Adv. Drug Deliv. Rev. , 56 , 1177 – 1192 . 
82. Nobs , L. , Buchegger , F. , Gurny , R. , and Allemann , E. ( 2004 ), Current methods for 
attaching targeting ligands to liposomes and nanoparticles , J. Pharm. Sci. , 93 , 1980 – 
1992 . 
83. Song , Y. K. , Liu , D. , Maruyama , K. Z. , and Takizawa , T. ( 1996 ), Antibody mediated lung 
targeting of long - circulating emulsions , PDA J. Pharm. Sci. Technol. , 50 , 372 – 377 . 
84. Lundberg , B. B. , Griffi ths , G. , and Hansen , H. J. ( 1999 ), Conjugation of an anti - B - cell 
lymphoma monoclonal antibody, LL2, to longcirculating drug - carrier lipid emulsions , 
J. Pharm. Pharmacol. , 51 , 1099 – 1105 . 
85. Lundberg , B. B. , Griffi ths , G. , and Hansen , H. J. ( 2004 ), Cellular association and cytotoxicity 
of anti - CD74 - targeted lipid drug - carriers in B lymphoma cells , J. Controlled Release , 
94 , 155 – 161 . 
86. Goldstein , D. , Nassar , T. , Lambert , G. , Kadouche , J. , and Benita , S. ( 2005 ), The design 
and evaluation of a novel targeted drug delivery system using cationic emulsion - 
antibody conjugates , J. Controlled Release , 108 , 418 – 432 . 
87. Rensen , P. C. N. , Van Dijk , M. C. M , Havenaar , E. C. , Bijsterbosch , M. K. , Kruijt , J. K. , 
and Van Berkel , T. J. C. ( 1995 ), Selective liver targeting of antivirals by recombinant 
chylomicrons — A new therapeutic approach to hepatitis B , Nat. Med. , 1 , 221 – 225 . 
88. Mahley , R. W. ( 1988 ), Apolipoprotein E: Cholesterol transport protein with expanding 
role in cell biology , Science , 240 , 622 – 630 . 
89. Barenholz , Y. , and Cohen , R. ( 1995 ), Rational design of amphiphile - based drug carriers 
and sterically stabilized carriers , J. Liposome Res. , 5 , 905 – 932 . 
90. Iwamoto , K. , Kato , T. , Kawahara , M. , Koyama , N. , Watanabe , S. , Miyake , Y. , and 
Sunamoto , J. ( 1991 ), Polysaccharide - coated oil droplets in oil - in - water emulsions as 
targetable carriers for lipophilic drugs , J. Pharm. Sci. , 80 , 219 – 224 . 
91. Grolier , P. , Azais - Braesco , V. , Zelmire , L. , and Fessi , H. ( 1992 ), Incorporation of carotenoids 
in aqueous systems: Uptake by cultured rat hepatocytes , Biochim. Biophys. Acta , 
1111 , 135 – 138 . 
92. Barratt , G. ( 2003 ), Colloidal drug carriers: Achievements and perspectives , Cell. Mol. 
Life Sci. , 60 , 21 – 37 . 
93. Kim , T. W. , Chung , H. , Kwon , I. C. , Sung , H. C. , and Jeong , S. Y. ( 2001 ), Optimization of 
lipid composition in cationic emulsion as in vitro and in vivo transfection agents , Pharm. 
Res. , 18 , 54 – 60 . 

94. Kim , Y. J. , Kim , T. W. , Chung , H. , Kwon , I. C. , Sung , H. C. , and Jeong , S. Y. ( 2001 ), Counterion 
effects on transfection activity of cationic lipid emulsion , Biotechnol. Bioproc. 
Eng. , 6 , 279 – 283 . 
95. Elbaz , E. , Zeevi , A. , Klang , S. , and Benita , S. ( 1993 ), Positively - charged submicron emulsion 
— A new type of colloidal drug carrier , Int. J. Pharm. , 96 , R1 – R6 . 
96. Guilatt , R. L. , Couvreur , P. , Lambert , G. , Goldstein , D. , Benita , S. , and Dubernet , C. 
( 2004 ), Extensive surface studies help to analyse zeta potential data: The case of cationic 
emulsions , Chem. Phys. Lipids , 131 , 1 – 13 . 
97. Calvo , P. , Remu a - L o pez , C. , Vila - Jato , J. L. , and Alonso , M. J. ( 1997 ), Development of 
positively charged colloidal drug carriers: Chitosan - coated polyester nanocapsules and 
submicro - emulsions , Coll. Polym. Sci. , 275 , 46 – 53 . 
98. Jumaa , M. , and M u ller , B. W. ( 1999 ), Physicochemical properties of chitosan - lipid emulsions 
and their stability during the autoclaving process , Int. J. Pharm. , 183 , 175 – 184 . 
99. Samama , J. P. , Lee , K. M. , and Biellmann , J. F. ( 1987 ), Enzymes and microemulsions. 
Activity and kinetic properties of liver alcohol dehydrogenase in ionic water - in - oil 
microemulsions , Eur. J. Biochem. , 163 , 609 – 617 . 
100. Teixeira , H. , Dubernet , C. , Puisieux , F. , Benita , S. , and Couvreur , P. ( 1999 ), Submicron 
cationic emulsions as a new delivery system for oligonucleotides , Pharm. Res. , 16 , 
30 – 36 . 
101. Choi , B. Y. , Chung , J. W. , Park , J. H. , Kim , K. H. , Kim , Y. I. , Koh , Y. H. , Kwon , J. W. , Lee , 
K. H. , Choi , H. J. , Kim , T. W. , Kim, Y. J. , Chung , H. , Kwon, I. C. , and Jeong , S. Y. (2002), 
Gene delivery to the rat liver using cationic lipid emulsion/DNA complex: Comparison 
between intra - arterial, intraportal and intravenous administration , Korean J. Radiol. , 3 , 
194 – 198 . 
102. Yi , S. W. , Yune , Y. , Kim , T. W. , Chung , H. , Choi , Y. W. , Kwon , I. C. , Lee , E. B. , and Jeong , 
S. Y. ( 2000 ), A cationic lipid emulsion/DNA complex as a physically stable and serum - 
resistant gene delivery system , Pharm. Res. , 17 , 314 – 320 . 
103. Kim , Y. J. , Kim , T. W. , Chung , H. , Kwon , I. C. , Sung , H. C. , and Jeong , S. Y. ( 2003 ), The 
effects of serum on the stability and the transfection activity of the cationic lipid emulsion 
with various oils , Int. J. Pharm. , 252 , 241 – 252 . 
104. Tamilvanan , S. ( 2004 ), Oil - in - water lipid emulsions: Implications for parenteral and 
ocular delivering systems , Prog. Lipid Res. , 43 , 489 – 533 . 
105. Benita , S. ( 1999 ), Prevention of topical and ocular oxidative stress by positively charged 
submicron emulsion , Biomed. Pharm. , 53 , 193 – 206 . 
106. Abdulrazik , M. , Tamilvanan , S. , Khoury , K. , and Benita , S. ( 2001 ), Ocular delivery of 
cyclosporin A II. Effect of submicron emulsion ’ s surface charge on ocular distribution 
of topical cyclosporin A , STP Pharma Sci. , 11 , 427 – 432 . 
107. Rojanasakul , Y. , and Robinson , J. R. ( 1989 ), Transport mechanisms of the cornea: Characterization 
of barrier permselectivity , Int. J. Pharm. , 55 , 237 – 246 . 
108. Klang , S. , Abdulrazik , M. , and Benita , S. ( 2000 ), Infl uence of emulsion droplet surface 
charge on indomethacin ocular tissue distribution , Pharm. Dev. Technol. , 5 , 521 – 532 . 
109. Wretlind , A. ( 1981 ), Parenteral nutrition . Nutr. Rev. , 39 , 257 – 265 . 
110. Davis , S. S. ( 1982 ), Emulsions systems for the delivery of drugs by the parenteral route , 
in Bundgaard , H. , Bagger Hansen , A. , and Kofod , H. , Eds., Optimization of Drug 
Delivery , Munksgaard , Copenhagen , pp. 333 – 346 . 
111. Klang , S. H. , Parnas , M. , and Benita , S. ( 1998 ), Emulsions as drug carriers — Possibilities, 
limitations, and future perspectives , in Muller , R. H. , Benita , S. , and Bohm , H. L. , Eds., 
Emulsions and Nanosuspensions for the Formulation of Poorly Soluble drugs , 
Medpharm , Stuttgart , pp. 31 – 65 . 
REFERENCES 1361 
n

1362 OIL-IN-WATER NANOSIZED EMULSIONS 
112. Yang , S. C. , and Benita , S. ( 2000 ), Enhanced absorption and drug targeting by positively 
charged submicron emulsions , Drug Dev. Res. , 50 , 476 – 486 . 
113. Takino , T. , Koreeda , N. , Nomura , T. , Sakaeda , T. , Yamashita , F. , Takakura , Y. , and Hashida , 
M. ( 1998 ), Control of plasma cholesterol - lowering action of probucol with various lipid 
carrier systems , Biol. Pharm. Bull. , 21 , 492 – 497 . 
114. Klang , S. H , Frucht - Pery , J. , Hoffman , A. , and Benita , S. ( 1994 ), Physicochemical characterization 
and acute toxicity evaluation of a positively - charged submicron emulsion 
vehicle , J. Pharm. Pharmacol. , 46 , 986 – 993 . 
115. Korner , D. , Benita , S. , Albrecht , G. , and Baszkin , A. ( 1994 ), Surface properties of mixed 
phospholipid – stearylamine monolayers and their interaction with a non - ionic surfactant 
(poloxamer) , Coll. Surf. B BioInterf. , 3 , 101 – 109 . 
116. Benita , S. , and Levy , M. Y. ( 1993 ), Submicron emulsions as colloidal drug carriers 
for intravenous administration: Comprehensive physicochemical characterization , 
J. Pharm. Sci. , 82 , 1069 – 1079 . 
117. Tamilvanan , S. , and Benita , S. ( 2004 ), The potential of lipid emulsion for ocular delivery 
of lipophilic drugs , Eur. J. Pharm. Biopharm. , 58 , 357 – 368 . 
118. Lee , V. H. L. , and Robinson , J. R. ( 1986 ), Review: Topical ocular drug delivery: Recent 
developments and future challenges , J. Ocul. Pharmacol. , 2 , 67 – 108 . 
119. Jumaa , M. , and M u ller , B. W. ( 1998 ), The effect of oil components and homogenization 
condition on the physicochemical properties and stability of parenteral fat emulsions , 
Int. J. Pharm. , 163 , 81 – 89 . 
120. Constantinides , P. P. , Tustian , A. , and Kessler , D. R. ( 2004 ), Tocol emulsions for drug 
solubilization and parenteral delivery , Adv. Drug Deliv. Rev. , 56 , 1243 – 1255 . 
121. Constantinides , P. P. , Han , J. , and Davis , S. S. ( 2006 ), Advances in the use of tocols as 
drug delivery vehicles , Pharm. Res. , 23 , 243 – 255 . 
122. Lundberg , B. B. ( 1997 ), A submicron lipid emulsion coated with amphiphathic polyethylene 
glycol for parenteral administration of paclitaxel (taxol) , J. Pharm. Pharmacol. , 
49 , 16 – 21 . 
123. Han , J. , Davis , S. S. , Papandreou , C. , Melia , C. D. , and Washington , C. ( 2004 ), Design and 
evaluation of an emulsion vehicle for paclitaxel. I. Physicochemical properties and 
plasma stability , Pharm. Res. , 21 , 1573 – 1580 . 
124. Muchtar , S. , and Benita , S. ( 1994 ), Emulsions as drug carriers for ophthalmic use , Coll. 
Surf. A Physicochem. Eng. Aspects , 91 , 181 – 190 . 
125. Ogawa , S. , Decker , E. A. , and McClements , D. J. ( 2002 ), Production and characterization 
of o/w emulsions containing cationic droplets stabilized by lecithin - chitosan membranes , 
J. Agric. Food Chem. , 51 , 2606 – 2812 . 
126. Sonne , M. R. ( 2001 ), Tocopherol compositions for delivery of biologically active agents, 
USPTO Patent Full - Text and Image Database No. 6,193,985, Dumex, Copenhagen, 
pp. 1 – 21 . 
127. Sznitowska , M. , Janicki , S. , Dabrowska , E. A. , and Gajewska , M. ( 2002 ), Physicochemical 
screening of antimicrobial agents as potential preservatives for submicron emulsions , 
Eur. J. Pharm. Sci. , 15 , 489 – 495 . 
128. Jumaa , M. , Furkert , F. H. , and M u ller , B. W. ( 2002 ), A new lipid emulsion formulation 
with high antimicrobial effi cacy using chitosan , Eur. J. Pharm. Biopharm. , 53 , 115 – 123 . 
129. Pongcharoenkiat , N. , Narsimhan , G. , Lyons , R. T. , and Hem , S. L. ( 2002 ), The effect of 
surface charge and partition coeffi cient on the chemical stability of solutes in o/w emulsions 
, J. Pharm. Sci. , 91 , 559 – 570 . 
130. Han , J. , and Washington , C. ( 2005 ), Partition of antimicrobial additives in an intravenous 
emulsion and their effect on emulsion physical stability , Int. J. Pharm. , 288 , 263 – 271 . 

131. Swietlikowska , D. W. , and Sznitowska , M. ( 2006 ), Partitioning of parabens between 
phases of submicron emulsions stabilized with egg lecithin , Int. J. Pharm. , 312 , 174 – 
178 . 
132. Myers , R. A. , and Stella , V. J. ( 1992 ), Systemic bioavailability of penclomedine (NSC - 
338720) from oil - in - water emulsion administered intraduodenally to rats , Int. J. Pharm. , 
78 , 217 – 226 . 
133. Ilan , E. , Amselem , S. , Weisspapir , M. , Schwartz , J. , Yogev , A. , Zawoznik , E. , and 
Friedman , D. ( 1996 ), Improved oral delivery of desmopressin via a novel vehicle: 
Mucoadhesive submicron emulsion , Pharm. Res. , 13 , 1083 – 1087 . 
134. Palin , K. J. , Phillips , A. J. , and Ning , A. ( 1986 ), The oral absorption of cefaxitin from oil 
and water emulsion vehicles in rats , Int. J. Pharm. , 33 , 99 – 104 . 
135. Kimura , T. , Takeda , K. , Kageyn , A. , Toda , M. , Kurpsaki , Y. , and Nkayama , T. ( 1989 ), 
Intestinal absorption of dolichol from emulsions and liposomes in rats , Chem. Pharm. 
Bull. , 37 , 463 – 466 . 
136. Charman , S. A. , Charman , W. N. , Rogge , M. C. , Wildon , T. D. , Dutko , F. J. , and Pouton , 
C. W. ( 1992 ), Self - emulsifying drug delivery systems: Formulation and biopharmaceutical 
evaluation of an investigational lipophilic compound , Pharm. Res. , 9 , 87 – 93 . 
137. Gursoy , R. N. , and Benita , S. ( 2004 ), Self - emulsifying drug delivery systems (SEDDS) 
for improved oral delivery of lipophilic drugs , Biomed. Pharmacother. Dossier: Drug 
Deliv. Drug Effi cacy , 58 , 173 – 182 . 
138. Takino , T. , Nagahama , E. , Sakaeda (nee Kakutani) , T. , Yamashita , F. , Takakura , Y. , and 
Hashida , M. ( 1995 ), Pharmacokinetic disposition analysis of lipophilic drugs injected 
with various lipid carriers in the single - pass rat liver perfusion system , Int. J. Pharm. , 
114 , 43 – 54 . 
139. Tamilvanan , S. , Schmidt , S. , Muller , R. H. , and Benita , S. ( 2005 ), In vitro adsorption of 
plasma proteins onto the surface (charges) modifi ed - submicron emulsions for intravenous 
administration , Eur. J. Pharm. Biopharm. , 59 , 1 – 7 . 
140. Alany , R. G. , Rades , T. , Nicoll , J. , Tucker , I. G. , and Davies , N. M. ( 2006 ), W/O microemulsions 
for ocular delivery: Evaluation of ocular irritation and precorneal retention , 
J. Controlled Release , 111 , 145 – 152 . 
141. Stjernschantz , J. , and Astin , M. ( 1993 ), Anatomy and physiology of the eye: Physiological 
aspects of ocular drug therapy , in Edman , P. , Ed., Biopharmaceutics of Ocular Drug 
Delivery , CRC Press , Boca Raton, FL , pp. 1 – 25 . 
142. Lee , V. H. L. ( 1993 ), Precorneal, corneal and postcorneal factors , in Mitra , A. K. Ed., 
Ophthalmic Drug Delivery System , Marcel Dekker , New York , pp. 59 – 81 . 
143. J a rvinen , K. , J a rvinen , T. , and Urtti , A. ( 1995 ), Ocular absorption following topical 
delivery , Adv. Drug Deliv. Rev. , 16 , 3 – 19 . 
144. Prausnitz , M. R. , and Noonan , J. S. ( 1998 ), Permeability of cornea, sclera and conjunctiva: 
A literature analysis for drug delivery to eye , J. Pharm. Sci. , 87 , 1479 – 1488 . 
145. Washington , N. , Washington , C. , and Wilson , C. ( 2001 ), Ocular drug delivery , in 
Washington , N. , Washington , C. , and Wilson , C. , Eds., Physiological Pharm., Barriers to 
Drug Absorption , 2nd ed., Taylor & Francis , New York, pp. 250 – 270. 
146. Ludwig , A. ( 2005 ), The use of mucoadhesive polymers in ocular drug delivery , Adv. Drug 
Deliv. Rev. , 57 , 1595 – 1639 
147. Chien , D. S. , Homsy , J. J. , Gluchowski , C. , and Tang - Liu , D. D. ( 1990 ), Corneal and conjunctival/
scleral penetration of p - aminoclonidine, AGN 190342, and clonidine in rabbit 
eyes , Curr. Eye Res. , 9 , 1051 – 1059 . 
148. Ahmed , I. , and Patton , T. F. ( 1985 ), Importance of the noncorneal absorption route in 
topical ophthalmic drug delivery , Investigative Ophthalmol. Vis. Sci. , 26 , 584 – 587 . 
REFERENCES 1363

1364 OIL-IN-WATER NANOSIZED EMULSIONS 
149. Kaur , I. P. , and Smitha , R. ( 2002 ), Penetration enhancers and ocular bioadhesives: Two 
new avenues for ophthalmic drug delivery , Drug Dev. Ind. Pharm. , 28 , 353 – 369 . 
150. Dey , S. , Anand , B. S. , Patel , J. , and Mitra , A. K. ( 2003 ), Transporters/receptors in the 
anterior chamber: Pathways to explore ocular drug delivery strategies , Expert Opin. Biol. 
Ther. , 3 , 23 – 44 . 
151. Beilin , M. , Bar - Ilan , A. , and Amselem , S. ( 1995 ), Ocular retention time of submicron 
emulsion (SME) and the miotic response to pilocarpine delivered in SME . Investigative 
Ophthalmol. Vis. Sci. , 36 , S166 . 
152. Aiache , J. M. , el Meski , S. , Beyssac , E. , and Serpin , G. ( 1997 ), The formulation of drug 
for ocular administration , J. Biomater. Appl. , 11 , 329 – 348 . 
153. Maurice , D. M. , and Mishima , S. ( 1984 ), Ocular pharmacokinetics , in Sears , M. L. , Ed., 
Pharmacology. of the Eye , Springer Verlag , New York , pp. 20 – 116 . 
154. Muchtar , S. , Almog , S. , Torracca , M. T. , Saettone , M. F. , and Benita , S. ( 1992 ), A submicron 
emulsion as ocular vehicle for delta - 8 - tetrahydrocannabinol: Effect on intraocular pressure 
in rabbits , Ophthalm. Res. , 24 , 142 – 149 . 
155. Naveh , N. , Weissman , C. , Muchtar , S. , Benita , S. , and Mechoulam , R. ( 2000 ), A submicron 
emulsion of HU - 211, a synthetic cannabinoid, reduces intraocular pressure in rabbits , 
Graefe ’ s Arch. Clin. Exp. Ophthalmol. , 238 , 334 – 338 . 
156. Naveh , N. , Muchtar , S. , and Benita , S. ( 1994 ), Pilocarpine incorporated into a submicron 
emulsion vehicle causes an unexpectedly prolonged ocular hypotensive effect in rabbits , 
J. Ocul. Pharmacol. , 10 , 509 – 520 . 
157. Zurowska - Pryczkowska , K. , Sznitowska , M. , and Janicki , S. ( 1999 ), Studies on the effect 
of pilocarpine incorporation into a submicron emulsion on the stability of the drug and 
the vehicle , Eur. J. Pharm. Biopharm. , 47 , 255 – 260 . 
158. Sznitowska , M. , Janicki , S. , Zurowska - Pryczkowska , K. , and Mackiewicz , J. ( 2001 ), In 
vivo evaluation of submicron emulsions with pilocarpine: The effect of pH and chemical 
form of the drug , J. Microencapsul. , 18 , 173 – 181 . 
159. Calvo , P. , Alonso , M. J. , Vila - Jato , J. L. , and Robinson , J. R. ( 1996 ), Improved ocular bioavailability 
of indomethacin by novel ocular drug carriers , J. Pharm. Pharmacol. , 48 , 
1147 – 1152 . 
160. Calvo , P. , Vila - Jato , J. L. , and Alonso , M. J. ( 1996 ), Comparative in vitro evaluation of 
several colloidal systems, nano - particles, nanocapsules, and nanoemulsions as ocular 
drug carriers , J. Pharm. Sci. , 85 , 530 – 536 . 
161. Anselem , S. , Beilin , M. , and Garty , N. ( 1993 ), Submicron emulsion as ocular delivery 
system for adaprolol maleate, a soft . - blocker , Pharm. Res. , 10 , S205 . 
162. Melamed , S. , Kurtz , S. , Greenbaum , A. , Haves , J. F. , Neumann , R. , and Garty , N. ( 1994 ), 
Adaprolol maleate in submicron emulsion, a novel soft . - blocking agent, is safe and 
effective in human studies , Investigative Ophthalmol. Vis. Sci. , 35 , 1387 – 1390 . 
163. Aviv , H. , Friedman , D. , Bar - Ilan , A. , and Vered , M. ( 1995 ), Submicron emulsions as 
ocular drug delivery vehicles, U.S. Patent 5,496,811 , March 5, 1996. 
164. Garty , N. , Lusky , M. , Zalish , M. , Rachmiel , R. , Greenbaum , A. , Desatnik , H. , Neumann , 
R. , Howes , J. F. , and Melamed , S. ( 1994 ), Pilocarpine in submicron emulsion formulation 
for treatment of ocular hypertension: A phase II clinical trial , Investigative Ophthalmol. 
Vis. Sci. , 35 , 2175 . 
165. Ding , S. , Tien , W. , and Olejnik , O. ( 1995 ), Nonirritating emulsions for sensitive tissue, 
U.S. Patent 5,474,979 (to Allergan), December 12. 
166. Ding , S. , and Olejnik , O. ( 1997 ), Cyclosporin ophthalmic oil/water emulsions. Formulation 
and characterization , Pharm. Res. , 14 , S41 . 

167. Acheampong , A. A. , Shackleton , M. , Tang - Liu , DD - S. , Ding , S. , Stern , M. E. , and Decker , 
R. ( 1999 ), Distribution of cyclosporin A in ocular tissues after topical administration to 
albino rabbits and beagle dogs , Curr. Eye Res. , 18 , 91 – 103 . 
168. Stevenson , D. , Tauber , J. , and Reis , B. L. ( 2000 ), Effi cacy and safety of cyclosporin A 
ophthalmic emulsion in the treatment of moderate - to - severe dry eye disease: A dose - 
ranging, randomized trial. The Cyclosporin A Phase 2 Study Group , Ophthalmology , 
107 , 967 – 974 . 
169. Sall , K. , Stevenson , O. D. , Mundorf , T. K. , and Reis , B. L. ( 2000 ), Two multicenter, randomized 
studies of the effi cacy and safety of cyclosporin ophthalmic emulsion in 
moderate - to - severe dry eye disease. CsA Phase 3 Study Group , Ophthalmology , 107 , 
631 – 639 . 
170. Turner , K. , Pfl ugfelder , S. C. , Ji , Z. , Feuer , W. J. , Stern , M. , and Reis , B. L. ( 2000 ), Interleukin 
- 6 levels in the conjunctival epithelium of patients with dry eye disease treated 
with cyclosporine ophthalmic emulsion , Cornea , 19 , 492 – 496 . 
171. Kunert , K. S. , Tisdale , A. S. , Stern , M. E. , Smith , J. A. , and Gipson , I. K. ( 2000 ), Analysis 
of topical cyclosporin treatment of patients with dry eye syndrome: Effect on conjunctival 
lymphocytes , Arch. Ophthalmol. , 118 , 1489 – 1496 . 
172. Brignole , F. , Pisella , P. J. , Saint - Jean , M. De. , Goldschild , M. , Goguel , A. , and Baudouin , 
C. ( 2001 ), Flow cytometric analysis of infl ammatory markers in KCS: 6 - month treatment 
with topical cyclosporin A . Investigative Ophthalmol. Vis. Sci. , 42 , 90 – 95 . 
173. Small , D. S. , Acheampong , A. , Reis , B. , Stern , K. , Stewart , W. , Berdy , G. , Epstein , R. , 
Foerster , R. , Forstot , L. , and Tang - Liu , D. D. - S. ( 2002 ), Blood concentrations of 
cyclosporin A during long - term treatment with cyclosporin A ophthalmic emulsions 
in patients with moderate to severe dry eye disease , J. Ocul. Pharmacol. Ther. , 18 , 
411 – 418 . 
174. Galatoire , O. , Baudouin , C. , Pisella , P. J. , and Brignole , F. ( 2003 ), Flow cytometry in 
impression cytology during keratoconjunctivitis sicca: Effects of topical cyclosporin A 
on HLA DR expression , J. Francais d ’ Ophtalmol. , 26 , 337 – 343 . 
175. Tang - Liu , D. D. , and Acheampong , A. ( 2005 ), Ocular pharmacokinetics and safety of 
ciclosporin, a novel topical treatment for dry eye , Clin. Pharmacokinet. , 44 , 247 – 261 . 
176. Hom , M. M. ( 2006 ), Use of cyclosporine 0.05% ophthalmic emulsion for contact lens - 
intolerant patients , Eye Contact Lens: Sci. Clin. Pract. , 32 , 109 – 111 . 
177. Sall , K. N. , Cohen , S. M. , Christensen , M. T. , and Stein , J. M. ( 2006 ), An evaluation of the 
effi cacy of a cyclosporine - based dry eye therapy when used with marketed artifi cial tears 
as supportive therapy in dry eye , Eye Contact Lens: Sci. Clin. Pract. , 32 , 21 – 26 . 
178. Klang , S. H. , Siganos , C. S. , and Benita , S. ( 1999 ), Evaluation of a positively - charged 
submicron emulsion of piroxicam in the rabbit corneum healing process following alkali 
burn , J. Controlled Release , 57 , 19 – 27 . 
179. Tamilvanan , S. , Khoury , K. , Gilhar , D. , and Benita , S. ( 2001 ), Ocular delivery of cyclosporin 
A. I. Design and characterization of cyclosporin A - loaded positively charged 
submicron emulsion , STP Pharma Sci. , 11 , 421 – 426 . 
180. Hussain, A. A. (1998), Intranasal drug delivery , Adv. Drug Deliv. Rev. , 29 , 39 – 49 . 
181. Kararli , T. T. , Needham , T. E. , Schoenhard , G. , Baron , D. A. , Schmidt , R. E. , Katz , B. , and 
Belonio , B. ( 1992 ), Enhancement of nasal delivery of a rennin inhibitor in the rat using 
emulsion formulations , Pharm. Res. , 9 , 1024 – 1028 . 
182. Ko , K. Y. , Needham , T. E. , and Zia , H. ( 1998 ), Emulsion formulations of testosterone for 
nasal administration , J. Microencapsul. , 15 , 197 – 205 . 
183. Aikawa , K. , Matsumoto , K. , Uda , H. , Tanaka , S. , Shimamura , H. , Aramaki , Y. , and 
Tsuchiya , S. ( 1998 ), Prolonged release of drug from o/w emulsion and residence in rat 
nasal cavity , Pharm. Dev. Technol. , 3 , 461 – 469 . 
REFERENCES 1365

1366 OIL-IN-WATER NANOSIZED EMULSIONS 
184. Mitra , R. , Pezron , I. , Chu , W. A. , and Mitra , A. K. ( 2000 ), Lipid emulsions as vehicles for 
enhanced nasal delivery of insulin , Int. J. Pharm. , 205 , 127 – 134 . 
185. Kim , T. W. , Chung , H. , Kwon , I. C. , Sung , H. C. , and Jeong , S. Y. ( 2000 ), In vivo gene 
transfer to the mouse nasal cavity mucosa using a stable cationic lipid emulsion , Mol. 
Cells , 10 , 142 – 147 . 
186. Jenkins , P. ( 1999 ), Mucosal vaccine delivery , Exp. Opin. Ther. Patients , 9 , 255 – 262 . 
187. Benita , M. B. , Oudshoorn , M. , Romeijn , S. , Meijgaarden , K. V. , Koerten , H. , Meulen , H. 
V. D , Lambert , G. , Ottenhoff , T. , Benita , S. , Junginger . H. , and Borchard , G. ( 2004 ), Cationic 
submicron emulsions for pulmonary DNA immunization , J. Controlled Release , 
100 , 145 – 155 . 
188. El - Hameed , M. D. A. , and Kellaway , I. W. ( 1997 ), Preparation and in vitro characterization 
of mucoadhesive polymeric microspheres as intranasal delivery systems , Eur. J. 
Pharm. Biopharm. , 44 , 53 – 60 . 
189. Friedman , D. I. , Schwarz , J. S. , and Weisspapir , M. ( 1995 ), Submicron emulsion vehicle 
for enhanced transdermal delivery of steroidal and nonsteroidal antiinfl ammatory 
drugs , J. Pharm. Sci. , 84 , 324 – 329 . 
190. Schwarz , J. S. , Weisspapir , M. R. , and Friedman , D. I. ( 1995 ), Enhanced transdermal 
delivery of diazepam by submicron emulsion (SME) creams , Pharm. Res. , 12 , 687 – 692 . 
191. Realdaon , N. , and Ragazzi , E. ( 1998 ), Study of drug availability from o/w emulsions 
obtained in different manufacturing conditions, in Proceedings of 2nd World Meeting 
APGI/APV , 775 – 776 . 
192. Welin - Berger , K. , and Bergenstahl , B. ( 2000 ), Inhibition of Ostwald ripening in local 
anesthetic emulsions — By using hydrophobic excipients in the disperse phase , Int. J. 
Pharm. , 200 – 202 , 249 – 260 . 
193. Welin - Berger , K. , Neelissen , J. A. , and Bergenstahl , B. ( 2001 ), The effect of rheological 
behaviour of a topical anaesthetic formulation on the release and permeation rates of 
the active compound , Eur. J. Pharm. Sci. , 13 , 309 – 318 . 
194. Dutta , S. ( 1999 ), Use of eutectic mixture of local anesthetics in children , Ind. J. Pediatr. , 
66 , 707 – 715 . 
195. Cordoni , A. , and Cordoni , L. E. ( 2001 ), Eutectic mixture of local anesthetics reduces 
pain during intravenous catheter insertion in the pediatric patient , Clin. J. Pain , 17 , 
115 – 118 . 
196. Ezra , R. , Benita , S. , Ginsberg , I. , and Kohen , R. ( 1996 ), Prevention of oxidative damage 
in fi broblast cell cultures and rat skin by positively - charged submicron emulsion of a - 
tocopherol , Eur. J. Pharm. Biopharm. , 42 , 291 – 298 . 
197. Piemi , M. P. , Korner , D. , Benita , S. , and Marty , J. P. ( 1999 ), Positively and negatively 
charged submicron emulsions for enhanced topical delivery of antifungal drugs , J. Controlled 
Release , 58 , 177 – 187 . 

1367 
INDEX 
Abortifacients, 850 
Acyclovir, 1037–1042 
Adjuvants, 635–637 
ADMET, 8–9 
AERx, 709–710 
AIDS, 4 
Alginate nanoparticles, 540–541 
Alginic acid, 295 
Anticancer drug delivery, 485–506 
Antimicrobials, 845–846 
API, 5 
Artifi cial neural networks, 1016 
Aseptic compounding, 107–108 
Auxiliary excipients, 894–895 
Avonex, 47 
Aztirelin, 620 
Bentonite, 295 
Benzodiazepines, 623–626 
Betaseron, 47 
Bioadhesion, 305–306 
Bioburden considerations, 26 
Bioconversion, 566, 572–574 
Biodegradable microspheres, 419–426 
Biodegradable polymeric nanoparticles, 
536–543 
Biodrug, 565–566 
Biogenerics, 35 
Biological half-life, 356 
Biopharmaceuticals Classifi cation System 
(BCS), 237–238, 961 
Boron Neutron Capture Therapy (BNCT), 
489 
Breast cancer, 492–497 
Breath actuation, 698–699 
CaCo-2, 960 
Caclyx, 497 
Calcitonin, 613–617 
Cancer therapy, 1238–1240 
Capillary Aerosol Generator (CAG), 
710–711 
Carbomer, 295–296 
Carbxymethyl cellulose, 655 
Carnauba wax, 274 
Carr index, 908 
Carrageenan, 296, 833 
Challenges in ocular drug delivery, 
730–737 
Characteristics of radiopharmaceuticals, 
60–61 
Chemical penetration enhancers, 803 
Chemically induced release, 384–385 
Chitosan nanoparticles, 541 
Chitosan, 608, 636, 655, 657, 658, 661–662, 
665–666, 833 
Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad 
Copyright © 2008 John Wiley & Sons, Inc.

1368 INDEX 
Cholera toxin, 637 
Ciliotoxicity, 668 
Classifi cation of hygroscopicity, 912 
Climatization, 1085–1086 
CMV, 481 
Coated tablets, 244–245 
Colloidal silicon dioxide, 296 
Colorants, 243 
Compactibility, 1138–1141 
Compendial gels, 307 
Compendial ointments, 289–291 
Compressibility, 917–918 
Container closure systems, 17–18 
Controlled-release delivery systems, 11 
Cryogenic spray drying, 401–402 
Cytotoxicity, 1240 
Defi nitions of density terms, 1178 
Dendrimers, 1272 
Depyrogenation, 117–120 
Diluents, 240–241 
Diphtheria Toxoid (DT), 420–421 
DirectHaler, 602–604 
Disintegration, 920–922 
DPI, 689 
Drug product stability, 21–25 
Drug-excipient compatibility, 969–970 
Dry Powder Inhalers (DPIs), 684, 
700–706 
Effective half life, 61 
Effervescent tablets, 251–252 
Electroresponsive release, 381–383 
Ethylcellulose, 297 
Excipients, 1344, 19–21, 239–244, 243–244, 
410–412, 412–419, 695, 822–823, 883– 
896, 884–885, 897 
Exubera, 704, 705–706 
Fair Packaging and Labeling Act, 
190–195 
FDA-approved transdermal patches, 794 
First-uterine-pass effect, 821 
Fluidized-bed coating, 1102–1103 
FluMist, 592 
Formulation approaches to improve ocular 
bioavailability, 737–753 
Formulation assessment, 7–8 
Formulation development, 15–16, 238 
Fortical, 52–53, 55 
Friability, 928 
Fullerenes, 1272, 1296–1297 
Gas and vapor sterilization, 119 
Gastrointestinal tract and absorption, 
356–357 
Gelatin capsules, 245–251 
Gelatin, 539 
Gelling agents, 293–301 
GELS, 288–310 
Giladin, 540 
Glucose-responsive insulin release device, 
384–385 
Glycerol behenate, 298–299 
Group B streptococcus vaccine, 420 
Guar gum, 297 
Hammer Mill, 1169 
Hausner ratio, 908 
Herpes simplex virus, 481 
HFA reformulation, 690–692 
High-throughput Screening (HTS), 934 
Human Growth Hormone (HGH), 34 
Hyaluronic acid, 499, 655,657, 833 
Hydralazine, 627 
Hydrogels, 291–292 
Hydrophilic matrix tablets, 1210–1211 
Hydroxyethyl Cellulose (HEC), 297–298 
Hydroxypropylmethyl Cellulose (HPMC), 
298 
Immunity after intranasal immunization, 
634–635 
Immunogenicity, 50, 53–54 
Inhalation drug products, 179 
Injectable microspheres, 407–408 
Insulin, 424–426 
Ionophoresis, 804 
Japanese Encephalitis Virus (JEV), 
423–424 
Kurve Technology, 601 
Labor inducers, 850 
Lanolin, 271 
Lipinski Rule of Five, 934 
Liposomal drugs approved for clinical 
application, 1263 
Liposome-based products currently under 
clinical testing, 484 
Liposomes, 365–367, 636, 747–748 
Liquid dosage forms, 338 
Low-molecular-weight heparins, 617–620 
Lozenges, 252–253

INDEX 1369 
Lung cancer, 497–502 
Lung toxicity, 1309–1310 
Lyophilization, 127–128 
Magnetically induced release, 383–384 
Marked medical and nonmedical 
emulsions, 1332 
MDI, 689 
Mechanisms of protein and peptide 
degradation, 22–23 
Metal as packaging material, 170–171 
Metered-dose Inhalers (MDIs), 684, 
690–700 
Microbicides, 843–845 
Microbiological quality, 334–335 
Microcrystalline cellulose (MCC), 653, 655 
Microemulsions, 1267, 748–750 
Microencapsulation, 358 
Microneedles, 803–804 
Milestones in early biologics regulation, 38 
MLVs (multilamellar vesicles), 444 
Mononuclear phagocyte system, 1333 
Mucoadhesion, 840 
Mucoadhesive microspheres, 657 
Mucoadhesive polymers, 744 
Mucosal toxicity screening method using 
the slug arion lusitanicus, 667 
Mucosal-associated lymphoid tissue 
(MALT), 635 
Musciliary clearance, 596 
Nanocapsules, 363 
Nanocarriers, 1258–1273 
Nanoemulsions, 1269 
Nanogels, 1271 
Nanomaterials in pharmacy, 1253–1254 
Nanomaterials, 1250–1252 
Nanomedicine technologies taxonomy, 
1292–1293 
Nanomedicine, 1278–1279 
Nanoparticles, 1231–1236, 1264–1269, 536, 
746–747 
Nasal delivery, 481–482 
Nasal delivery of nonpeptide molecules, 
622–630 
Nasal delivery of vaccines, 633–637 
Nasal dry powder formulations, 652–655 
Nasal route, 1352–1353 
Nasal vaccination delivery systems, 
636–637 
Nasal vasculature, 594–595 
Nebulizers, 706–707 
Niosomes, 367, 748 
Nitroglycerin, 627–628 
Noncovalent binding of ligands, 465–466 
Nose-associated Lymphoid Tissue 
(NALT), 635 
Nose-to-Brain Delivery, 632 
Ocular delivery, 477–481 
Ocular drug delivery, 738–741, 784–785 
Ocular routes, 1347–1352 
Offi cial creams, 282 
Offi cial gels, 304 
Oil-in-water nanosized emulsions, 
1329–1341 
Ointments and creams, 269–270 
Omnitrope, 51, 53–56 
OptiNose, 601–602 
Oral drug delivery, 781–782 
Oral ER formulations, 1193–1195 
Orally disintegrating tablets, 259–262 
Organogels, 292 
Ovarian cancer, 502–506 
Pan coating, 1102 
Parenteral drug delivery, 783–784 
Parenteral routes, 1346–1347 
Partition coeffi cient, 352, 956–957 
PEGylated liposomes, 469–472 
Percolation theory, 1013–1016, 1030–1042 
Permeability enhancement methods, 964 
Preservatives, 20–21 
PET radiopharmaceuticals, 83 
Petrolatum, 272 
Photostability, 23 
pH-sensitive polymeric nanoparticles, 547 
Physiochemical properties of liposomes, 
449–456 
Plastic additives, 164 
Plastic as packaging material, 166–170 
Poloxamer, 299 
Poly (lactic acid), 543–544 
Polyethylene oxide, 299–300 
Polymorphism, 936–942 
Polysaccharides, 539–540 
Polyvinyl Alcohol (PVA), 300 
Povidone, 300 
Preformulation approaches for tablet 
production, 883 
Principles for extended drug release, 
1196–1197 
Principles of radiation protection, 63–64 
Production of radionuclides, 75–76

1370 INDEX 
Production of radiopharmaceuticals, 78–88 
Propylene Glycol Alginate (PGA), 300 
Quantum dots, 1293–1295 
Radiation sterilization, 119 
Radioactive decay, 61–63 
Radiochemical purity, 90–91 
Radionuclides, 65 
Reaction calorimetry, 141–142 
Respmat, 708–709 
Route of administration, 8–10 
Salmon calcitonin, 52 
Salt selection, 952–956 
Scanning tunneling microscopy, 1306–1308 
Selected drugs administered in vagina, 853 
Selection guideline of pharmaceutical 
excipients, 895–896 
Selection of microemulsion ingredients, 
773 
Sodium alginate, 300–301, 538, 655 
Soft Mist Aerosols, 707–708 
Solubility characteristics, 950–965 
Sonophoresis, 804 
Spermicides, 849–850 
Stability, 336–337 
Stability-indicating methodologies, 14–15 
Stability of liposomes, 455–456 
Sterile products, 169–170 
Sterilization, 117–120 
Sterilization by fi ltration, 119–120 
Sterilization of radiopharmaceuticals, 
73–74 
Surface hydrophilicity, 550 
Synthesis of PET radiopharmaceuticals, 86 
Synthetic cervical mucus, 816 
Systemic uptake of nanoparticles, 
1310–1311 
Tablet coating methods, 1102–1103 
Tablet tooling terminology, 1147 
Tableting machines, 1058–1067 
Tableting process, 1055–1056 
Tetanus toxoid, 421–423 
Thermoresponsive drug release dosage 
forms, 379–381 
Thermosensitive polymeric nanoparticles, 
546–547 
TNO gastrointestinal tract model, 569–571 
Topical route, 1353–1355 
Toxicological effects of dry powder 
formulation, 666–667 
Tragacanth, 301 
Transdermal drug delivery, 368, 782–783 
Ultrasonic atomization, 403 
Ultrasound-assisted tableting, 1043–1045 
United States Pharmacopoeia Center for 
the Advancement of Patient Safety, 195 
U.S. Pharmacopoeia, 177 
USP, 281–282, 304, 903 
Vaccines, 420–424, 851–852 
Vaginal and uterine controlled-release 
dosage forms, 371 
Vaginal fi lms, 831 
Vaginal fl uid stimulant, 816 
Vaginal foams, 831 
Vaginal rings, 826–830 
Vaginal sponges, 832 
Vibrio cholerae vaccine, 423