throbber
PHARMACEUTICAL
`MANUFACTURING
`HANDBOOK
`
`Production and
`Processes
`
`SHAYNE COX GAD, PH.D., D.A.B.T.
`Gad Consulting Services
`Cary, North Carolina
`
`A JOHN WILEY & SONS, INC., PUBLICATION
`
`Novartis Exhibit 2336.001
`Regeneron v. Novartis, IPR2021-00816
`
`

`

`PHARMACEUTICAL
`MANUFACTURING
`HANDBOOK
`
`Production and
`Processes
`
`Novartis Exhibit 2336.002
`Regeneron v. Novartis, IPR2021-00816
`
`

`

`PHARMACEUTICAL
`MANUFACTURING
`HANDBOOK
`
`Production and
`Processes
`
`SHAYNE COX GAD, PH.D., D.A.B.T.
`Gad Consulting Services
`Cary, North Carolina
`
`A JOHN WILEY & SONS, INC., PUBLICATION
`
`Novartis Exhibit 2336.003
`Regeneron v. Novartis, IPR2021-00816
`
`

`

`Copyright © 2008 by John Wiley & Sons, Inc. All rights reserved
`
`Published by John Wiley & Sons, Inc., Hoboken, New Jersey
`Published simultaneously in Canada
`
`No part of this publication may be reproduced, stored in a retrieval system, or transmitted in any
`form or by any means, electronic, mechanical, photocopying, recording, scanning, or otherwise, except
`as permitted under Section 107 or 108 of the 1976 United States Copyright Act, without either the
`prior written permission of the Publisher, or authorization through payment of the appropriate per-
`copy fee to the Copyright Clearance Center, Inc., 222 Rosewood Drive, Danvers, MA 01923, (978)
`750-8400, fax (978) 750-4470, or on the web at www.copyright.com. Requests to the Publisher for per-
`mission should be addressed to the Permissions Department, John Wiley & Sons, Inc., 111 River
`Street, Hoboken,
`NJ 07030, (201) 748-6011, fax (201) 748-6008, or online at http://www.wiley.com/go/permission.
`
`Limit of Liability/Disclaimer of Warranty: While the publisher and author have used their best efforts
`in preparing this book, they make no representations or warranties with respect to the accuracy
`or completeness of contents of this book and specifi cally disclaim any implied warranties of
`merchantability or fi tness for a particular purpose. No warranty may be created or extended by sales
`representatives or written sales materials. The advice and strategies contained herein may not be
`suitable for your situation. You should consult with a professional where appropriate. Neither the
`publisher nor author shall be liable for any loss of profi t or any other commercial damages, including
`but not limited to special, incidental, consequential, or other damages.
`
`For general information on our other products and services or for technical support, please contact
`our Customer Care Department within the United States at (800) 762-2974, outside the United States
`at (317) 572-3993 or fax (317) 572-4002.
`
`Wiley also publishes its books in a variety of electronic formats. Some content that appears in print
`may not be available in electronic formats. For more information about Wiley products, visit our web
`site at www.wiley.com.
`
`Library of Congress Cataloging-in-Publication Data is available.
`
`ISBN: 978-0-470-25958-0
`
`Printed in the United States of America
`
`10 9 8 7 6 5 4 3 2 1
`
`Novartis Exhibit 2336.004
`Regeneron v. Novartis, IPR2021-00816
`
`

`

` CONTRIBUTORS
`
`
`
`
`
` Susanna Abrahms é n - Alami, AstraZeneca R & D Lund, Lund, Sweden,
`Extended - Release Formulations
`
` Oral
`
` 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 é d ’ Auvergne, Clermont - Ferrand, France, Recombinant
`Saccharomyces Cerevisiae as New Drug Delivery System to Gut: In Vitro Valida-
`tion and Oral Formulation
`
` Sacide Alsoy Altinkaya, Izmir Institute of Technology, Urla - Izmir, Turkey, Con-
`trolled Release of Drugs from Tablet Coatings
`
` Maria Helena Amaral, University of Porto, Porto, Portugal,
`Delivery
`
` Vaginal Drug
`
` 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
`
`Novartis Exhibit 2336.005
`Regeneron v. Novartis, IPR2021-00816
`
`

`

`vi
`
` CONTRIBUT ORS
`
` 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,
`Delivery
`
` Vaginal Drug
`
` 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, Cyclo-
`dextrin - Based Nanomaterials in Pharmaceutical Field
`
` St é phanie Blanquet, Universit é d ’ Auvergne, Clermont - Ferrand, France, Recombi-
`nant 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 Phar-
`macy, 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 é 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
`č č
` Jelena Filipovi ç - Gr i , Faculty of Pharmacy and Biochemistry, University of
`Zagreb, Zagreb, Croatia, Nasal Powder Drug Delivery
`
`Novartis Exhibit 2336.006
`Regeneron v. Novartis, IPR2021-00816
`
`

`

`CONTRIBUTORS
`
` vii
`
` Eddy Castellanos Gil, Center of Pharmaceutical Chemistry and University of
`Havana, Havana, Cuba; University of Sevilla, Seville, Spain; University of Mont-
`pelier 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, Nanotechnol-
`ogy 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 ö 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, Semi-
`solid 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
`
`Novartis Exhibit 2336.007
`Regeneron v. Novartis, IPR2021-00816
`
`

`

`viii
`
` CONTRIB UTORS
`
` Iv á n Pe ñ 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 Produc-
`tion 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 Phar-
`macy, 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 ê s Rocha Miritello Santoro, Department of Pharmacy, Faculty of Pharma-
`ceutical Sciences, University of S ã o Paulo, S ã o Paulo, Brazil, Packaging and
`Labeling
`
` Helton M.M. Santos, University of Coimbra, Coimbra, Portugal,
`Compression
`
` Tablet
`
` 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 ã o Paulo, S ã o Paulo, Brazil, Packaging and Labeling
`
` Jo ã o J.M.S. Sousa, University of Coimbra, Coimbra, Portugal,
`Compression
`
` Tablet
`
` Shunmugaperumal Tamilvanan, University of Antwerp, Antwerp, Belgium, Prog-
`ress 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, Microemul-
`sions as Drug Delivery Systems
`
` Hui Zhai, Queen ’ s University Belfast, Belfast, Northern Ireland, Effects of Grind-
`ing in Pharmaceutical Tablet Production
`
`
`
`Novartis Exhibit 2336.008
`Regeneron v. Novartis, IPR2021-00816
`
`

`

`CONTENTS
`
`PREFACE
`
`SECTION 1 MANUFACTURING SPECIALTIES
`
`1.1 Biotechnology-Derived Drug Product Development
`
`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
`Erin Oliver, Stephen M. Carl, Kenneth R. Morris, Gerald W. Becker, and
`Gregory T. Knipp
`
`
`
`1.3 Radiopharmaceutical Manufacturing
`
`Brit S. Farstad and Iván Peñuelas
`
`SECTION 2 ASEPTIC PROCESSING
`
`2.1
`
`
`Sterile Product Manufacturing
`James Agalloco and James Akers
`
`SECTION 3 FACILITY
`
`3.1
`
`
`
`From Pilot Plant to Manufacturing: Effect of Scale-Up on
`Operation of Jacketed Reactors
`B. Wayne Bequette
`
`xiii
`
`1
`
`3
`
`33
`
`59
`
`97
`
`99
`
`137
`
`139
`
`ix
`
`Novartis Exhibit 2336.009
`Regeneron v. Novartis, IPR2021-00816
`
`

`

`x
`
` CONTENTS
`
`3.2 Packaging and Labeling
`
`Maria Inês Rocha Miritello Santoro and Anil Kumar Singh
`
`3.3 Clean-Facility Design, Construction, and Maintenance Issues
`
`Raymond K. Schneider
`
`SECTION 4 NORMAL DOSAGE FORMS
`
`4.1
`
`
`4.2
`
`
`Solid Dosage Forms
`Barbara R. Conway
`
`Semisolid Dosages: Ointments, Creams, and Gels
`Ravichandran Mahalingam, Xiaoling Li, and Bhaskara R. Jasti
`
`4.3 Liquid Dosage Forms
`
`Maria V. Rubio-Bonilla, Roberto Londono, and Arcesio Rubio
`
`SECTION 5 NEW DOSAGE FORMS
`
`5.1 Controlled-Release Dosage Forms
`
`Anil Kumar Anal
`
`5.2 Progress in the Design of Biodegradable Polymer-Based
`Microspheres for Parenteral Controlled Delivery of Therapeutic
`Peptide/Protein
`Shunmugaperumal Tamilvanan
`
`
`
`5.3 Liposomes and Drug Delivery
`
`Sophia G. Antimisiaris, Paraskevi Kallinteri, and Dimitrios G. Fatouros
`
`5.4 Biodegradable Nanoparticles
`
`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
`Stéphanie Blanquet and Monique Alric
`
`
`
`5.6 Nasal Delivery of Peptide and Nonpeptide Drugs
`
`Chandan Thomas and Fakhrul Ahsan
`
`5.7 Nasal Powder Drug Delivery

`č ć
`
`Jelena Filipovi -Gr i and Anita Hafner
`
`5.8 Aerosol Drug Delivery
`
`Michael Hindle
`
`5.9 Ocular Drug Delivery
`
`Ilva D. Rupenthal and Raid G. Alany
`
`5.10 Microemulsions as Drug Delivery Systems
`
`Raid G. Alany and Jingyuan Wen
`
`159
`
`201
`
`233
`
`235
`
`267
`
`313
`
`345
`
`347
`
`393
`
`443
`
`535
`
`565
`
`591
`
`651
`
`683
`
`729
`
`769
`
`Novartis Exhibit 2336.0010
`Regeneron v. Novartis, IPR2021-00816
`
`

`

`CONTENTS
`
` xi
`
`5.11 Transdermal Drug Delivery
`
`C. Scott Asbill and Gary W. Bumgarner
`
`5.12 Vaginal Drug Delivery
`
`José das Neves, Maria Helena Amaral, and Maria Fernanda Bahia
`
`SECTION 6 TABLET PRODUCTION
`
`6.1 Pharmaceutical Preformulation: Physicochemical Properties of
`Excipients and Powers and Tablet Characterization
`Beom-Jin Lee
`
`
`
`6.2 Role of Preformulation in Development of Solid Dosage Forms
`
`Omathanu P. Perumal and Satheesh K. Podaralla
`
`6.3 Tablet Design
`
`Eddy Castellanos Gil, Isidoro Caraballo, and Bernard Bataille
`
`6.4 Tablet Production Systems
`
`Katharina M. Picker-Freyer
`
`6.5 Controlled Release of Drugs from Tablet Coatings
`
`Sacide Alsoy Altinkaya
`
`6.6 Tablet Compression
`
`Helton M. M. Santos and João J. M. S. Sousa
`
`6.7 Effects of Grinding in Pharmaceutical Tablet Production
`
`Gavin Andrews, David Jones, Hui Zhai, Osama Abu Diak, and
`Gavin Walker
`
`6.8 Oral Extended-Release Formulations
`
`Anette Larsson, Susanna Abrahmsén-Alami, and Anne Juppo
`
`SECTION 7 ROLE OF NANOTECHNOLOGY
`
`7.1 Cyclodextrin-Based Nanomaterials in Pharmaceutical Field
`
`Erem Bilensoy and A. Attila Hincal
`
`7.2 Nanotechnology in Pharmaceutical Manufacturing
`
`Yiguang Jin
`
`7.3 Pharmaceutical Nanosystems: Manufacture, Characterization,
`and Safety
`D. F. Chowdhury
`
`
`
`7.4 Oil-in-Water Nanosized Emulsions: Medical Applications
`
`Shunmugaperumal Tamilvanan
`
`INDEX
`
`793
`
`809
`
`879
`
`881
`
`933
`
`977
`
`1053
`
`1099
`
`1133
`
`1165
`
`1191
`
`1223
`
`1225
`
`1249
`
`1289
`
`1327
`
`1367
`
`Novartis Exhibit 2336.0011
`Regeneron v. Novartis, IPR2021-00816
`
`

`

` 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 medi-
`cine. These 34 chapters cover the full range of approaches to developing and pro-
`ducing 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 therapeu-
`tic 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 funda-
`mental problems that stand in the way of so many potentially successful pharmaco-
`therapeutic interventions.
`
`xiii
`
`Novartis Exhibit 2336.0012
`Regeneron v. Novartis, IPR2021-00816
`
`

`

`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
` 2.1.2
`
` 2.1.3
`
` 2.1.4
`
` 2.1.5
`
` Introduction
` Process Selection and Control
` 2.1.2.1
` Formulation and Compounding
` 2.1.2.2
` Primary Packaging
` 2.1.2.3
` Process Objectives
` 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
` Aseptic Processing Facility Alternatives
` 2.1.4.1
` Expandability
` 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.
`
`99
`
`Novartis Exhibit 2336.0013
`Regeneron v. Novartis, IPR2021-00816
`
`

`

`100
`
`STERILE PRODUCT MANUFACTURING
`
` 2.1.6
`
` 2.1.7
` 2.1.8
`
` 2.1.9
`
` 2.1.10
` 2.1.11
`
` 2.1.12
` 2.1.13
`
`
`
`
`
`
`
` 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
` Facility and System: Qualifi cation and Validation
` Environmental Control and Monitoring
` 2.1.8.1
` Sanitization and Disinfection
` 2.1.8.2
` Monitoring
` 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
` Personnel
` 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
` Terminal Sterilization
` 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 contami-
`nated 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 require-
`ments 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
`
`Novartis Exhibit 2336.0014
`Regeneron v. Novartis, IPR2021-00816
`
`

`

`PROCESS SELECTION AND DESIGN
`
`101
`
`(FDA) in its 2004 Guideline on Sterile Drug Products Produced by Aseptic Process-
`ing 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 Par-
`enteral 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 compli-
`ance has been adopted in Europe, Japan, and the United States [4, 5] . Sterile prod-
`ucts, 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 (typi-
`cally for large - volume parenteral production), down to less than 50 mL for radio-
`pharmaceuticals 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 sensi-
`tive 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 gener-
`ally mobile and positioned in individual processing booths or rooms as needed.
`
`Novartis Exhibit 2336.0015
`Regeneron v. Novartis, IPR2021-00816
`
`

`

`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 compound-
`ing 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 clean-
`ing of vessels prior to reuse, but confi rmation of material compatibility is required.
`Disposable equipment is often used with products manufactured in small to moder-
`ate 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 (technolo-
`gies 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 requir-
`ing 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 process-
`ing 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 pro-
`duction 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
`
`Novartis Exhibit 2336.0016
`Regeneron v. Novartis, IPR2021-00816
`
`

`

`PROCESS SELECTION AND DESIGN
`
`103
`
`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, dia-
`phragms, 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 clo-
`sures 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 con-
`tamination 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 continu-
`ous 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 post-
`processing. This is generally accomplished by short - term storage in an ISO 5 envi-
`ronment often accompanied by covering within a lidded tray. There are suppliers
`that offer depyrogenated glass vials and partially assembled syringes in sealed pack-
`ages for fi lling at a customer ’ s site. In this instance, the supplier assumes responsibil-
`ity 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 γ irradiation is accom-
`plished 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 per-
`formed in house, though many common components are becoming available prest-
`erilized 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

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket