What You Should Know When You Make Manufacturing Changes to Biotechnology Products May 16-18, 2011 | Beijing, China Mark Rosolowsky, Ph.D. Vice President,

Slides:



Advertisements
Similar presentations
Testing Relational Database
Advertisements

Project Quality Plans Gillian Sandilands Director of Quality
Biopharmaceutical Quality
Statistical Evaluation of Dissolution for Specification Setting and Stability Studies Fasheng Li Associate Director, Pharmaceutical Statistics Worldwide.
Safety and Extrapolation Steven Hirschfeld, MD PhD Office of Cellular, Tissue and Gene Therapy Center for Biologics Evaluation and Research FDA.
Strengthening the Medical Device Clinical Trial Enterprise
Reprocessing Biotech Products
1 Implementation of Quality by Design (QbD): Status, Challenges and Next Steps Moheb M. Nasr, Ph.D. Office of New Drug Quality Assessment (ONDQA), OPS,
Stability data required by WHO-PQP Mercy Acquaye.
1 The critical challenge facing banks and regulators under Basel II: improving risk management through implementation of Pillar 2 Simon Topping Hong Kong.
Batch Reworking and Reprocessing
Integrating CMC Review & Inspection Industry Recommendations Joe Anisko April 24, 2003.
World Health Organization
1 MANUFACTURING AND PRODUCTION OF BIOLOGICAL PRODUCTS (ERT 455) HAZARD ANALYSIS AND CRITICAL CONTROL POINT (HACCP) SYSTEM Munira Mohamed Nazari School.
Determine impurity level in relevant batches1
U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES NATIONAL INSTITUTES OF HEALTH Working with FDA: Biological Products and Clinical Development Critical Path.
The ICH E5 Question and Answer Document Status and Content Robert T. O’Neill, Ph.D. Director, Office of Biostatistics, CDER, FDA Presented at the 4th Kitasato-Harvard.
What Do Toxicologists Do?
Pharmaceutical Product Quality Assurance Through CMC Drug Development Process Presented by Darlene Rosario (Aradigm) 21 October 2003 Meeting of the Advisory.
COMPARABILITY PROTOCOLS ACPS March 12-13, 2003 Stephen K. Moore, Ph.D. Chemistry Team Leader CDER/Office of New Drug Chemistry Co-Chair, Comparability.
Clinical Trials of Traditional Herbal Medicines In India Y.K.Gupta Professor & Head, Department of Pharmacology, All India Institute of Medical Sciences,
Training Workshop on Pharmaceutical Development with a Focus on Paediatric Medicines / October |1 | Regulatory Requirement on Dossier of Medicinal.
QUALITY CONTROL OF PHYSICO-Chemical METHODS Introduction :Validation توثيق المصدوقية.
GLOBAL REGULATORY STRATEGY CONSIDERATIONS SCIENTIFIC SARAH POWELL EXECUTIVE DIRECTOR, REGULATORY STRATEGIES SEPTEMBER 14-17, 2008 BOSTON, MA.
Learnings from Pre-approval Joint Inspection of a GSK QbD Product with US-FDA & EMA and the application of Continuous Verification 17 May 2011, Beijing,
Codex Guidelines for the Application of HACCP
Application of the principles of QbD in vaccines production Andrea Pranti.
Allergen regulation in the future : what will be the place for recombinant allergens ? Jacqueline DAYAN-KENIGSBERG European Academy of Allergology and.
Achieving and Demonstrating “Quality-by-Design” with Respect to Drug Release/dissolution Performance for Conventional or Immediate Release Solid Oral Dosage.
Nonclinical Studies Subcommittee Advisory Committee for Pharmaceutical Science CMC Issues for Screening INDs Eric B. Sheinin, Ph.D. Acting Deputy Director.
1 Supplements and Other Changes to an Approved Application By: Richard J. Stec Jr., Ph.D. February 7, 2007.
Barry Cherney, Ph.D., Deputy Director DTP/OBP/CDER/ FDA Perspectives on Comparability of Biotechnology Derived Protein Products.
A substance used in the diagnosis, treatment, or prevention of a disease or as a component of a medication A substance used in the diagnosis, treatment,
© Grant Thornton | | | | | Guidance on Monitoring Internal Control Systems COSO Monitoring Project Update FEI - CFIT Meeting September 25, 2008.
Drug Submissions: Review Process Agnes V. Klein, MD Biologics and Genetic Therapies Directorate February, 2003 www/hc-sc.gc.ca/hpb-dgps/therapeut.
Exploratory IND Studies
Planned Emergency Research Exception from Informed Consent Requirements September 2007.
Nonclinical Perspective on Initiating Phase 1 Studies for Small Molecular Weight Compounds John K. Leighton, PH.D., DABT Supervisory Pharmacologist Division.
Multimedia Assessment for New Fuels: Stakeholders’ Meeting September 13, 2005 Sacramento, CA Dean Simeroth, California Air Resources Board Dave Rice, Lawrence.
Chapter 6 CRISIS MANAGEMENT. Introduction - Crisis: ◦is a situation that specifically involves a pharmaceutical product, medical device or activity with.
1-7.The ICH Q8 “Minimal Approach” to Pharmaceutical Development
Important informations
Marcel H.N. Hoefnagel 2 November 2007 BIOSIMILARS are not Generics But similar.
CHALLENGES FACED IN THE DEVELOPMENT OF BIOSIMILARS Dr.G.Hima Bindu MD; PG dip. diabetology Asst.Professor Dept. of Pharmacology Rajiv Gandhi Institute.
Overview of FDA's Regulatory Framework for PET Drugs
PhRMA Perspective on FDA Final Report FDA Advisory Committee on Pharmaceutical Sciences October 20, 2004 G.P. Migliaccio, Pfizer Inc.
Molecule-to-Market-Place Quality
Workshop Session 3 Questions 1 How would a control strategy look different in a traditional submission vs a QbD submission? How would parameters that are.
The New Drug Development Process (www. fda. gov/cder/handbook/develop
COMPARABILITY PROTOCOLUPDATE ADVISORY COMMITTEE FOR PHARMACEUTICAL SCIENCE Manufacturing Subcommittee July 20-21, 2004 Stephen Moore, Ph.D. Chemistry Team.
1 Recommendations Now that 40 GbE has been adopted as part of the 802.3ba Task Force, there is a need to consider inter-switch links applications at 40.
Agenda for Session Compliance in Clinical Research
International Atomic Energy Agency Roles and responsibilities for development of disposal facilities Phil Metcalf Workshop on Strategy and Methodologies.
General Aspects of Quality assessment of multisource interchangeable medicines Rutendo Kuwana Technical Officer, WHO, Geneva Training workshop: Assessment.
Company LOGO. Company LOGO PE, PMP, PgMP, PME, MCT, PRINCE2 Practitioner.
WORKSHOP ON ACCREDITATION OF BODIES CERTIFYING MEDICAL DEVICES INT MARKET TOPIC 9 CH 8 ISO MEASUREMENT, ANALYSIS AND IMPROVEMENT INTERNAL AUDITS.
In vitro - In vivo Correlation
A substance used in the diagnosis, treatment, or prevention of a disease or as a component of a medication recognized or defined by the U.S. Food, Drug,
In the name of God. Common Technical Document On Biotech.
POST APPROVAL CHANGE MANAGEMENT PROTOCOLS IN THE EUROPEAN UNION
Guidance for review of studies involving HCT/Ps and IND Basics
Hina M. Pinto, MSE Scientific Reviewer
Pharmaceutical Quality Control & current Good Manufacturing Practice
FDA’s IDE Decisions and Communications
Right-sized Evaluation
Northeast Biomanufacturing Center and Collaborative
The Lifecycle of Pharmaceutical products
Implementation of Quality by Design (QbD): Status, Challenges and Next Steps Moheb M. Nasr, Ph.D. Office of New Drug Quality Assessment (ONDQA), OPS, CDER.
Quality guidelines on impurities
Quality guidelines on impurities
Presentation transcript:

What You Should Know When You Make Manufacturing Changes to Biotechnology Products May 16-18, 2011 | Beijing, China Mark Rosolowsky, Ph.D. Vice President, Global Regulatory Sciences-CMC Bristol-Myers Squibb

Disclaimers –The information within this presentation represents the views of the presenter and is based on the presenter’s expertise and experience 2

3 Overview –Comparability - key points for consideration –Discuss changes to biotechnology products: During development Post-marketing

4 Why are Changes to Biotechnology Products so Complex? Small Molecules –Usually synthetic, organic compounds having well defined structures and chemical characteristics –Typically produced through chemical synthesis –Usually micromolecules having molecular weights of less than 500 Daltons –Generally very stable, and not extremely sensitive to heat Biotechnology Products –Usually a protein- or carbohydrate-based product with complex structure –Either composed of / or extracted from a living organism or produced via cell culture –Macromolecular by nature, and usually have a molecular weight greater than 500 Daltons –Tend to be rather labile, and are usually very heat- and sheer- sensitive –Tend to be immunogenic

What is Comparability? ICHQ5E Definitions Comparable: –A conclusion that products have highly similar quality attributes before and after manufacturing process changes and that no adverse impact on the safety or efficacy, including immunogenicity, of the drug product occurred. This conclusion can be based on an analysis of product quality attributes. In some cases, nonclinical or clinical data might contribute to the conclusion. Comparability Exercise: –The activities, including study design, conduct of studies, and evaluation of data, that are designed to investigate whether the products are comparable. 5

Comparability Exercise Considerations –No one right answer when deciding a comparability package –Decision must be made with consideration of: Product clinical development plan Complexity of the product Stage of development of the product Robustness of analytical methods Existence of relevant animal models Previous health authority interactions 6

What is “Highly Similar”? –Comparability does not require quality attributes of pre-change and post-change product to be identical –“Highly similar” depends upon whether: Existing knowledge can adequately support that differences in quality attributes have no adverse impact upon safety or efficacy –Side-by-side analysis of “post-change” vs. “pre- change” product is useful for contemporaneous evaluation 7

8 Comparability is a Sequential Process Comparability is a Sequential Process Non-clinical Clinical and/or Pharmacovigilance Quality Confirmatory clinical testing may be necessary Non-clinical studies may be necessary, if non-clinical studies cannot discern relevant differences, then… If the analytical procedures used are not sufficient to discern relevant differences, then… OVERALL GOAL: Assess potential impact to safety and efficacy of the product

Parameters to Consider –Production step where changes are introduced. –Potential impact of changes to: purity, physicochemical and biological properties considering complexity and degree of knowledge (e.g., impurities, product related substances). –Availability of suitable analytical techniques to detect potential modifications –Understanding of relationship between quality attributes and safety and efficacy, based on overall nonclinical and clinical experience. –Relevant physicochemical and biological characterization data regarding quality attributes; –Need for stability data, including accelerated or stress conditions, to provide insight into potential product differences in the degradation pathways 9

Parameters to Consider (continued) –Batches used for demonstration of manufacturing consistency; –Historical data that provide insight into potential “drift” of quality attributes –Critical control points in the manufacturing process that affect product characteristics, –Impact of the process change on the quality of in-process materials & ability of downstream steps to accommodate material from a changed cell culture process; –Adequacy of the in-process controls (critical control points & in- process testing: In-process controls for post-change process should be confirmed, modified, or created, as appropriate, to maintain product quality –Nonclinical or clinical characteristics of the drug product and its therapeutic indications 10

Risk Assessment –Utilize prior knowledge and development studies –Categorize risk change of process parameters based upon potential to impact product quality –Examples: High: Change to Master Cell Bank (MCB) Moderate: Media composition change using established raw materials, <50% output Low: Step optimization (e.g. flow rate, wash volumes, elution collection criteria) –Provides an effective tool for internal discussion of change & subsequent communications to regulators regarding the assessment 11

Risk Assessment Model

Example Risk Assessment Tool 13  Each risk factor category has built-in drop-down boxes with potential values  Higher values indicate greater risk  Team discussion is critical to document thought process that drove the scores (“no one right answer”) Total risk score generated by multiplying individual risk factors

Evaluating Changes During Development 14

Phase-based Approach to Changes Early Development: –Before nonclinical studies: Comparability is not generally not a concern Subsequent nonclinical and clinical studies using the post-change product as part of the development process support change –Early phases of nonclinical and clinical studies: Comparability testing is generally not as extensive as for an approved product –As knowledge and information accumulate, the comparability exercise will generally become more comprehensive

Phase-based Approach to Changes During Pivotal Clinical Studies: –Changes are discouraged –Sponsor should seek scientific advice from the relevant health authorities If process changes are introduced in late (post- pivotal studies) stages of development: –Thorough comparability exercise is generally required: Physicochemical and biological in vitro studies Clinical pharmacokinetic and / or pharmacodynamic comparability studies may also be required –If comparability exercise cannot rule out impact to the efficacy and safety profile: Additional clinical studies may be required 16

Guidelines for Acceptance Criteria Setting for Analytical Comparability a Orthogonal support for release test or independent attribute assessment for structure-function relationship b If a stability indicating attribute is identified under recommended storage conditions c If a forced degradation or stressed condition degradation product attribute is identified and believed relevant to structure-function

Case Study - Development 18

Case Study Process Changes Overview of Changes –Drug Substance New MCB (higher producing subclone of current MCB) New DS manufacturing site (Site “A”  Site “B”) New DS manufacturing process (cell culture and purification) –Drug Product New DP manufacturing site (Site “X”  Site “Y”) Minor change to sterile filtration (0.1 µm  0.2 µm) Reason for Changes –Increase drug substance yield ~4X –Manufacturing control –COG

Comparability Exercise Goal - to ensure the quality, safety and efficacy of drug product produced by a changed manufacturing process How - through collection and evaluation of the relevant data based on process and product knowledge –Analytical assays –Biological assays –Nonclinical data –Clinical data

Analytical Comparability Established at multiple points –In-process –Release Release tests Extended characterization tests –Stability profile Recommended storage condition Accelerated/Stress storage conditions –Downstream Drug substance changes may only be seen in the drug product (release, stability profile )

Analytical Acceptance Criteria In-process –Comparable process/product related impurity/adventitious agent clearance Release tests –Current (pre-change) specification Additional tests may be needed Extended characterization –May need to evaluate additional pre-change batches to establish appropriate acceptance criteria Evaluation against historical data (i.e. clinical experience) Side-by-side comparison of pre- and post-change product by various analytical characterization techniques

Binding Kinetics by Surface Plasmon Resonance Association Dissociation

Electron Spray Ionization Mass Spectrometry Electron Spray Ionization Mass Spectrometry Reference material (pre-change) Post-change product Slight increase in one subtype observed

Tryptic Peptide Maps Overlay of pre- and post-change materials

Comparison of Carbohydrate Profiles by HPAEC-PAD 26 Pre-change material Post-change material

Isoelectrically Focused Isoelectrically Focused Lane 1: Reference material (pre-change) Lanes 2 through 5: Post-change material

Cation Exchange Profiles Gray line: Pre-change material Black line: Post-change material

Evaluating Changes for Marketed Products 29

Changes to Marketed Products Changes to Marketed Products Changes during life-cycle are inevitable: –Yield increases necessary to meet market demand / address cost of goods issues –Quality improvements are necessary to adhere to current GMPs –Unexpected events require corrective action, such as process parameter changes –Vendor / supply issues necessitate use of alternate materials 30

Hypothetical Process Evolution Hypothetical Process Evolution Process Step Phase IIPhase III & Commercial Process CProcess DProcess E Cell Bank XYZ-01XYZ-01.1Same Media DECD-CHO, eRDF & Yeastolate Same, Plus Additional Minerals Same Production Conditions No temp. shift Two phase temp. shift Same Single temp. shift Downstream Sequence Six columns Five columns SameChange of one resin Other Process Changes N/A Centrifugation & filtration parameters Filter changesN/A

Case Study – Marketed Product 32

Case Study: Change in Media –Quality: Media component change resulted in minor differences to quality attributes –Nonclinical: Study conducted using non- human primate model with previously well- established concordance to human PK for the product Model had demonstrated sensitivity to changes in: –Minor glycosylation alterations –Moderate sialic acid profile shifts 33

Non-Human Primate Results Red line = pre-change Blue line = post-change Time in Hours Serum Concentration [ug/mL]

FDA Feedback –Quality attributes evaluated against historic data alone deemed insufficient “…the Agency strongly encourages the use of side-by-side analysis as the most rigorous assessment of comparability…” –Side-by-side comparison required: “Since the product approved for marketing authorization was made using the…media..., this product should be directly compared to the product produced from the proposed process…”

Regulatory Experiences with Comparability Regulatory Experiences with Comparability –Comparability exercises can be successfully used to support changes –Most changes can be supported on Quality attributes alone Specifications alone are generally NOT sufficient to support comparability –Additional characterization required –Side-by-side analysis preferred by FDA –Additional non-clinical & clinical data may be required –Safety & efficacy data are generally not required 36

In Summary –Comparability is a key issue for biotech products –There is no single, correct strategy to demonstrate comparability –Decision must be made with consideration of multiple factors: Complexity of the product Stage of development of the product Process knowledge & robustness of analytical methods Existence of relevant animal models Previous health authority interactions –Comparability should be approached stepwise: Quality Non-clinical Clinical 37

Acknowledgements Cheryl Watson Reb Russell Dave Peck Charlene Craig Gary Lazarus 38