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 compl