throbber
Pharmaceutical
`
`Dosage Forms:
`
`Parenteral Medications
`
`Volume 2: Facility Design;
`Sterilization ond Processing
`
`John D. Ludwig
`
`, f
`Enigma
`
`Edited by
`
`Sandeep Nema
`
`Pu.e,""' ..I. V.L ...,.-.,,v
`Pufiw I. UL "IAIV
`
`Regeneron Exhibit 1016.001
`Regeneron Exhibit 1016.001
`
`

`

`Pharmaceutical Dosage
`Forms
`
`Regeneron Exhibit 1016.002
`
`

`

`This page intentionally left blank
`
`Regeneron Exhibit 1016.003
`
`

`

`

`

`First published in 1984 by Marcel Dekker, Inc., New York, New York.
`This edition published in 2010 by Informa Healthcare, Telephone House, 69 77 Paul Street, London EC2A
`4LQ, UK.
`
`Simultaneously published in the USA by Informa Healthcare, 52 Vanderbilt Avenue, 7th Floor, New York,
`NY 10017, USA.
`
`Informa Healthcare is a trading division of Informa UK Ltd. Registered Office: 37 41 Mortimer Street,
`London WlT 3JH, UK. Registered in England and Wales number 1072954.
`
`?::2010 Informa Healthcare, except as otherwise indicated
`
`No claim to original U.S. Government works
`
`Reprinted material is quoted with permission. Although every effort has been made to ensure that all
`owners of copyright material have been acknowledged in this publication, we would be glad to
`acknowledge in subsequent reprints or editions any omissions brought to our attention.
`
`All rights reserved. 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, or otherwise,
`unless with the prior written permission of the publisher or in accordance with the provisions of the
`Copyright, Designs and Patents Act 1988 or under the terms of any licence permitting limited copying
`issued by the Copyright Licensing Agency, 90 Tottenham Court Road, London WlP OLP, UK, or the
`Copyright Clearance Center, Inc., 222 Rosewood Drive, Danvers, MA 01923, USA (http:/ /www.
`copyright.com/ or telephone 978 750 8400).
`
`Product or corporate names may be trademarks or registered trademarks, and are used only for
`identification and explanation without intent to infringe.
`
`This book contains information from reputable sources and although reasonable efforts have been made
`to publish accurate information, the publisher makes no warranties (either express or implied) as to the
`accuracy or fitness for a particular purpose of the information or advice contained herein. The publisher
`wishes to make it clear that any views or opinions expressed in this book by individual authors or
`contributors are their personal views and opinions and do not necessarily reflect the views/ opinions of
`the publisher. Any information or guidance contained in this book is intended for use solely by medical
`professionals strictly as a supplement to the medical professional's own judgement, knowledge of the
`patient's medical history, relevant manufacturer's instructions and the appropriate best practice
`guidelines. Because of the rapid advances in medical science, any information or advice on dosages,
`procedures, or diagnoses should be independently verified. This book does not indicate whether a
`particular treatment is appropriate or suitable for a particular individual. Ultimately it is the sole
`responsibility of the medical professional to make his or her own professional judgements, so as
`appropriately to advise and treat patients. Save for death or personal injury caused by tl1e publisher's
`negligence and to the fullest extent otherwise permitted by law, neither the publisher nor any person
`engaged or employed by the publisher shall be responsible or liable for any loss, injury or damage caused
`to any person or property arising in any way from the use of this book.
`
`A CIP record for this book is available from the British Library.
`
`Library of Congress Cataloging in Publication Data available on application
`
`ISBN 13: 9781420086454
`ISBN 13: 9781420086539 (three volume set)
`
`Orders may be sent to: Informa Healthcare, Sheepen Place, Colchester, Essex C03 3LP, UK
`Telephone: +44 (0)20 7017 5540
`Email: CSDhealthcarebooks@informa.com
`Website: http:/ /informahealthcarebooks.com/
`
`For corporate sales please contact: CorporateBooksIHC@informa.com
`For foreign rights please contact: RightsIHC@informa.com
`For reprint permissions please contact: PermissionsIHC@informa.com
`
`Typeset by MPS Limited, A Macmillan Company
`Printed and bound in Tndia
`
`Regeneron Exhibit 1016.005
`
`

`

`We dedicate this work to those who have inspired us.
`To my parents Walter and Ruth Ludwig and my wife Sue Ludwig
`To my parents Hari and Pratibha Nema and my wife Tina Busch Nema
`
`Regeneron Exhibit 1016.006
`
`

`

`This page intentionally left blank
`
`Regeneron Exhibit 1016.007
`
`

`

`Foreword
`
`I was a faculty member at the University of Tennessee and a colleague of Dr. Kenneth Avis
`when he conceived, organized, and edited (along with H.A. Lieberman and L. Lachman) the
`first edition of this book series that was published in 1984. It was so well received by the
`pharmaceutical science community that an expanded three-volume second edition was
`published in 1992. Dr. A vis did not survive long enough to oversee a third edition, and it was
`questionable whether a third edition would ever be published until two of his graduate
`students, Drs. Nema and Ludwig, took it upon themselves to carry on Dr. Avis' tradition.
`Their oversight of this third edition is work that their mentor would be highly pleased
`and proud of. From 29 chapters in the second edition to 43 chapters in this new edition, this
`three-volume series comprehensively covers both the traditional subjects in parenteral science
`and technology as well as new and expanded subjects. For example, separate chapter topics in
`this edition not found in previous editions include solubility and solubilization, depot delivery
`systems, biophysical and biochemical characterization of peptides and proteins, container(cid:173)
`closure integrity testing, water systems, endotoxin testing, focused chapters on different
`sterilization methods, risk assessment in aseptic processing, visual inspection, advances in
`injection devices, RNAi delivery, regulatory considerations for excipients, techniques to
`evaluate pain on injection, product specifications, extractables and leachables, process
`analytical technology, and quality by design.
`The editors have done an outstanding job of convincing so many top experts in their
`fields to author these 43 chapters. The excellent reputations of the authors and editors of this
`book will guarantee superb content of each chapter. There is no other book in the world that
`covers the breadth and depth of parenteral science and technology better than this one. In my
`opinion, the editors have achieved their primary objectives publishing a book that contains
`current and emerging sterile product development and manufacturing information, and
`maintaining the high standard of quality that readers would expect.
`
`Michael J. Akers
`Baxter BioPharma Solutions
`Bloomington, Indiana, U.S.A.
`
`Regeneron Exhibit 1016.008
`
`

`

`This page intentionally left blank
`
`Regeneron Exhibit 1016.009
`
`

`

`Preface
`
`Pharmaceutical Dosage Forms: Parenteral Medications was originally published in 1984 and
`immediately accepted as a definitive reference in academic institutions and the pharmaceutical
`industry. The second edition was published in 1993. The ensuing years have produced
`incredible technological advancement. Classic small-molecule drugs are now complemented
`by complex molecules such as monoclonal antibodies, antibody fragments, aptamers,
`antisense, RNAi therapeutics, and DNA vaccines. There have been significant innovations in
`delivery devices, analytical techniques, in-silico modeling, and manufacturing and control
`technologies. In addition, the global regulatory environment has shifted toward greater
`emphasis on science-based risk assessment as evidenced by the evolving cGMPs, quality by
`design (QbD), process analytical technology (PAT), continuous processing, real time release,
`and other initiatives. The rapidly changing landscape in the parenteral field was the primary
`reason we undertook the challenging task of updating the three volumes. Our objectives were
`to (i) revise the text with current and emerging sterile product development and
`manufacturing science and (ii) maintain the high standard of quality the readers expect.
`The third edition not only reflects enhanced content in all the chapters, but also more
`than half of the chapters are new underscoring the rapidly advancing technology. We have
`divided the volumes into logical subunits volume 1 addresses formulation and packaging
`aspects; volume 2, facility design, sterilization and processing; and volume 3, regulations,
`validation and future directions. The authors invited to contribute chapters are established
`leaders with proven track records in their specialty areas. Hence, the textbook is authoritative
`and contains much of the collective experience gained in the (bio)pharmaceutical industry over
`the last two decades. We are deeply gratefiil to all the authors who made this work possible.
`Volume 1 begins with a historical perspective of injectable drug therapy and common
`routes of administration. Formulation of small molecules and large molecules is presented in
`depth, including ophthalmic dosage forms. Parenteral packaging options are discussed
`relative to glass and plastic containers, as well as elastomeric closures. A definitive chapter is
`provided on container closure integrity.
`Volume 2 presents chapters on facility design, cleanroom operations, and control of the
`environment. A chapter discussing pharmaceutical water systems is included. Key quality
`attributes of sterile dosage forms are discussed, including particulate matter, endotoxin, and
`sterility testing. The most widely used sterilization techniques as well as processing
`technologies are presented. Volume 2 concludes with an in-depth chapter on lyophilization.
`Volume 3 focuses on regulatory requirements, risk-based process design, specifications,
`QbD, and extractables/leachables. In addition, we have included chapters on parenteral
`administration devices, siRNA delivery systems, injection site pain assessment, and control,
`PAT, and rapid microbiology test methods. Volume 3 concludes with a forward-looking
`chapter discussing the future of parenteral product manufacturing.
`These three volumes differ from other textbooks in that they provide a learned review on
`developing parenteral dosage forms for both small molecules and biologics. Practical guidance
`is provided, in addition to theoretical aspects, for how to bring a drug candidate forward from
`discovery, through preclinical and clinical development, manufacturing, validation, and
`eventual registration.
`The editors wish to thank Judy Clarkston and Lynn O'Toole-Bird (Pfizer, Inc.) for their
`invaluable assistance and organizational support during this project, and Sherri Niziolek and
`Bianca Turnbull (Informa Healthcare) for patiently leading us through the publishing process.
`
`Regeneron Exhibit 1016.010
`
`

`

`X
`
`PREFACE
`
`We also acknowledge the assistance of Pfizer, Inc. colleagues Lin Chen and Min Huang for
`reviewing several of the chapters.
`We would like to express special gratitude to the late Kenneth E. Avis (University of
`Tennessee College of Pharmacy) for his dedication to teaching and sharing practical
`knowledge in the area of parenteral medications to so many students over the years,
`including us. Finally, we acknowledge the contributions of Dr Avis, Leon Lachman, and
`Herbert A. Lieberman who edited the earlier editions of this book series.
`
`Sandeep Nema
`John D. Ludwig
`
`Regeneron Exhibit 1016.011
`
`

`

`Contents
`
`Foreword Michael J. Akers
`Preface
`ix
`Contributors
`
`xiii
`
`vii
`
`1
`1. Aseptic manufacturing facility design
`Mark Caldwell, Bob Helt, Beth Holden, Francesca McBride, and Kevi11 Schreier
`
`2. Personnel and their impact on clean room operations
`Jeanne Moldrnhauer
`
`56
`
`3. The fundamentals of an environmental control program
`William H. Miele
`
`80
`
`4. Water systems for parenteral facilities
`Joseph ]. Manfredi
`
`91
`
`5. Particulate matter: subvisible
`D. Scott Aldrich
`
`114
`
`6. Endotoxin testing
`Michael E. Dawson
`
`146
`
`7. The compendia! sterility tests
`Scott V. W. Sutton
`
`187
`
`8.
`
`Industrial sterilization technologies: principles and overview
`Anne F. Booth
`
`195
`
`9. Steam sterilization
`James Agalloco
`
`221
`
`10. Gas, vapor, and liquid chemical sterilization
`James Agalloco
`
`241
`
`11. Dry heat depyrogenation and sterilization
`Deborah Havlik a11d Kevin Trupp
`
`255
`
`268
`12. Radiation sterilization
`Barry P. Fairand and Dusan Razem
`
`Regeneron Exhibit 1016.012
`
`

`

`xii
`
`CONTENTS
`
`297
`13. Filters and filtration
`Maik W. Jornitz and Theodore H. Meltzer
`
`334
`14. Processing of small volume parenterals and large volume parenterals
`Donald A. Eisenhauer, Roland Schmidt, Christine Martin, and Steven G. Schultz
`
`15. Freeze-drying: principles and practice
`Steven L. Nail and Larry A. Gatlin
`
`353
`
`Tndex
`
`.38.3
`
`Regeneron Exhibit 1016.013
`
`

`

`Contributors
`
`James Agalloco Agalloco & Associates, Belle Mead, New Jersey, U.S.A.
`
`D. Scott Aldrich Ultramikro, LLC, Richland, Michigan, U.S.A.
`
`Anne F. Booth Booth Scientific, Inc., Hendersonville, North Carolina, U.S.A.
`
`Mark Caldwell Jacobs Engineering Group, Inc., Conshohocken, Pennsylvania,
`U.S.A.
`
`Michael E. Dawson Associates of Cape Cod, Inc., Falmouth, Massachusetts, U.S.A.
`
`Donald A. Eisenhauer Abbott Laboratories, North Chicago, Illinois, U.S.A.
`
`Barry P. Fairand Sterigenics International, Dublin, Ohio, U.S.A.
`
`Larry A. Gatlin PK, LLC, Groton, Connecticut, U.S.A.
`
`Deborah Havlik R&D Microbiology, Hospira, Inc., Lake Forest, Illinois, U.S.A.
`
`Bob Helt Jacobs Engineering Group, Inc., Conshohocken, Pennsylvania, US.A.
`
`Beth Holden Jacobs Engineering Group, Inc., Conshohocken, Pennsylvania, U.S.A.
`
`Maik W. Jornitz Sartorius Stedim North America, Inc., Bohemia, New York, U.S.A.
`
`Joseph J. Manfredi GMP Systems, Inc., Fairfield, New Jersey, U.S.A.
`
`Christine Martin Abbott GmbH & Co. KG, Ludwigshafen, Germany
`
`Francesca McBride Jacobs Engineering Group, Inc., Conshohocken, Pennsylvania, U.S.A.
`
`Theodore H. Meltzer Capitola Consultancy, Bethesda, Maryland, U.S.A.
`
`William H. Miele Pfizer Global Manufacturing, Kalamazoo, Michigan, U.S.A.
`
`Jeanne Moldenhauer Excellent Pharma Consulting, Mundelein, Illinois, U.S.A.
`
`Steven L. Nail Baxter Pharmaceutical Solutions, Bloomington, Indiana, U.S.A.
`
`Dusan Razem Ruder Boskovic Institute, Zagreb, Croatia
`
`Roland Schmidt Abbott GmbH & Co. KG, Ludwigshafen, Germany
`
`Kevin Schreier Jacobs Engineering Group, Inc., Conshohocken, Pennsylvania, U.S.A.
`
`Regeneron Exhibit 1016.014
`
`

`

`xiv
`
`CONTRIBUTORS
`
`Steven G. Schultz Abbott Laboratories, North Chicago, Illinois, U.S.A.
`
`Scott V. W. Sutton Microbiology Network, Inc., Rochester, New York, U.S.A.
`
`Kevin Trupp Global Sterilization Engineering, Hospira, Inc., Lake Forest, Illinois, U.S.A.
`
`Regeneron Exhibit 1016.015
`
`

`

`Aseptic manufacturing facility design
`
`Mark Caldwell, Bob Helt, Beth Holden, Francesca McBride, and Kevin Schreier
`
`INTRODUCTION
`Sterile products may be in liquid or powder form (among others) as drug products and may be
`presented in formats including ampoules, vials, prefilled syringes, presterilized bottles, and
`blow-fill-seal containers. Product form and presentation influence processing conditions,
`equipment selection, and therefore, facility design. The sterile envelope refers to all the steps
`carried out during and following the final sterile filtration step through process completion,
`which occurs after filled product containers are sealed and a risk of environmental
`contamination to the product is eliminated. These steps include:
`
`• Adjuvant, buffer and media formulation
`• Addition of excipients
`• Adjustment of concentration to achieve target potency
`• Sterile filtration
`• Component preparation
`• Filling, stoppering/plugging, and sealing of product in final dosage containers
`
`The design of the facility must meet all applicable regulatory guidelines, and meet GMP
`and safety guidelines. Current Good Manufacturing Practice (GMP) requires that areas of
`operation used for aseptic processing must prevent contamination from particles and microbes
`that may be present in the air, on product contact surfaces, or shed from personnel (1 5).
`When processing biological products, such as live virus vaccines, attenuated vaccines
`and viral vectors, the biohazard nature of these products place extra demands on the facility.
`Potent compounds, like some biological products, also pose a risk to the operator and
`environment. Therefore, the facility and process design must also ensure both product and
`personnel safety.
`This chapter establishes a basis for compliance with the global regulatory expectations
`for facility design, equipment interfaces, and utility requirements applicable to sterile
`processing and the manufacture of sterile products (6 10).
`
`FACILITY DESIGN DRIVERS
`As each facility is being designed, process requirements specific to each product must be
`considered. Each different type of product has different facility needs. Also, the number of
`products to be manufactured and the production campaign strategy will impact the facility
`design.
`
`Product Types
`Chemical Bulk Drug Substances (API)
`Sterile chemical bulk drug substances are derived from chemical reactions. Facilities producing
`sterile API will be required to provide protection of the product during synthesis, isolation,
`and bulk filling. An adjuvant produced by precipitation is an example of a sterile APL
`
`Potent Compounds
`Potent compounds are classified as those chemical drug substances that are considered to be
`toxic to humans when exposure limits are exceeded, and may cause allergic reactions, birth
`defects, cancer, or other conditions. For this reason, it is required to ensure protection of
`operators working with potent compounds, ensure containment of all operations, and
`prevent release of products into the environment. It is acceptable to permit production of
`potent compounds in multiproduct facilities, provided the suite is segregated from other
`operations.
`
`Regeneron Exhibit 1016.016
`
`

`

`2
`
`VOLUME 2: FACILITY DESIGN, STERILIZATION AND PROCESSING
`
`Following filling, it is recommended to wash the exterior of vials produced in potent
`compound facilities to limit uncontrolled exposure to the product during downstream
`operations.
`
`Antibiotics
`Antibiotics are drugs produced to treat bacterial or fungal infections. Antibiotics are considered
`to be sensitizers and can generate mild to severe allergic reactions in patients and operators. It is
`required to segregate production operations from personnel outside the production area.
`Campaigns of antibiotic must be segregated from other products, as the potential for cross(cid:173)
`contamination between products can occur. In addition, ~-lactam (penicillin) and nonpenicillin
`based (cephalosporin) antibiotic products are not permitted to be produced in the same facility,
`as there is evidence that intolerance can occur for one antibiotic type, and not another.
`To accomplish this segregation, it is a requirement that a separate dedicated suite be
`constructed for each antibiotic family. This suite can be housed inside a common structure
`with other functions. At no time should antibiotic production personnel come into contact with
`personnel operating in media or fermentation areas while gowned.
`Following filling, it is recommended to wash the exterior of vials produced in antibiotic
`facilities to limit uncontrolled exposure to the product during downstream operations.
`
`Biological Product
`This category includes therapeutic proteins generated by fermentation or cell culture and
`inactivated vaccines. The facility is to be designed in same way as API production, except that
`terminal sterilization is often not feasible, due to the fragility of the product.
`
`Live Virus Vaccines
`Vaccines containing a live virus, or viral vector, must be designed to provide containment of
`the organism to protect both operators and the environment. Viral vectors are virus-like
`particles that inject genetic material into the cells of the organism being treated and are treated
`in a similar manner to live viruses. The biosafety level designated for the organism will drive
`decisions regarding the level of environmental controls that is required for the facility.
`Typically, these products require the use of adjuvants and will be suspension products,
`incapable of being filter sterilized. Products cannot be terminally sterilized. Live virus vaccine
`products can be campaigned in a multiproduct facility, as long as the vaccine production area
`is segregated from the remainder of the facility. Following the completion of the live virus
`campaign, the suite will need to be completely decontaminated prior to use for another
`product.
`
`Facility Types
`Single Product, Dedicated
`This facility is designed to produce a single product at any one time, throughout the year,
`without concern for cross-contamination with a second product. The facility can be operated to
`produce multiple products in a series of campaigns, converting between products.
`
`Multiproduct Multisuite
`This facility is designed to produce multiple products simultaneously in multiple sterile suites.
`Sterile operations in each suite are to be segregated from one another to ensure that cross(cid:173)
`contamination is prevented. It is a recommendation to clean and decontaminate any used
`components or equipment prior to exiting the suite and entering the return corridor.
`
`Production Area Description
`Conventional Aseptic Technology: Open
`In conventional aseptic processing, the product is exposed to the room environment during
`operation. For this reason, aseptic operations are required to be performed under ISO 5
`
`Regeneron Exhibit 1016.017
`
`

`

`ASEPTIC MANUFACTURING FACILITY DESIGN
`
`3
`
`conditions, by sufficiently gowned operators, trained in aseptic technique. Sterility assurance
`levels for aseptic operations, including filling of vials or syringes, can be maximized through
`the use of barriers such as restricted access barrier systems or isolators to limit the size of the
`aseptic environment and remove operators from the ISO 5 fill area.
`
`Terminally Sterilized Product
`Whenever possible, it is required to terminally sterilize filled units of product. Application of
`an "overkill" sterilization methodology can provide a sterility assurance level of 10-6
`, or better.
`Sterilization can be by steam, dry heat, gas, or radiation.
`
`Restricted Access Barrier: Open and Closed
`As defined by ISPE: A restricted access barrier system (RABS) is an advanced aseptic
`processing system that can be utilized in many applications in a fill-finish area. RABS provides
`an enclosed environment to reduce the risk of contamination to product, containers, closures,
`and product contact surfaces compared to the risks associated with conventional clean
`room operations. RABS can operate as "doors closed" for processing with very low risk of
`contamination similar to isolators, or permit rare "open door interventions" provided
`appropriate measures are taken (11).
`In representative installations recently constructed, there is a wide variety of equipment
`configurations referred to as RABSs. In construction detail, some approximate isolators in their
`level of segregation of the clean environment from the background environment, while other
`examples employ less rigorous segregation between the clean environment and the
`background room.
`When barrier doors are required to be opened, the opening must be protected within an
`ISO 5 zone, to the extent necessary to ensure continuous ISO 5 coverage. The door must be
`permitted to be opened without leaving the ISO 5 zone.
`
`Isolation Technology
`As defined by ISPE: An isolator is a leak-tight enclosure designed to protect operators from
`hazardous/potent processes or protect processes from people or detrimental external environ(cid:173)
`ments or both. A basic enclosure consists of a shell, viewing window, glove/ sleeve assemblies,
`supply and exhaust filters, light (s), gauge (s), input and output openings (equipment door
`airlocks, RTPs, etc.), and various other penetrations. There are two types of isolators:
`
`Closed isolators.
`Isolators operated as closed systems do not exchange unfiltered air or
`contaminants with adjacent environments. Their ability to operate without personnel access to
`the critical zone makes isolators capable of levels of separation between the internal and
`external environment unattainable with other technologies. Because the effectiveness of this
`separation, closed isolators are ideally suited for application in the preparation of sterile
`and/or toxic material. Aseptic and containment isolators are two types of closed isolators.
`
`Open isolators. Open isolators differ from closed isolators in that they are designed to allow
`for the continuous or semicontinuous egress of materials during operation while maintaining a
`level of protection over the internal environment. Open isolators are decontaminated while
`closed, and then opened during manufacturing. Open isolators typically are used for the
`aseptic filling of finished pharmaceuticals (11).
`
`Closed Processing
`Closed system sterile processing. A closed system is one that does not contain any open
`aseptic manipulations or interventions by design or operation and does not allow microbial
`ingress. Validated sterilization cycles must be provided. The product is separated from the
`surrounding room environment by a sterilizing grade vent filters. Leak testing must occur pre(cid:173)
`and post use to demonstrate the system integrity.
`
`Regeneron Exhibit 1016.018
`
`

`

`4
`
`VOLUME 2: FACILITY DESIGN, STERILIZATION AND PROCESSING
`
`EQUIPMENT AND PROCESS SYSTEM IMPACT ON FACILITY
`It is important to start the facility design with an understanding of each process step involved
`with the manufacture of a sterile product. In this section, an overview is provided for some of
`the more common process steps, including a description of the major equipment, material
`flows, and facility impacts.
`
`Nonactive Materials
`Nonactive materials are transported from the warehouse to the weigh/dispense area, where
`they are dispensed into containers under a hood with high efficiency particulate air (HEP A)
`filtration. The hoods used in the dispensing operation shall be designed for protection of the
`product and may also need to protect the operator from exposure to potentially hazardous
`materials used in the formulation process.
`The number of hoods, and type, will be determined by evaluating the number of
`weighing operations that must be performed, the size of the weighing operations, the
`compatibility of materials that must be weighed, and any special ergonomic or personnel
`safety concerns. Materials that are not compatible may need to be dispensed in separate hoods
`to prevent any cross-contamination concerns. Also, dispensing operations involving large bulk
`containers will require either a lift assist or pallet jack access to prevent operator injury,
`requiring the hood to be designed as a walk-in type.
`Hoods being designed for product protection typically recirculate air back to the
`dispensing room to reduce the HV AC consumption for the area. However, when weighing
`hazardous materials, the air from the hood may need to be captured and exhausted to the roof
`through some type of environmental control device. Hoods of this type are typically designed
`to be negatively pressurized with respect to the dispensing room.
`Containers may be either single-use or reusable. Single-use containers are disposable.
`Reusable containers are required to be tracked and controlled to prevent cross-contamination
`of clean and used containers. Prior to reuse, used containers should be brought to a parts
`preparation washroom for cleaning and then placed in controlled storage.
`Nonactive materials are dispensed into bags or plastic bottles and can be placed into
`plastic bins as part of preassembled kits. Preweighed nonactive materials are stored as kits and
`staged until they are ready to be transferred to the formulation area. Identifying labels should
`be placed on the containers.
`It is typical to provide a local WFI drop feeding a sink in the weigh/ dispense area, with
`WFI temperature controlled by a local WFI drop cooler. The WFI drop is periodically flushed
`and sampled per SOP. WFI is used to prepare solutions of nonactive materials in bottles. This
`operation may be performed with equipment such as a laboratory agitator.
`Bench scales are used for dispensing of smaller-scale materials. Floor scales are used for
`larger quantities. Balances and measuring equipment should be of an appropriate range and
`precision.
`
`Active Materials
`Receipt/Storage
`Active materials, API or drug substance (DS), may be received in a wide variety of container
`types, in either frozen or liquid form, or as a solid. Careful consideration should be given to the
`form and container type, since this affects the storage, transport, preparation, and handling of
`APL The following list is provided to indicate the diversity of some common examples.
`
`1. Frozen in cryovessels, ranging in size from 50 to 300 L
`2. Frozen in small containers, ranging in size from 1 to 20 L
`3. As liquid in small containers, ranging in size from <1 to 20 L
`4. As powder in canisters, ranging in size from <1 to 50 kg
`
`Weighing and dispensing can occur for either sterile or nonsterile material.
`It is recommended that sterile material be dispensed in an isolator to prevent
`contamination. The isolator should be fitted with a rapid decon antechamber to facilitate the
`addition of product and containers to be dispensed.
`
`Regeneron Exhibit 1016.019
`
`

`

`

`

`6
`
`VOLUME 2: FACILITY DESIGN, STERILIZATION AND PROCESSING
`
`Thawing
`If API is received in frozen form, an area adjacent to the formulation area should be designated
`as an API thaw room. Sufficient space shall be provided, depending on the API production
`requirements and duration of the thaw cycle.
`If frozen in cryovessels, ample thaw/shaker stations shall be provided for the required
`number of cryovessels. At each station, a shaker mechanism is provided to gently mix the API
`during the thawing process. The thaw module will allow for precise control of the temperature
`profile during the process.
`If frozen in small containers, consideration will be given to the selection of the thawing
`equipment. A shaker thaw bath will thaw several containers quickly, but needs a water supply.
`An environmental chamber will also thaw several bottle, but the operation will be performed
`at a much slower rate, due to the lower heat transfer rate of the air. The number of units will be
`determined by the quantity of bottles to be thawed for each batch, and the time allotted to
`perform the operation. If there are concerns regarding the accumulated time out of
`refrigeration, more units may be required.
`Local freezers and refrigerators should be provided in the thaw area to provide staging
`space for material that is awaiting the thaw process as well as thawed material that is awaiting
`transport to the formulation room. The refrigerators and freezers should employ some system
`for segregation to maintain lot-to-lot integrity of the same material as well as containers of
`different products, avoiding any potential cross-contamination and product loss.
`If live virus product is thawed, secondary containment is required during the thawing
`operation in the event of a container rupture. This can be accomplished by use of an overwrap
`or use of the thaw bath as secondary containment. Any use of water as a thawing medium for
`live virus should be routed to biowaste collection.
`
`Formulation
`The typical formulation module includes one formulation tank, positioned inside the
`formulation room, adjacent to a utility panel on the wall. Piping, electrical and instrumentation
`for the tank are connected to the utility panel. This tank has been cleaned via CIP and has been
`sanitized by clean steam. Following the sanitization operation, it has been maintained under
`positive pressurize with filtered process air.
`Room height should account for elevation of equipment, including open charging
`operations, agitator clearance, and vent filters. Portable equipment must be able to fit under
`the door frame without disassembly. Room lay

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