Download full The challenge of cmc regulatory compliance for biopharmaceuticals, 4th 4th edition joh

Page 1


Visit to download the full and correct content document: https://ebookmass.com/product/the-challenge-of-cmc-regulatory-compliance-for-bioph armaceuticals-4th-4th-edition-john-geigert/

More products digital (pdf, epub, mobi) instant download maybe you interests ...

Human Metabolism: A Regulatory Perspective 4th Edition

https://ebookmass.com/product/human-metabolism-a-regulatoryperspective-4th-edition/

The xVA Challenge : Counterparty Risk, Funding, Collateral, Capital and Initial Margin 4th Edition Jon Gregory

https://ebookmass.com/product/the-xva-challenge-counterpartyrisk-funding-collateral-capital-and-initial-margin-4th-editionjon-gregory/

Fundamentals of Phonetics: A Practical Guide for Students (4th Edition ) 4th…

https://ebookmass.com/product/fundamentals-of-phonetics-apractical-guide-for-students-4th-edition-4th/ Oxford Handbook of Urology 4th Edition Edition John Reynard

https://ebookmass.com/product/oxford-handbook-of-urology-4thedition-edition-john-reynard/

Synopsis Of Clinical Opthalmology 4th Edition John R. Salmon

https://ebookmass.com/product/synopsis-of-clinicalopthalmology-4th-edition-john-r-salmon/

Kanski's Synopsis of Clinical Ophthalmology 4th Edition

John F.Salmon

https://ebookmass.com/product/kanskis-synopsis-of-clinicalophthalmology-4th-edition-john-f-salmon/

Introduction to Toxicology 4th Edition John Timbrell

https://ebookmass.com/product/introduction-to-toxicology-4thedition-john-timbrell/

Fundraising For Dummies, 4th Edition Dr. Beverly A. Browning

https://ebookmass.com/product/fundraising-for-dummies-4thedition-dr-beverly-a-browning/

A Book of Visual Signalwave 4th Edition John D. Riselvato

https://ebookmass.com/product/a-book-of-visual-signalwave-4thedition-john-d-riselvato/

John Geigert

The Challenge of CMC Regulatory Compliance for Biopharmaceuticals

Fourth Edition

The Challenge of CMC Regulatory Compliance for Biopharmaceuticals

The Challenge of CMC Regulatory Compliance for Biopharmaceuticals

Fourth Edition

Carlsbad, CA, USA

ISBN 978-3-031-31908-2 ISBN 978-3-031-31909-9 (eBook) https://doi.org/10.1007/978-3-031-31909-9

© The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Nature Switzerland AG 2004, 2013, 2019, 2023

This work is subject to copyright. All rights are solely and exclusively licensed by the Publisher, whether the whole or part of the material is concerned, specifcally the rights of translation, reprinting, reuse of illustrations, recitation, broadcasting, reproduction on microflms or in any other physical way, and transmission or information storage and retrieval, electronic adaptation, computer software, or by similar or dissimilar methodology now known or hereafter developed.

The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication does not imply, even in the absence of a specifc statement, that such names are exempt from the relevant protective laws and regulations and therefore free for general use.

The publisher, the authors, and the editors are safe to assume that the advice and information in this book are believed to be true and accurate at the date of publication. Neither the publisher nor the authors or the editors give a warranty, expressed or implied, with respect to the material contained herein or for any errors or omissions that may have been made. The publisher remains neutral with regard to jurisdictional claims in published maps and institutional affliations.

This Springer imprint is published by the registered company Springer Nature Switzerland AG The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland

Preface

Preparing the fourth edition of my book was again a most humbling experience for me. My primary purpose for updating the previous edition was to continue to provide relevant insight and practical suggestions for a common sense, cost-effective, risk-managed approach to meeting the Chemistry, Manufacturing and Controls (CMC) regulatory compliance requirements and expectations for biopharmaceuticals as human medicinal products. But the scope of this approach was almost overwhelming as there was so much that could not be included in this latest updated edition. I trust that my choices will be of the most beneft to the upcoming next generation of Project Management, Process Development, Manufacturing, Analytical Development, Quality Control, Quality Assurance, and Regulatory Affairs staff who take responsibility for ensuring the quality, effcacy, and safety of these biopharmaceutical medicines for patients.

So many changes continue in the advancement of the protein-based biopharmaceuticals. Over 250 recombinant proteins, monoclonal antibodies, bispecifc antibodies, fusion proteins, antibody Fab and Fc fragments, antibody-drug conjugates (ADCs) are now in the marketplace in both the United States of America (USA) and European Union (EU). And not to forget that over 80 biosimilars are also now available across all major classes of off-patent recombinant proteins and monoclonal antibodies.

The increasing entrance into the marketplace over the past 5 years of the gene therapy-based biopharmaceuticals. Using genetically engineered viruses, genes (DNA) are delivered either directly to the patient or to collected patient cells that then get administered back to the patient, so that the patient becomes the “bioreactor” producing the needed protein product. About 20 gene therapy-based biopharmaceuticals are now market-approved, with an estimate of about 10 new therapies to be approved every year going forward.

Another measure of the rapid degree of change occurring in the biopharmaceutical feld is refected in over 400 CMC regulatory compliance references listed in this book that were either issued or updated since the release of the last edition.

I am indebted to two major regulatory authorities: the United States Food and Drug Administration (FDA) and the European Medicines Agency (EMA). These

two regulatory authorities publish on their websites an abundance of CMC regulatory guidances (recommendations and expectations) to help the developing and changing biopharmaceutical industry. In addition, they both upload (after a biopharmaceutical medicine is market-approved) discussions, reviews, and meeting minutes during the biopharmaceutical review process, thus providing insights into how the regulatory authority currently views the current application of manufacturer’s biopharmaceutical CMC regulatory compliant strategy. Many of the references listed in this book are from this information that is readily downloaded from their websites. Thanks also goes to the International Council of Harmonisation (ICH), in their harmonizing of consensus CMC guidelines. ICH has issued harmonized CMC guidelines not only for the content to be included in biopharmaceutical submissions seeking market approval (e.g., for viral safety evaluation, comparability of biopharmaceuticals after a manufacturing process change, etc.) but also for the strategic control for the complex biopharmaceutical manufacturing processes (e.g., for pharmaceutical development, quality risk management, pharmaceutical quality system, etc.). While the initial focus for ICH was on the protein-based biopharmaceuticals, as their guidances are updated, aspects for the gene-therapy biopharmaceuticals are being included. The ICH regulatory guidelines have driven the biopharmaceutical industry to a higher standard of manufacturing excellence and quality control, introducing the principles of Quality by Design (QbD), Quality Risk Management (QRM), Pharmaceutical Quality Systems (PQS), Knowledge Management (KM), Established Conditions (EC), and Post-Approval Change Management Protocols (PACMPs). It is for this reason that I have provided the FDA, EMA, and ICH website locations (listed at the end of each chapter) for the many regulations, guidance documents, and case examples that I have used in the preparation of this book.

Expediting clinical pathways have shortened the time in clinical development –Breakthrough Therapy designation (BTD) and Regenerative Medicines Advanced Therapy (RMAT) designation in the USA; PRIME (PRIority MEdicine) designation in the European Union. This shortening of the clinical study time from First-inHuman (FIH) entry studies to market approval (estimated to be at least a 2–3 year savings) has placed great demands upon the CMC regulatory compliance strategy with ever-decreasing time to complete all of the required development, optimization, validation, site changes, etc. for the challenging biopharmaceutical manufacturing processes and complex products. This enhanced pressure on the CMC teams has not gone unnoticed by the regulatory authorities and they are keenly aware of the potential delays in biopharmaceutical product market approval that now can be due to CMC issues. Discussion on how to effectively manage the CMC regulatory compliance strategy under clinical expedited pathways is provided in this book.

Thanks also go to the companies who stumbled in their CMC regulatory compliance strategy, resulting in delay or rejection of their biopharmaceutical, so that we can learn from their mistakes. At times, an effective CMC regulatory compliance strategy can seem like a mystery. Sometimes this mystery is self-induced in our companies, when the CMC team is not aware that an effective CMC regulatory

compliance strategy can be at hand. Through means of this fourth edition, I want to show clearly the “good news” that CMC regulatory compliance no longer has to be a mystery. But I also want to caution against the “bad news” that there can be too much CMC regulatory compliance information available, “an information overload.” This is where this book becomes invaluable (along with the help of a good CMC consultant of course) in sifting through all of the public guidance available to determine which pieces are relevant for each specifc biopharmaceutical manufacturing processes and product types. To reinforce that no commercial proprietary information is revealed in this book, I have provided internet website addresses for the comments on the various companies and their CMC biopharmaceutical issues, mentioned in this book.

Throughout this book, I use the terms “biologic” or “biological” whenever I am discussing CMC issues that apply across the board to pharmaceuticals that are (1) derived from living organisms, (2) have challenging manufacturing processes, and (3) are complex products. But, I use the terms “biopharmaceutical” or “recombinant DNA-derived” whenever I am discussing CMC issues specifc for biologics manufactured from genetically engineered living organisms.

In Chap. 1, defning the terminology used in CMC regulatory compliance of biopharmaceuticals is paramount to being able to effectively communicate not only throughout our industry but also with the regulatory authorities. Also, the increasing diversity of biopharmaceutical product types is unveiled, with a discussion of the four major “waves” of product types that have entered the marketplace from the early 1990s through today. Today, there are the protein-based biopharmaceuticals (the recombinant proteins and the monoclonal antibodies) and there are the everincreasing number of gene therapy-based biopharmaceuticals (the viral vectors and the genetically modifed patient cells). In addition, a non-viral vector (mRNA) is now being pursued in the clinic. Time will tell how abundant these human patient “bioreactors” will become. In Chap. 2, the various regulatory pathways for initiating a clinical trial, for maintaining the clinical trial during its development and then for seeking market approval for biopharmaceuticals, is examined, within the USA and European Union regions. Differences in the regulatory pathways are discussed. In Chap. 3, biopharmaceuticals are shown to be defnitely different from chemical drugs. This is not a perception, but a reality, and it is refected by the statements on regulatory authority websites and in the wording of the regulatory guidances that they issue. Differences due to starting materials, differences due to the ability to control the manufacturing process, and differences due to the molecular complexity of the products are examined across four product types. Avoidance of the word “biogeneric” for biosimilars is discussed. In Chap. 4, why the risk-based approach is absolutely necessary to effectively manage the minimum CMC regulatory compliance continuum, due to the challenge in manufacturing and complexity of the resulting biopharmaceuticals, is examined. The ICH-recommended risk-based approach for biopharmaceuticals – Quality by Design (QbD)/Quality Risk Management (QRM) – is also discussed and shown to be an invaluable tool for establishing and

maintaining adequate and appropriate control for all biopharmaceutical types. In Chap. 5, the four primary adventitious agents of concern for biopharmaceuticals are examined in detail – prions, viruses, mycoplasma, bacteria/fungi. While each manufacturing process type has a different level of risk due to adventitious agent contamination, there is no biopharmaceutical manufacturing process that carries no risk of adventitious agent contamination. In Chap. 6, the signifcant differences between the starting materials for the protein-based biopharmaceuticals (Master Cell Bank) and the many starting materials for the gene therapy-based biopharmaceuticals (i.e., the viral vectors, transduced patient cells, mRNA non-viral vector) are evaluated. A problem here can carry all the way through the manufacturing process to the fnal administered biopharmaceutical product. In Chaps. 7 and 8, the risk-based requirements and expectations for an adequate and appropriate design and control of the biopharmaceutical drug substance manufacturing stages are examined. Similarities and differences between what is expected and what is doable for the drug substance manufacture, across the four types of biopharmaceuticals (recombinant proteins and monoclonal antibodies, viral and non-viral vectors, and genetically modifed patient cells), are compared and contrasted. In Chap. 9, the risk-based requirements and expectations for an adequate and appropriate design and control of the biopharmaceutical drug product manufacturing stages – formulation, container closure, and aseptic processing of flling/sealing – are examined. Sometimes between the purifed drug substance and formulation, the drug substance is conjugated (e.g., antibody-drug conjugates, PEGylation). In Chaps. 10 and 11, compared to chemical drugs, biopharmaceuticals have a large, complex biomolecular structure, seemingly endless structural variants, and, in addition, a highly complex process-related impurity profle, primarily due to the use of living systems involved in their manufacturing process. The challenges for the physicochemical and functional characterization of the different biopharmaceutical types – recombinant proteins, monoclonal antibodies, genetically engineered viruses, genetically engineered cells, mRNA nonviral vector – are examined, along with the use of a risk-based approach for process-related impurity control (and hopefully reduction or removal through the purifcation process). In Chap. 12, it is shown that because of the size and complexity of a biopharmaceutical functional/therapeutic activity, assays are required for strength/ potency measurement. In this chapter, the three types of functional activity assays for measuring potency are examined: bioassay, surrogate, and assay matrix. In Chap. 13, the seven major categories of Critical Quality Attributes (CQAs) are explored. Specifc testing to meet the requirements of each of these quality attributes, for each biopharmaceutical type, is discussed. In Chap. 14, the art of specifcation setting for biopharmaceuticals is examined – both for the release of batches and for setting the shelf-life. The use of a risk-based approach to set the limits or ranges through clinical development and into market approval for a

biopharmaceutical is discussed. The concept of an interim regulatory specifcation for a to-be-marketed biopharmaceutical, especially when so few batches are available today to statistically set specifcations, is explored. In Chap. 15, the three riskbased concerns that need to be addressed by an effective comparability study are examined. Demonstrating comparability for a biopharmaceutical after a manufacturing process change is no easy task, whether it be for a recombinant protein, monoclonal antibody, viral vector, or genetically modifed patient cells. In Chap. 16, the critical importance of communicating with the regulatory authorities on the CMC regulatory compliance strategy is stressed. Finally, in this chapter, an encouragement is given to senior management to take advantage of CMC-focused meetings available with the regulatory authorities.

CMC regulatory compliance strategy does not determine the direction of the clinical development program; its primary purpose is to support it, but that does not mean that we in CMC should avoid the tough decisions that scientists must make when advancing the applications of genetic engineering toward human. Francis S. Collins, the former Director of the United States National Institutes of Health (NIH) and the Human Genome Project (HGP) laid out this thought for all to consider [1]:

Is the science of genetics and genomics beginning to allow us to “play God”? That phrase is the one most commonly used by those expressing concern about these advances, even when the speaker is a nonbeliever. Clearly the concern would be lessened if we could count on human beings to play God as God does, with infnite love and benevolence. Our track record is not so good. Diffcult decisions arise when a confict appears between the mandate to heal and the moral obligation to do no harm. But we have no alternative but to face those dilemmas head-on, attempt to understand all of the nuances, include the perspectives of all the stakeholders, and try to reach a consensus. The need to succeed at these endeavors is just once more compelling reason why the current battles between the scientifc and spiritual worldviews need to be resolved – we desperately need both voices to be at the table, and not to be shouting at each other.

Learning never ceases in the area of biopharmaceutical CMC regulatory compliance strategy. After 40 years in the biopharmaceutical industry, I would have thought by now that there would be “nothing new under the sun” to learn. But I am constantly amazed at the energy and creativity by my colleagues continually developing new manufacturing process technologies and new product types, which demand challenging CMC strategies to effectively manage and ensure their regulatory compliance. It is my sincere desire that this book will be of help to those who work in these biopharmaceutical companies both today and for years to come. I encourage the users of this book to seek to learn more on their own about CMC regulatory compliance strategy for biopharmaceuticals.

Carlsbad, CA, USA

John Geigert

1. Collins FS.  The language of god – a scientist presents evidence for belief. Free Press/Simon & Schuster, Inc; 2007.

Photo courtesy of Nicki Geigert Photography

Acknowledgments

Many colleagues during my 50 years of service in the biopharmaceutical industry have impacted my understanding of CMC regulatory compliance strategy and have indirectly contributed to the writing of this book. I would like to especially acknowledge my friends and colleagues at my former companies (all of which now have been acquired by larger biopharmaceutical companies and no longer exist as separate entities) – Cetus Corporation, Immunex Corporation, and IDEC Pharmaceuticals – for the insights and experiences that we shared. I would also like to acknowledge my new friends and colleagues in the many biopharmaceutical companies that I now serve as their consultant – for the many CMC regulatory compliance strategies that we wrestle with.

A special expression of appreciation goes to my wife, Nicki, who understood the time commitment and mental exhaustion that comes along with trying to update a book of this magnitude and for her patient support and strong encouragement again throughout this entire lengthy process.

Quoting from Albert Einstein, Nobel laureate in Physics, “The most beautiful thing we can experience is the mysterious. It is the source of all art and science. He to whom this emotion is a stranger, who can no longer pause to wonder and stand rapt in awe, is as good as dead; his eyes are closed.” I have been in awe watching my fellow scientists unravel the ever-increasing intricate complexity and design of life – carrying out genetic engineering on living cells to re-design them either to over-produce recombinant proteins or monoclonal antibodies or to become a living biopharmaceutical product itself such as a genetically engineered T-cell. As a scientist who believes that God is the ultimate genetic engineer, I trust that scientists will eventually comprehend and appreciate His original creative work.

Carlsbad, CA, USA

John Geigert

2.3.3

Terms: ‘Biologic’ and ‘Biological’

Terms: ‘Biopharmaceutical’ and ‘rDNA-Derived’

Terms: ‘ATMP’ and ‘CGTP’

5.4.3

5.5 Adventitious Bacteria/Fungi

5.5.1 Bacteria/Fungi Risk Assessment .

5.5.2 Bacteria/Fungi Risk Control

5.5.3 CMC Strategy Tip: Not All Discoveries Have Been Made Yet

5.6 ‘Not Detected’ Is Not Confrmation of Absence .

References

6 Starting Materials for Manufacturing the Biopharmaceutical Drug Substance

6.1 In the Beginning

6.2 Starting Material for Recombinant Proteins and Monoclonal Antibodies

6.2.1 Development Genetics – Steps Prior to Cell Banking

6.2.2 Importance of

6.2.3

6.2.4

6.2.5

6.2.6

6.2.7

6.3

7.3

Upstream Cell Culture Production of Recombinant

7.3.4 Meeting CMC Regulatory Compliance for Upstream Production

7.4 Upstream Production of Genetically Modifed Patient Cells .

7.4.1 Assembling the Transduction Process

7.4.2 Applying the Minimum CMC Regulatory Compliance Continuum

7.4.3 Meeting CMC Regulatory Compliance for Upstream Production

7.5 Upstream IVT Production of Non-Viral Vector (mRNA)

7.5.1 Assembling the Non-Viral mRNA Production Process

7.5.2 Meeting CMC Regulatory Compliance for Upstream Production

8.2

9.3 Formulation

9.3.1 Formulation of Recombinant Proteins & Monoclonal Antibodies .

9.3.2 Formulation of Gene Therapy Viral Vectors

9.3.3 Formulation of Genetically Modifed Patient Cells

9.3.4 Formulation of mRNA Non-viral Vector

9.3.5 Formulation Changes – Tread Carefully

9.4 The Container Closure System

9.4.1 Close Encounters Not Wanted: Product – Container-Closure Interactions

9.4.2 Applying the Minimum CMC Regulatory Compliance Continuum

11.3 Viral Vectors

11.3.1 Sources of Biomolecular Structural Variants

11.3.2 Issue of Empty Capsids

11.3.3 Applying the Minimum CMC Regulatory Compliance Continuum

11.4 Genetically Modifed Patient Cells

11.4.2 VCN – Not Too Much, Not Too Little

Applying the Minimum CMC Regulatory Compliance Continuum

13.5.2 Regulatory Guidance on Quantity

13.5.3 Measurement of Quantity

13.6 Safety

13.6.1 Safety from Adventitious Agents

13.6.2 Replication Competent Virus

13.7 General .

13.8 Compiled Tables of CQAs for Different Biopharmaceutical Types

13.8.1 FDA Released CQA Test Results

15.1.4 The Comparability Study Must Address Three Risk-Based Concerns .

15.2 Level of Risk Due to Stage of Clinical Development When Change Is Planned

15.3 Level of Risk Due to Nature (Type, Extent, Location) of Planned Process Change

15.3.1 Assigning Risk Levels to Proposed Manufacturing Process Changes

15.3.2 Risk Levels for Recombinant Proteins and Monoclonal Antibodies.

15.3.3 Risk Levels for Viral Vectors and Genetically Modifed Patient Cells

15.3.4 Established Conditions (ECs)

15.4 Level of Risk Due to Residual Uncertainty Still Remaining 517

15.5

List of Figures

Fig. 1.1 Amino acid sequence for the 51 amino acid recombinant human insulin

Fig. 1.2 Amino acid linear schematic for the 570 amino acid recombinant human Xenpozyme

Fig. 1.3 X-ray crystal 3-dimension structure of the 497 amino acid recombinant human Vpriv

Fig. 1.4 Two-dimensional schematic of the IgG monoclonal antibody

Fig. 1.5 Two-dimensional schematic of a bispecifc antibody

Fig. 1.6 Molecular schematic of some market-approved engineered antibody fragments.

Fig. 1.7 Molecular schematic of some commercial antibody-drug conjugates (ADCs) .

Fig. 1.8 FDA’s description of a biosimilar

Fig. 1.9 Manufacture of protein-based biopharmaceuticals (waves 1, 2, 3)

Fig. 1.10 Manufacture of gene therapy-based biopharmaceuticals (wave 4)

Fig. 1.11 Two main approaches to inserting gene therapy-base biopharmaceuticals into patients

Fig. 1.12 Schematic illustrating the structure of a recombinant AAV viral vector

Fig. 1.13 Schematic illustrating the anti-CD19 CAR transduced onto a T-cell

12

14

16

17

19

21

Fig. 2.1 The pharmaceutical regulatory system in the United States 33

Fig. 2.2 Regulatory drug development pathways in the USA

Fig. 2.3 The pharmaceutical regulatory system in the European Union

Fig. 2.4 Regulatory drug development pathway in the European Union

Fig. 3.1 Difference due to type of starting material across the four product types

Fig. 3.2 Difference due to inconsistency of manufactured product across the four product types

List of Figures

Fig. 3.3 Difference due to complexity of molecular structure across the four product types

Fig. 3.4 Illustration of size of recombinant proteins and monoclonal antibodies

Fig. 3.5 Linkage between manufacturing process and product across the four product types

Fig. 3.6 Comparison of studies for market-approval – innovator (reference) versus biosimilar

Fig. 4.1 Minimum CMC regulatory compliance continuum

Fig. 4.2 Three interactive CMC regulatory compliance components

Fig. 4.3 CMC Regulatory component

Fig. 4.4 cGMPs component

Fig. 4.5 Quality System component

Fig. 4.6 QbD/QRM risk-based approach .

Fig. 4.7 Quality target product profle (QTPP)

Fig. 4.8 Critical Quality Attribute (CQA)

Fig. 4.9 Critical Process Parameter (CPP)

Fig. 4.10 Determining a CPP versus a non-CPP by the degree of % CQA impact.

Fig. 4.11 Control Strategy (CS)

Fig. 4.12 Illustration of design space applied to an anion exchange chromatography step

Fig. 5.1 Risk mitigation/risk preparation/response recovery for a facility virus contamination 153

Fig. 6.1 Starting Materials: frst stage in the manufacture of the drug substance

Fig. 6.2 Schematic of the genetic construction process for a recombinant cell

Fig. 6.3 Schematic of the process of selecting a single recombinant cell for the MCB/WCB

Fig. 6.4 Threefold screening steps to select the desired clone

Fig. 6.5 Manufacture of the four starting materials for rAAV viral vector

Fig. 6.6 The three DNA plasmid starting materials for transient rAAV vector manufacture

Fig. 6.7 Transgene plasmid for Luxturna rAAV in vivo gene therapy

Fig. 6.8 Manufacture of the fve starting materials for rLV viral vector

Fig. 6.9 Manufacture of the linearized DNA plasmid starting material for mRNA production

Fig. 7.1 Upstream production: second stage in the manufacture of the drug substance

Fig. 7.2 Overview of upstream production processes for four types of biopharmaceuticals

Fig. 7.3 Schematic of fed-batch mode bioreactor operations

Fig. 7.4 Illustration of the recommended cell culturing conditions for genetic stability assessment

Fig. 7.5 Schematic of the viral vector upstream production process

Fig. 7.6 Schematic of the ex vivo transduction of patient cells process

Fig. 7.7 T cell selection from patient cells – before and after enrichment

Fig. 7.8 Schematic of manufactured mRNA viral vector

Fig. 8.1 Downstream purifcation: third stage in the manufacture of the drug substance

Fig. 8.2 Overview of downstream purifcation processes for four types of biopharmaceuticals

Fig. 8.3 Typical downstream purifcation process for IgG monoclonal antibodies

Fig. 8.4 Some typical CPPs for a downstream purifcation process for a monoclonal antibody

Fig. 8.5 Typical downstream purifcation process for rAAV viral vector . . . .

Fig. 8.6 Typical downstream purifcation process for mRNA non-viral vector

Fig. 9.1 Manufacture of the biopharmaceutical drug product 300

Fig. 9.2 Conjugation of recombinant proteins and monoclonal antibodies 301

Fig. 9.3 Drug-linker intermediate for manufacture of Trodelvy ADC

Fig. 9.4 Manufacture of Trodelvy ADC bulk drug substance 305

Fig. 9.5 Formulation stage in the drug product manufacturing process pathway 307

Fig. 9.6 Formulated bulk drug product in the drug product manufacturing process pathway 310

Fig. 9.7 Aseptic processing stage in the drug product manufacturing process 322

Fig. 9.8 Typical drug product manufacturing process for the four biopharmaceutical types 328

Fig. 10.1 Potential sources of process-related impurities in a biopharmaceutical process

Fig. 10.2 Illustration of the ELISA test method for residual host cell proteins

Fig. 11.1 Potential sources of biomolecular structural variants in a biopharmaceutical process 383

Fig. 11.2 N-glycan profles of two market-approved highly similar trastuzumab mAbs

Fig. 11.3 Site of disulfde scrambling in the Fc-fusion protein etanercept

Fig. 12.1 A cell proliferation inhibition cell-based bioassay for a market-approved mAb

Fig. 12.2 Functional activities across the molecular structure of rituximab, a monoclonal antibody

List of Figures

Fig. 12.3 Four mechanisms of rituximab-mediated B-cell death

Fig. 14.1 Interrelationship between the four testing categories

Fig. 14.2 Illustration of visually viewing data – not just looking at the mean values

Fig. 15.1 Risk concern increases as the stage of clinical development advances

Fig. 15.2 Risk concern increases as the perceived risk of the nature of the change increases

Fig. 15.3 Stepwise approach to reducing residual uncertainty for a process change

Fig. 16.1 Clinical development pathway milestones for regulatory approval

Fig. 16.2 Timeline commitments for PDUFA (Type B) meetings with the FDA

Fig. 16.3 Eight (8) steps for EMA scientifc advice

Fig. 16.4 Different perspectives on readiness of BLA/MAA fling

Fig. 16.5 BLA review process – Clinical and CMC milestones

Fig. 16.6 BLA review process – CMC milestones only

Fig. 16.7 MAA review process: Step 2 – Decision on need for cGMP inspection

Fig. 16.8 MAA review process: Step 7 – Day 120 List of Questions (LoQ)

546

List of Tables

Table 1.1 General description of some commercial recombinant proteins .

Table 1.2 General description of some commercial monoclonal antibodies . . . 13

Table 1.3 General description of two commercial bispecifc antibodies (bsAbs)

Table 1.4 General description of some commercial engineered antibody fragments

Table 1.5 General description of some commercial antibody-drug conjugates (ADCs)

Table 1.6 Comparison of general biosimilar description compared to innovator

Table 1.7

Table 1.8 General description of some commercial genetically modifed patient cells

Table 2.1 Partial outline of 21 CFR Part 312 IND clinical development requirements

Table 2.2 Partial outline of 21 CFR Part 314 NDA market approval requirements

Table 2.3 Partial outline of 21 CFR Parts 601–610 BLA market approval requirements

Table 2.4 CFR Title 21 Part 312.23(a)(7) – General guidance for CMC information in an IND

Table 2.5 CFR Title 21 Part 312.23(a)(7) – Required CMC information in an IND

Table 2.6 Module 3.2.S – Outline of required CMC information for market approval of DS

Table 2.7 Module 3.2.P – Outline of required CMC information for market approval of DP

Table 2.8 Added bioqualifers for some market-approved biopharmaceuticals

Table 2.9 Comparison of handling of market-approved biopharmaceuticals

Table 4.1 Basic CMC Regulatory information to be submitted to regulatory authorities . .

Table 4.2 FDA/EMA guidance on CMC Regulatory content to be included in submissions .

Table 4.3 ICH ‘Q’ guidelines on CMC Regulatory content to be included in submissions .

Table 4.4 Risk-based and clinical stage-appropriate CMC content fexibility in IMPD submissions

Table 4.5 Risk-based and clinical stage-appropriate CMC content fexibility in IND submissions

Table 4.6 FDA/EMA guidance on cGMPs for biopharmaceutical manufacturing and control

Table 4.7 General guidance on fexible risk-based cGMPs during early clinical stage development 91

Table 4.8 Flexible, risk-based cGMPs for gene-based biopharmaceuticals in clinical development 92

Table 4.9 Basics for the Quality System during clinical development 93

Table 4.10 Risk-based Quality Unit fexible expectations for ATMP manufacturing 96

Table 4.11 Illustration of a general TPP/QTPP for a monoclonal antibody

Table 4.12 Illustration of a general QTPP for a viral vector (AAV)

Table 4.13 QTPP used as a guide for development of Phesgo (pertuzumab/trastuzumab)

Table 4.14 Example of a risk assessment to assign criticality to QAs for a monoclonal antibody

Table 4.15 Example of a risk assessment to assign criticality to QAs for an in vivo AAV product

Table 4.16 Multiple risk assessment approaches used to determine CQAs for Gazyvaro mAb

Table 4.17 FDA recommended table format for communicating CPPs

Table 4.18 FDA recommended table format for communicating the Control Strategy

Table 5.1 Known prion diseases

Table 5.2 Non-animal-derived material substitutes for animal-derived materials

Table 5.3 List of some viruses known to harm human health

Table 5.4 Virus tests recommended to be performed once for animal/human cell lines

Table 5.5 Minimum requirements for viral clearance safety evaluation – clinical versus for market

Table 5.6 Mycoplasma species that have contaminated mammalian cell cultures

Table 5.7 Mycoplasma testing to be performed across the protein-based manufacturing process

Table 5.8 Mycoplasma testing to be performed across the viral vector manufacturing process .

Table 5.9 Mycoplasma testing to be performed across ex vivo gene therapy manufacturing process . .

Table 5.10 A holistic approach to microbial risk control

Table 5.11 Bacteria/Fungi testing requirements/recommendations for a mAb process

Table 5.12 Bacteria/fungi control data to be included in BLA fling – Module 3.2.S

Table 5.13 Bacteria/fungi control data to be included in BLA fling – Module 3.2.P.3.3/.4

Table 5.14 Bacteria/fungi control data to be included in BLA fling – Module 3.2.P.3.5 171

Table 5.15 Bacteria/fungi control for the drug substance (Module 3.2.S) 172

Table 5.16 Bacteria/fungi control for the drug product (Module 3.2.P) 173

Table 5.17 FDA caution about sterility testing challenges for gene therapy-based biopharmaceuticals 174

Table 5.18 EMA caution about sterility testing challenges for gene therapy-based biopharmaceuticals

Table 5.19 FDA background on Leptospira licerasiae cell culture contamination

Table 6.1 Some advantages/disadvantages between different host cell lines

Table 6.2 PIC/S regulatory authority aid for GMP inspecting cell bank maintenance

Table 6.3 Some suggested characterization testing for the E. coli MCB and WCB

Table 6.4 Some suggested characterization testing for the CHO MCB and WCB

Table 6.5 Starting materials – manufacture of rAAV for in vivo gene therapy.

Table 6.6 Starting materials – manufacture of genetically modifed cells (using rLV)

Table 6.7 Starting materials – manufacture of the mRNA non-viral vector

Table 7.1 Some upstream cell production bioreactor parameters and output measurements

Table 7.2 PIC/S regulatory aid for GMP inspection of the DS upstream production process

Table 7.3 Elapsed time from clinical start (IND) to seeking market approval (BLA)

Table 8.1 PIC/S regulatory aid for GMP inspection of the DS downstream purifcation process

Table 8.2 Some reduced-scale studies for the downstream purifcation process of rproteins

Table 8.3 Some reduced-scale studies for the downstream purifcation process of viral vectors .

Table 9.1 Some CMC regulatory compliance concerns associated with the manufacture of ADCs

Table 9.2 Monoclonal antibody intermediate for manufacture of Trodelvy ADC

Table 9.3 Formulation of Humira (adalimumab) and its biosimilars .

Table 9.4 Formulations of rituximab

Table 9.5 Formulations of commercial rAAV vectors for in vivo administration

Table 9.6 Formulations of commercial transduced patient cells for administration

Table 9.7 Formulations of market-approved COVID-19 mRNA vaccines .

Table 9.8 Some container closures used for market-approved biopharmaceutical drug products

Table 9.9 FDA suggestions for meeting design controls of preflled syringe cGMPs .

Table 9.10 Drug product process control data to be included in BLA fling – 3.2.P.3.3/.4

Table 9.11 Drug product process validation/evaluation to be included in BLA fling – 3.2.P.3.5

Table 10.1 Potential process-related impurities from a rprotein/mAb manufacturing process

Table 10.2 Antibiotic use in cell culture manufacturing of recombinant proteins/mAbs

Table 10.3 Potential process-related impurities for the viral vector manufacturing process

Table 10.4 Potential process-related impurities for the genetically modifed patient cells process

Table 10.5 Potential process-related impurities for the mRNA non-viral vector process

Table 11.1 Origin of biomolecular structural variants across the recombinant protein/mAb process

Table 11.2 Known structural variants for protein-based biopharmaceuticals

Table 11.3 Origin of biomolecular structural variants across the viral vector process

Table 11.4 Origin of biomolecular structural variants across the genetically modifed patient cells

Table 11.5 Origin of biomolecular structural variants across the mRNA non-viral vector process

Table 13.1 Some descriptions for appearance with market-approved biopharmaceuticals

Table 13.2 Some identity tests for biopharmaceutical products

Table 13.3 Some purity/impurities tests for biopharmaceutical products .

Table 13.4 Some descriptions of potency found in market-approved biopharmaceuticals

Table 13.5 Some descriptions for quantity found in market-approved biopharmaceuticals

Table 13.6 Testing in the seven CQA categories for a market-approved monoclonal antibody

Table 13.7 Testing in the seven CQA categories for a market-approved viral vector drug product

Table 13.8 Proposed testing in each CQA category for a viral vector 451

Table 13.9 Proposed testing in each CQA category for genetically modifed patient cells

Table 13.10 Proposed USP testing in each CQA category for mRNA non-viral vectors 455

Table 14.1 Stability test samples storage conditions .

Table 14.2 Recommended minimum testing frequency in stability program

Table 14.3 In-use handling instructions for market-approved biopharmaceuticals

Table 14.4 Interim regulatory specifcation post-market approval commitments

Table 15.1 Process change risk levels: protein-based biopharmaceuticals in clinical development 512

Table 15.2 Regulatory authority guidances on process change risk levels post-market approval 512

Table 15.3 Process change risk levels: protein-based biopharmaceuticals post-market approval 513

Table 15.4 Process change risk levels: gene therapy-based biopharmaceuticals post-market approval 515

Table 15.5 Example of Established Conditions (ECs) template 517

Table 16.1 Timeline commitments for all BsUFA meetings with the FDA 543

Abbreviations

AAV Adeno-associated virus

ACF Animal component-free

ADC Antibody drug conjugate

ADCC Antibody-dependent cellular cytotoxicity

AEX Anion exchange chromatography

ANDA Abbreviated New Drug Application

API Active pharmaceutical ingredient

ARM Alliance for Regenerative Medicine

ASO Antisense oligonucleotide

ATMP Advanced Therapy Medicinal Product

AUC Analytical ultracentrifugation

BHK Baby hamster kidney

BLA Biologic License Application

BPCI Biologics Price Competition and Innovation Act of 2009

BSA Bovine serum albumin

BSE Bovine spongiform encephalopathy

BsUFA Biosimilar User Fee Act

BTD Breakthrough Therapy Designation

CAR Chimeric antigen receptor

CAT Committee for Advanced Therapies

CBER Center for Biologics Evaluation and Research

CBMP Cell-based medicinal product

CBP Cells beyond production

CD Cluster of differentiation

CDC Complement-dependent cytotoxicity

CDC Center for Disease Control and Prevention

CDER Center for Drug Evaluation and Research

CDRH Center for Devices and Radiological Health

CDSCO Central Drugs Standard Control Organization

CFDA China Food and Drug Administration

CFR Code of Federal Regulations

Abbreviations

CFU Colony-forming unit

CGE Capillary gel electrophoresis

CGMP Current good manufacturing practice

CGTP Cell and Gene Therapy Product

CHMP Committee for Medicinal Products for Human Use

CHO Chinese hamster ovary

CI Confdence interval

CJD Creutzfeldt-Jakob disease

CMC Chemistry, Manufacturing and Controls

CMO Contract manufacturing organization

CP Comparability protocol

CPP Critical process parameter

CQA Critical quality attribute

CRL Complete Response Letter

CTA Clinical Trial Authorisation

CTD ICH common technical document

CTIS Clinical Trials Information System

CTL Contract testing laboratory

CTN Clinical Trial Notifcation

Da Dalton

DF Diafltration

DNA Deoxyribonucleic acid

DOE Design of experiments

DP Drug product

d-PCR Digital-PCR

DS Drug substance

E&L Extractables and leachables

EC Established Condition

EC European Commission

eCTD Electronic common technical document

EDQM European Directorate for the Quality of Medicines and Healthcare

ELA Establishment License Application

ELISA Enzyme-linked immunosorbent assay

EMA European Medicines Agency

EOP2 End of phase 2

EPAR European Public Assessment Report

EPC End of production cells

EU Endotoxin unit

EU European Union

Fab Antigen-binding fragment

FBS Fetal bovine serum

Fc Fragment crystallizable

FD&C Food, Drug, and Cosmetics Act

FDA Food and Drug Administration

FIH First-in-human

Turn static files into dynamic content formats.

Create a flipbook
Issuu converts static files into: digital portfolios, online yearbooks, online catalogs, digital photo albums and more. Sign up and create your flipbook.