Survey
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
CMC, Preclinical and Clinical Considerations for Biosimilars and Follow-on Biologics By Raymond A. Huml, MS, DVM, RAC; Peter Hicks, PhD; Kamali Chance, MPH, PhD, RAC; Kevin Howe, PhD; and Ross M. Tonkens, MD, FACP A s noted in the article “Follow-on Biologics in the EU and US,” on page 8 of this issue, biologics are created from living organisms, either naturally or via genetic manipulation (e.g., monoclonal antibodies), or are manufactured from complex building blocks of living organisms (e.g., siRNA, aptamers, etc.). In either case, they demonstrate considerable molecular complexity and heterogeneity, and are more difficult to characterize physicochemically than synthetic chemical entities. These differences are reflected in the regulatory agencies’ refusal to adopt the same paradigm for generic biologic drugs as for traditional small molecule products. This article summarizes some of the issues pertaining to biosimilars in Europe that are also likely to impact eventual FOB approval testing in the US. It focuses on the key parts of the development package for an FOB marketing application, including the comparability protocol exercise and regulatory authorities’ preclinical and clinical expectations. CMC Concerns for Biosimilars/FOBs As for all other drug products, a biosimilar submission to the regulatory authorities requires a complete CMC package, which is provided as Module 3 in the Common Technical Document (CTD). This CTD section provides complete information on the development, manufacture and control of both the active drug substance and the drug product. EMEA’s Guideline on Similar Biological Medicinal Products Containing Biotechnology-derived Proteins as Active Substance: Quality Issues is a good starting point. In addition—and fundamental to defining the overall development package—is the CMC comparability exercise, reported as a separate section of the filing. The manufacturer must carefully design the comparability exercise based upon full knowledge of the molecular structure and its relevance to the mode of action. The result is a series of physicochemical tests, alone or in combination with such biological tests as in vitro or in vivo bioassays, and receptor binding studies. These tests are applied to the biosimilar and the selected reference product to demonstrate similarities and differences between the two products. Where such testing cannot establish similarity, or where differences arise, the outstanding issues must be addressed through supporting preclinical and or clinical work. Biosimilar comparability testing builds upon earlier guidance on comparability of recombinant products arising from manufacturing changes. Changes introduced early in a biosimilar’s development will give rise to some predictable differences from the reference product, assuming sufficient information on the reference product is in the public domain and the mechanism of action—at the molecular level—is well-established. These differences should be assessed as part of the comparability review and appropriate testing must be performed. An example would be change of the host cell line and vector. This is a common change that can have far-ranging consequences, such as host cell protein and DNA (type and level) differences requiring that the levels present be justified and qualified. Cell line change may also go from internal protein storage requiring cell disruption to a protein expressed directly into the media, which may impact the overall product profile with regard to related materials and impurities. Another difference that may arise from changing the cell line is an alteration in the degree and type of glycosylation with potential changes to product half-life and receptor affinity and important changes to immunogenicity. These points and their resulting impacts illustrate why tight coordination among the three disciplines of quality, safety and efficacy is paramount before instituting a biotechnology-derived medicinal development program. An important consideration for any comparability exercise is the lack of access to the reference product drug substance. In a best-case scenario, the formulation adopted for the reference product may allow direct comparison and be amenable to the battery of tests to be employed. However, coformulants may potentially render some testing inapplicable or interfere with other procedures, thereby requiring extraction of the drug substance. Extraction, itself, requires careful assessment, as it may induce changes that render the results inapplicable to the product. Before a definitive comparability testing protocol can be designed, preliminary studies may be required to determine test method applicability and whether the drug product or extracted drug substance will be used. A corollary to the formulation impact is the ability to assay the reference product since certificates of analysis frequently are not available or the reference product is partially through its shelf life. Using a nominal value may have marked effects on demonstrating equivalence in comparator trials or preclinical programs where the product has a wide specification. Thus, materials should be assayed for potency prior to use. Intrinsic to establishing similarity is the complexity of the molecule under investigation. Proteins (the current focus of biosimilars RA focus 21 Table 1: Acronyms CD Circular Dichroism CE Capillary Electrophoresis CHMP Committee for Medicinal Products for Human Use CMC Chemistry, Manufacturing and Controls CTD Common Technical Document DNA Deoxyribonucleic Acid EMEA European Medicines Agency FOB Follow-on Biologic IEF Isolectric Focusing LCMS Liquid Chromatography/Mass Spectrometry NCE New Chemical Entity NMR Nuclear Magnetic Resonance PAGE Polyacrylamide Gel Electrophoresis PD Pharmacodynamic PK Pharmacokinetic siRNA small (or short) interfering Ribonucleic Acid TK Toxicokinetic UV Ultraviolet Spectroscopy and FOBs) can range from polypeptides to multi-domain proteins with or without posttranslational changes, even ignoring degrees of heterogeneity. The EU guidance recognizes the characterization difficulties and issues of variability with the expectation that the biosimilar approach be applied to highly purified product that can be thoroughly characterized—essentially a biotechnology-derived medicinal product. EU authorities also require full confirmatory clinical data for follow-on products derived from blood or plasma, and the case-by-case consideration of immunologic modulators. Current emphasis remains on licensing similar first-generation proteins coming off patent. Similar guidance and comparability concepts will need to be extended to chemically modified proteins (e.g., pegylated) as their patents expire. With these products, subtle differences between proteins may also affect actual amino acid coupling site(s) (and potential resistance to cleavage) that may markedly affect the mean residence time and pharmacodynamic profiles of these 22 January 2008 agents. Nevertheless, while chemically modified proteins will add yet another layer of complexity to comparability exercises, the underlying guidance principles will remain applicable. Characterization of proteins as either drug substances or drug products utilizes combinations of techniques. Testing is conducted to establish the primary and higher-order structures present, the presence of related materials (amino acid variants, dimers, etc.) and contaminants arising from the cell line and processing (host cell proteins, plasmid DNA, peptidase inhibitors, etc.), as well as the protein’s physicochemical parameters (molecular weight, isoelectic point, degree and type of glycosylation, etc). Techniques employed include, but are not limited to, primary amino acid analysis, peptide mapping, LCMS (and its variants), CE, CD, NMR, UV, IEF and PAGE (normal and denaturing). In many cases, however, there is no direct link between the data generated and the protein’s overall conformation and pharmacodynamic effect. Therefore, an in vitro or in vivo biological assay relevant to the mode of action is required, which may also necessitate supplementary receptor binding studies. The testing program must be able to demonstrate both differences and similarities. Data produced by the comparability exercise must be reviewed and put into context. By the very nature of the material in question and the processes used to produce it, differences may arise. Such differences themselves are not the issue; the ultimate pharmacodynamic impact and overall safety they may confer must be assessed. The answer to this can lie outside the CMC domain. For some, answers are obtained from biological testing, but for others, the answers are found in the preclinical and clinical arenas. CMC comparability is essential to any biosimilar and is absolutely fundamental in defining the overall development program. However, it must be recognized that a detailed, comprehensive program necessary to support a regulatory application will be expensive not only in terms of money, but also in terms of key personnel and resources (a major consideration for smaller organizations). For platform-based companies looking at numerous biosimilars, the overall costs are multiplied and will be significant—often beyond the means of all but the most robustly capitalized. Once a detailed comparability program has been established, based in part upon preliminary CMC data, regulatory scrutiny should be sought via the EMEA Scientific Advice program. Companies seeking advice will not only validate their approaches, but will benefit from the regulators’ current thinking on any one product as well. Smaller companies looking to out-license will particularly enhance their negotiating leverage by following this proactive approach. Efficacy Comparisons, Preclinical Safety, Pharmacokinetics and Clinical Studies for FOBs Current guidelines adopted in the EU are likely to form the backbone of future FDA‑specific guidance regarding FOBs. EMEA’s Guideline on the Nonclinical and Clinical Issues to be Considered for the Development of Similar Biological Medicinal Products (EMEA/CHMP/BMWP/42832/2005) details EU issues and expectations regarding efficacy, safety and clinical testing, but these could ultimately differ in the US. The requirements depend upon existing knowledge of the reference biological medicinal product and the claimed therapeutic indication(s) in addition to reference drug availability and confirmation of that agent’s identity. Available product- and/or disease-specific guidelines should be followed, acknowledging that the FOB manufacturing process will be optimized throughout development. It is highly recommended that clinical data required for the comparability study be generated in parallel with final manufacturing process design to represent the FOB quality profile expected of the commercial batches. The clinical comparability exercise is a stepwise procedure that begins with pharmacokinetic (PK) and pharmacodynamic (PD) studies, followed by clinical efficacy and safety trial(s) or, in certain cases, PK/PD studies only for demonstrating clinical comparability. For preclinical and in vitro comparability testing, assays like receptor-binding studies or other cell-based assays may be available from quality-related bioassays. Preclinical and in vitro comparability testing should be conducted to determine comparability in reactivity and likely causative factor(s), if comparability cannot be established. Animal studies should be designed to maximize information and to compare both reference and FOB intended for clinical trial use. Animal studies should therefore be performed in species that are relevant to humans and should employ state-ofthe-art technology, ideally monitoring a number of clinically relevant pharmacodynamic endpoints. For toxicology studies, non-human primates are likely to be the most relevant species for a human protein, but occasionally no species may How do you get the latest medical device intelligence in a global industry that’s always changing? Problem: You need the latest regulatory information, right now. Nothing the new Tarius® medical device web portal can’t solve. Special discounts for RAPS members. Tarius® is a single web resource that gives you access to regulations directives, guidelines, forms and more—with daily updates! tarius.com Why not try it for free? The result: A simpler way to find, use and share critical information in your dayto-day work. For more information and free trial, go to www.tarius.com today! RA focus 23 HORIZ 2 0 0 8 R A P S H O R I Z O N S C 26–28 March 2008 • San Francisco • The Fairmont • www.raps.org/horizons2008 Dig in! See for yourself why 2008 RAPS Horizons Conference & Exhibition challenges you to think 2008 RAPS Horizons Conference strategically and creatively about hot topics and vital issues that directly impact & Exhibition is the conference to your ability to excel in today’s fast-paced regulatory environment. attend for regulatory professionals who aren’t satisfied with simply skimming the surface of today’s most critical regulatory topics. This unique learning and networking experience is designed for experienced regulatory affairs professionals, cutting across product lines and addressing critical aspects of the healthcare product lifecycle. You will walk away with innovative approaches and measurable strategies What is 2008 RAPS Horizons Conference & Exhibition: • Advanced perspectives on cutting– edge topics • Challenging, in-depth discussions that can be effectively implemented within your organization and support your professional development. Visit www.raps.org/horizons2008 to access important program information and register online today. • Unique networking opportunities • Expert speakers from industry and regulatory agencies • Hot new products, services and technologies • Regulatory resources: RAPS Bookstore, RAPS Booth and more! Explore leading-edge regulatory topics that you’ll find nowhere else in multitrack sessions cutting across products lines: devices, drugs, combination products, biologics and more! O ZONS N F E R E N C E & E X H I B I T I O N Think. Challenge. Excel. Key opinion leaders from around the globe discuss the impact of current and future regulatory trends, market innovations, new technologies, global challenges and much more in detailed, explorative education sessions that challenge you and provoke critical thinking. Each day, topics focus on vital subjects affecting the typical product lifecycle, including: Early Lifecycle (Day 1) • What’s on the horizon in medical science and technology? • Business strategic role of the RA professional • Navigating the product development process • Risk management with new technologies • Regulatory strategy when outsourcing global clinical trials Submission to Approval (Day 2) • Dissemination of information about investigational products • Mitigating liability Issues with new technologies • Regulatory authority meetings and effective engagement of regulatory bodies • Product-specific submissions Postapproval (Day 3) • Global regulatory changes • EU vigilance reporting requirements • Special challenges with postapproval combination products • Crisis management WHO SHOULD ATTEND Experienced regulatory affairs professionals involved in the research, development, regulatory submission, manufacture or marketing of healthcare products throughout all product areas, as well as mid- and senior-level professionals involved in preclinical, regulatory review and postmarketing aspects of the product lifecycle. Levels II, III, IV VALUABLE NETWORKING OPPORTUNITIES Expand your professional circle, gain important regulatory insight and make the most of your time away from the office with these opportunities to engage in meaningful networking and in-depth discussions of the day’s events: • Networking reception • Dine–arounds at renowned San Francisco restaurants • Hot topic roundtable exchanges • Exhibit Hall featuring new products and valuable resources Already know the basics of regulatory affairs? Attend 2008 RAPS Horizons Conference & Exhibition and… • Think critically about today’s hottest topics. • Examine fresh solutions to regulatory challenges. • Get the skills you need to excel in your career. be relevant. Therefore, direct reference to the original dossier’s study designs is required for consideration of species selection. At least one repeat-dose toxicity study containing toxicokinetic (TK) measurements is expected. These TK measurements should include determination of antibody titers and neutralizing capacity. These latter factors are most relevant in the required clinical trial and should be built into the clinical protocol including isotype characterisation. Toxicity study duration should be sufficient to enable detection of relevant differences in toxicity and/or immune responses between the FOB and the reference medicinal product. If there are specific safety concerns, these might be addressed by including relevant observations (i.e., local tolerance) in the same repeat-dose toxicity study. Other standard NCE-based toxicological studies, such as ICH S7 safety pharmacology, reproductive toxicology, mutagenicity and carcinogenicity studies are not usually required for FOBs unless indicated by repeat-dose study results, with reference to the original product submission. Generally, one comparative clinical trial is required to demonstrate clinical comparability. In certain cases, however, comparative PK/PD studies between the FOB and the reference medicinal product may be sufficient to demonstrate clinical comparability. Clinical comparability margins should be prespecified and justified based upon clinical grounds. As for all clinical comparability trial designs, assay sensitivity should be demonstrable and reproducible. If a clinical comparability trial design is not feasible, other designs should be explored and their use discussed with the relevant regulatory authorities. Overall, and based upon current experience in the EU, safety data derived from the comparator clinical trial—focusing on immune response endpoints—are required. However, since the comparator trial may not generate sufficient data on long-term safety, the sponsor is expected to conduct postmarketing (phase 4) studies to adequately address long-term safety. Ongoing monitoring of FOB clinical safety, including a risk-benefit assessment, must be conducted in the postapproval phase. The sponsor must provide a risk specification in the application dossier for the medicinal product under review in the EU. This should include a description of possible safety issues related to tolerability of the medicinal product that may result from a manufacturing process, as contrasted with the originator, comparator medicinal product. Ultimately, during the authorization process, the applicant is expected to present a risk management program/pharmacovigilance plan in accordance with current guidelines. In conclusion, the issues outlined in this article that pertain to biosimilar testing currently required in Europe are also likely to influence the eventual FOB approval testing requirements in the US, once legislation is passed. Summary 26 January 2008 •CMC comparability remains the cornerstone of establishing an overall development package for biosimilars. •Direct chemical comparison remains essential. In any comparability exercise individuals experienced in proving quality are as essential as those proving safety and efficacy. •Only with time, further advances in analytical techniques and better REFERENCES 1.The Pink Sheet. “Follow-on biologics deal gives brands 12 years of exclusivity.” Vol 19, Issue 121, Number 008; 22 June 2007. 2.110th Congress; Bill for Biosimilars (http://help.senate.gov/ Hearings/2007_06_27_E/Biologics.pdf) Accessed 26 July 2007. For additional information, see references and tables in “Follow-on Biologics in the EU and US,” on page 8 of this issue AUTHORS Raymond A. Huml, MS, DVM, RAC, is Executive Director of Global Due Diligence for NovaQuest, the product partnering group of Quintiles Transnational Corp. He has worked in various roles for Quintiles since 1993 and currently serves as a project leader for global due diligence activities. Huml can be reached at [email protected]. Peter Hicks, Btech, PhD, is Executive Director of Global Due Dili- gence for NovaQuest. Following a post-doctoral fellowship with Boehringer Ingelheim, he was a lecturer in pharmacology at the University of Manchester. Hicks has 30 years of pharmaceutical and CRO industry experience and can be reached at [email protected]. Kamali Chance, MPH, PhD, RAC, is Director of Global Regulatory Affairs and Medical Writing at Quintiles, Inc. She has more than 20 years of management experience in the healthcare industry and over nine years of regulatory experience in the pharmaceutical/biotechnology industry. Chance can be reached at [email protected]. Kevin Howe, BPharm, PhD, CChem, MRSC, CBiol, MIBiol, MRPharmS, MIoD, Qualified Person, is owner of Epi-Cure Consulting Ltd, a specialist drug development and regulatory organization. He has over 20 years of varied postgraduate experience in academia, the National Health Service, pharmaceutical industry and Quintiles where he was he was Head of The Pharma Consulting Group in Europe. Howe can be reached at [email protected]. Ross Tonkens, MD, is chief medical officer of Regado Biosciences, a company pioneering in discovery and development of antidote-controlled therapeutics directed against specific molecular targets. He practiced invasive and noninvasive cardiology for more than 20 years in Beverly Hills, and then Las Vegas, also teaching at UCLA and University of Nevada Schools Medicine and participated as principal investigator in over three dozen clinical trials. Formerly, Tonkens was Global Head of Cardiovascular Therapeutics for Quintiles. He can be reached at [email protected]. Acknowledgement The authors would like to thank Dr. Judith Beach, PhD, Esq., Vice President and Senior Associate General Counsel and Regulatory and Government Affairs Chief Privacy Officer for Quintiles Transnational Corp., for her editorial assistance with this manuscript. Principles &Practices of EU and US Medical Devices 10-11 March 2008 Munich ArabellaSheraton Westpark www.raps.org/pandp/munich Want to accelerate your RA career? Looking to refresh your knowledge of medical devices? Join the Regulatory Affairs Professionals Society (RAPS) for this popular two-day conference on the principles and practices of medical devices in the EU and US markets and get the basics you need to get ahead. SESSION TOPICS • Overview of EU Medical Device Regulatory • Medical Device Directives • Medical Device Premarket Requirements • Medical Device Postmarket Requirements • Introduction to US Medical Devices • Medical Device Submissions • Advertising, Labelling and Promotion of Medical Devices • Quality System Regulation R e to gis da te y! r understanding of relationships between molecular structure and function will the development of FOBs become more focused. •With improved scientific insight, our development of biosimilars may reach a position analogous to that of generic synthetic chemical entities. •It is imperative that companies contemplating FOBs consult with respective regulatory authorities early in the process to obtain buy-in for the strategic development plan. RA focus 27