Regulatory Challenges for the Licensure of Future Vaccines

Similar documents
FDA Draft Guidance on Immunogenicity Testing

Clinical qualification of specifications - a Regulator s view

MAIN OUTCOMES OF DISCUSSION FROM WHO CONSULTATION ON NUCLEIC ACID VACCINES. I. Knezevic, R. Sheets Feb, 2018 Geneva, Switzerland

Changes to a Potency Bioassay for Biotechnology Products: a Regulatory Perspective Kathleen A. Clouse, Ph.D., Director Division of Monoclonal Antibodi

London, 11 October 2006 Doc. Ref. EMEA/CHMP/BWP/271475/2006 COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP)

CASE STUDY: THE USE OF PRIOR KNOWLEDGE IN ESTABLISHMENT OF AN INTEGRATED CONTROL STRATEGY AND CLINICALLY RELEVANT SPECIFICATIONS

Guideline for the quality, safety and efficacy of follow-on biological medicinal products

Biosimilar Monoclonal Antibodies: Registration Requirements. Henry M. J. Leng

Regulatory Perspective on Analytical Method Validation During Product Development

Stability of Biological Products

Regulatory perspective on setting clinically relevant specifications. Joslyn Brunelle, PhD Team Leader Office of Biotechnology Products

Control strategy and validation. Emanuela Lacana PhD Office of Biotechnology Products CDER/FDA

Phase-Appropriate Analytical Method Validation: A Regulator s Perspective

Risk Assessments for Host Cell Protein Control Strategies: CDER Experiences

Archived Content. This content was archived on June 24, 2013.

Guideline for the Quality, Safety, and Efficacy Assurance of Follow-on Biologics

Essentials in Stability Analysis and Expiry Determination

Substituting In Vitro for In Vivo Potency and Safety Assays: Science Versus the Fear Factor*

fact sheet 3 Introduction to Biosimilars & Regulatory Requirements

FDA Perspective on the Preclinical Evaluation of Biological Therapies for Cancer

Characterization of Biotechnology Products: A Regulatory Perspective

R&D of Biological Products. Rochapon Wacharotayankun, Ph.D., R.Ph.

Guidelines on the nonclinical evaluation of vaccine adjuvants and adjuvanted vaccines

Update on the new immunogenicity guideline in the EU

DCVMN workshop 7-10 May 2018, Hyderabad Hilde Depraetere, PhD European Vaccine Initiative DCVMN WORKSHOP, HYDERABAD

Copyright. Jeremiah J. Kelly (2015). All rights reserved. Further dissemination without express written consent strictly prohibited.

CLINICAL CONSIDERATIONS FOR EVALUATION OF FOR PREQUALIFICATION 1. Points to consider for manufacturers of human vaccines

Critical Quality Attributes for Biotechnology Products: A Regulatory Perspective

DMTC Technology Readiness Levels Guideline

Setting Specifications for Biotech Products

FDA Public Hearing: Approval Pathway for Biosimilar. Products. November 2-3, 2010

Introduction of Development Center for Biotechnology TAIWAN

Guidelines for Pharmaceutical Equivalence Requirements

Basis for setting acceptance criteria

Notice Our file number:

Affigenix Biosolutions Private Ltd, 265/ 1F, KSSIDC Industrial Area Bangalore, Karnataka,India

Scientific and Regulatory Perspectives on Measuring Protein Aggregation of Biotechnology Products

Strategies for IND Filing Success: Chemistry, Manufacturing and Controls

Rasha Sayed Salama, MD, PhD, UAE

Regulatory Issues and Drug Product Approval for Biopharmaceuticals

Vaccines based on Recombinant Proteins and Adjuvant Systems: GSK's malaria vaccine candidate as a case study.

Phase Appropriate GMPs for IMPs. Presented by: Karen S. Ginsbury For: IFF, October 31, Nov 02, 2017

VLP Vaccines, an Example of Product Profile Specification Setting

Reference Standards for Monoclonal Antibodies: Key Challenges Addressed

Cell Therapy Product Manufacturing Considerations. July 17, 2017 CMC Strategy Forum Mo Heidaran, Ph.D.

European Medicines Agency Evaluation of Medicines for Human Use

Professor Andrea Laslop, MD, Austria

The Role of Mass Spectrometry for Developing Biotherapeutics: Regulatory Perspectives

Division of Dockets Management (HFA-305) Food and Drug Administration 5630 Fishers Lane, rm Rockville, MD 20852

Dealing with Post-market Issues: OOS Case Study

Oral vaccines to protect patients against Clostridium difficile infection

Design and Interpretation of Accelerated Stability Studies of Biologics

Lessons Learned from QbD Application of Biologics in Japan: Perjeta Case

ANALYTICAL VALIDATION CHALLENGES DURING THE RAPID DEVELOPMENT OF KEYTRUDA

Regulatory Expectations for Method Transfers: Health Canada's Perspective CMC Strategy Forum Methods on the move January 23, 2017

NDA Advisory Services Ltd

Application of ICH Q12 Tools and Enablers Post-Approval Lifecycle Management Protocols

Introduction regarding aggregate/particle issues in biopharmaceuticals. Yasushi Shikata Eisai Co., Ltd.

ADVAC ALUMNI MEETING DURING SAGE

Replacing Analytical Methods for Release and Stability Testing CBER Perspective

Appropriate Control Strategies Eliminate the Need for Redundant Testing of Pharmaceutical Products

Dr. Earl Dye CMC/GMP Considerations for Expedited Development Programs

Antibody Discovery at Evotec

Comparability of Biotechnology Derived Protein Products: Lessons from the U.S Experience

Guidelines on procedures and data requirements for changes to approved biotherapeutic products

CMC Considerations for Manufacturing of CAR T-Cell Product

Applied Protein Services

Paradigm Shift in Comparability Assessment:

Synopsis of Issues Concerning 9 CFR and VSM

Data & Materials Sharing Agreement. Collaboration for AIDS Vaccine Discovery. Clinical Trials Data Sharing Addendum & Related Information

Harmonizing clinical trials for Biogenerics. Dr. Akhilesh Sharma M.D.;C Clin. Research & P.V. (UCBC - USA & Luton - UK)

A Regulator s Perspective on Risk Based and Phase Appropriate Comparability. Marjorie Shapiro, Ph.D. Division of Monoclonal Antibodies OBP, CDER,FDA

Health Canada s Experience with Bioassays used for Consistency Testing of Biotherapeutic Products

Reflection Paper. The Role of Product-specific Monographs for Biotherapeutic Products in Pharmacopoeias

Table 12: Design of Stability Studies for Biotechnology Product Development and Lifecycle Management A New USP Chapter

Re: Docket No. FDA-2017-D-5525: Statistical Approaches to Evaluate Analytical Similarity

Immunogenicity Assay Strategies for Antibody-Drug Conjugates

Guide. recombinant DNA proteins. for the elaboration of monographs on synthetic peptides and. European Pharmacopoeia

Wrap Up Summary CMC Strategy Forum. Bridging Analytical Methods Jan 27, 2014

A Regulatory Perspective on Characterization and Control of Process-Related Impurities

Table 37: Design of Stability Studies for Biotechnology Product Development and Lifecycle Management A New USP Chapter

WHO/BS/ ENGLISH ONLY. EXPERT COMMITTEE ON BIOLOGICAL STANDARDIZATION Geneva, 29 October to 2 November 2018

Field trial with veterinary vaccine

Biosimilars: Questions and Answers Regarding Implementation of the Biologics Price Competition and Innovation Act of 2009

Guidelines on the nonclinical evaluation of vaccine adjuvants and adjuvanted vaccines. Proposed guidelines

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP)

MenAfriVac clinical development

Overview Everything You Wanted to Know About GLPs. but were afraid to ask

Pharmaceutical Reference Standards: Overview and Role in Global Harmonization

Submission of comments on Guideline on Similar Biological Medicinal Products (CHMP/437/04 Rev1)

Common Issues in Qualification and Validation of Analytical Procedures

Understanding the Use of Bioassays in a Biomanufacturing Facility. Bernardo E. Cáceres Associate Principal Scientist Biotech Development

Reduction, Refinement and Replacement of Animals in Biological Testing. Animal Health Firm s Perspective VBS Public Meeting April 6, 2004

Outline CLINICALLY RELEVANT SPECIFICATIONS. ISPE Process Validation Conference September 2017 Bethesda, MD

MHRA Telephone Ref: 1099/Flutcore Ms Nathalie Gilmore Direct line: (0)

PROCESS VALIDATION ROCHAPON WACHAROTAYANKUN, PH.D.

GUIDELINE FOR THE STABILITY TESTING

Method Validation Studies How GLP Interacts With Guidance Documents

COMPARABILITY OF NOVEL THERAPEUTICS: CASE STUDIES FOR A BISPECIFIC T-CELL ENGAGER AND ONCOLYTIC VIRUS. FRANK MAGGIO, Ph.D. PRODUCT QUALITY PRINCIPAL

Notes from Round Table 5. January 26 and 27, 2016

Transcription:

Regulatory Challenges for the Licensure of Future Vaccines Tong Wu, Ph.D. Bacterial & Combination Vaccine Division, BGTD, Health Canada June 26-29, 2018, Seoul, Korea, the Global Bio Conference 1

Disclaimer The views and opinions expressed in the following presentation are those of the individual presenter and should not be attributed to Health Canada. 2

Outline Introduction Potency assay Reference standard Specification Clinical development to support quality specifications 3

Introduction 4

Challenges in vaccine development What does it take to market a new vaccine? 15-20 years on average. an investment of $US 800 million or more. thousands of individuals or more in clinical studies. regulatory scrutiny is unpredictable and not always science-based. Overall, the development of a new vaccine, especially the first against an infectious agent, is a complicated, time-consuming, expensive, risky, and competitive process. 5

Regulatory framework in Canada Life cycle approach Source: The Progressive Licensing Framework Concept Paper for Discussion, Health Canada, 2006; www.hc-sc.gc.ca/dhp-mps/alt_formats/hpfbdgpsa/pdf/prodpharma/proglic_homprog_concept-eng.pdf. 6

Requirements for regulatory approval of vaccines in Canada Regardless of manufacturing platform, target population or disease to be prevented, a vaccine must be: safe (acceptable safety profile) efficacious (or effective) of suitable quality No prescriptive or product-specific requirements guided by up-to-date science and technology Considerations for review and approval Case by case Risk/benefit assessment 7

Our goal is Quality Safety Efficacy Vaccine availability Science based regulatory standards and risk benefit based decision making are essential for ensuring timely access to new vaccines as well as managing vaccine supply postapproval. 8

Potency Assay 9

Why are potency assays required? A suitable potency assay, combined with a well managed reference standard and an appropriate specification: provides a bridge between clinical lots and commercial lots. assures that commercial lots are comparable to clinical lots that are shown to be safe and efficacious/effective in clinical studies. is a critical tool for life cycle management of a vaccine product. 10

Potency assays in vivo vs in vitro In vivo challenge models Relevance to clinical efficacy In vivo functional serological assays In vitro immunoassays In vitro physicochemical methods Assay sensitivity and consistency 11

Strategy for potency assessment throughout product life cycle At early development stage, in vivo potency assay is critical for Proof of concept Decision on excipients (e.g. adjuvant) and final formulation At clinical development stage, both in vivo and in vitro potency assays may be used to characterize antigenicity and stability of the vaccine verify suitability of in vitro potency assay for stability monitoring establish potency specifications Post-approval commercial manufacturing in vitro potency assay preferred assay for routine lot release, stability monitoring, supporting manufacturing changes in vivo potency assay may be used to support manufacturing changes (demonstrate product comparability) 12

Why in vitro potency assay for commercial manufacturing? Quality of commercial vaccine lots is assured through a combination of cgmp compliance, validated manufacturing process and QC testing. Consistency approach The goal of potency assessment assure that commercial lots have comparable potency results to that of clinical lots shown to be effective in clinical trials, therefore, expected to perform similarly in clinical. Use of in vivo potency assay (even challenge assays) alone can t predict clinical outcome, due to differences in immune systems (human vs animal models) 13

Why in vitro potency assay for commercial manufacturing? (cont d) A potency assays suitable for quality control of commercial lots should have consistent performance (low variability). Highly variable in vivo assay likely leads to high invalidity rate, and delays decision for lot release. adds challenge to management of reference standards In recent years, rejection of lots were largely due to method and references related issues (use of highly variable in vivo assay), not vaccine quality issues. Test issues, rather than vaccine quality, have led to vaccine shortages. Both vaccine manufacturers and regulatory scientists are currently working towards replacing in vivo potency assay with suitable in vitro potency assay. 14

Potency assay method development Research Understand mechanism of action Development Incorporate new science and technology Prevalidation Optimize assay parameters Validation Determine assay performance Regulatory scrutiny Regulatory review and approval 15

Points to consider when developing and validating potency assay In vivo potency assays are highly variable and less sensitive, it is critical to select appropriate immunization schedule. In general, one injection is preferred, and multiple injections should be justified. The immunization schedule should ensure that the immune response are within dose response curve. In vitro potency assay select appropriate antibody to ensure assay is stability indicating (e.g. monoclonal antibody specific to functional epitopes) For both in vivo and in vitro potency assays Establish assay sensitivity (e.g. using samples with known antigen content) Use appropriate reference standard 16

Reference Standard 17

Reference standard critical reagent for potency assay A carefully managed reference standard for potency is essential for assuring comparable vaccine potency throughout its life cycle. Reference standard for potency assay is used to: Calculate relative potency, in which the biological activity of a test sample is directly compared to that of the reference standard Set test validity criteria to ensure consistent performance of the potency assay over time Management of potency reference standard is challenging, especially when the new vaccine is the 1 st against an infectious agent. 18

Reference standard - challenges Vaccines are complex biologics Development and maintenance of vaccine reference standards is often more challenging when compared with those for well characterized biologics or small molecule drugs. Use of highly variable in vivo assays for reference calibration difficult to ensure accuracy Complex formulation/matrix difficult to preserve potency as freezingthaw may impact product integrity Critical QC tests are relative design lack of appropriate tools for stability monitoring All the above affect the values assigned to subsequent reference replacement, and impact the validity or the comparability of the test results. 19

Reference standard selecting suitable candidate material ICH Q6B requires that candidate material be representative of production and clinical materials Alternatively, a reference standard may be different from the test vaccine lots in its composition and/or storage conditions. However, the candidate material should have the following characteristics: The dose response curve of the candidate material is comparable to that of commercial vaccine lots to allow the calculation of the relative strength of the test sample The candidate material can be stored for a long period of time without decay. 20

Reference standard ensuring stability The composition and storage conditions (frozen, lyophilized or added stabilizer) of a reference standard should be carefully selected in order to preserve the integrity of the reference avoid the need for frequent replacement that may lead to drift/shift of the reference standard. Stability monitoring should be meaningful. Implement and trend a test readout, that is independent of the reference standard, if possible (e.g. ED 50 ) Link potency unit, especially arbitrary unit, to other quality attribute measured independently (e.g. protein content based on total nitrogen). 21

Specification 22

Potency specification regulatory expectation Specific to each vaccine product. ICH Q6B requires that specifications be linked to: clinical studies ensure specifications at end of shelf life (lowest for potency and highest for degradation impurities) not worse than those of clinical lots demonstrated to be safe and efficacious in clinical trials manufacturing process ensure comparable quality attributes of commercial and clinical lots stability of both drug substance and drug product set different specifications for release and end of shelf life for quality attributes that change during storage. analytical procedure comparable test performance (clinical development vs commercial production) and assay variability. 23

Potency specification: based on product release model WHO TRS 999, Annex 5 24

Potency specification: based on product release model (cont d) Different specifications for release and end of shelf-life for potency that declines through shelf-life. Potency specification at end of shelf-life (lower limit) should be supported by clinical lots demonstrated to be safe and efficacious (or immunogenic) in clinical studies. Potency specification at release should ensure vaccine lots meet specification at the end of shelf-life, based on rates of changes during storage be within manufacturing capability 25

How to support the lowest potency specification? Ideal situation: Vaccine lots with potency at the lower limit are tested in clinical study and shown to be efficacious or immunogenic. Reality: The lower boundary of potency, for most licensed vaccines, has not been tested in clinical studies. Flexibility is required to set a practical potency range for commercial manufacturing. Consider age of clinical materials at the point of use, combined with established potency decay rate, were considered during licensure. Commitment for post-licensure studies. For future vaccines: clinical development program should incorporate elements to support quality specifications. 26

Support quality specifications with clinical experience 27

How to support quality specifications with clinical experience? Test aged lots or lots stored under forced degradation conditions in clinical trials. This approach is ethical. Study subjects are monitored closely. The aged/degraded materials are likely to be safe and effective based on existing experience and knowledge. This type of clinical studies have been approved by other agencies (e.g. FDA for live attenuated viral vaccines), as well as by our division recently (intend to support the upper limit for free polysaccharide). Consider the results of dose-ranging study. The dose-ranging study should include a lose lower than what is intended for commercial product. Lots with different antigen content should be tested using the potency assay account for the performance of potency assay. 28

How to support potency specification when correlate of protection has not been established? It is impractical and cost prohibitive to conduct dose-ranging study using protection as end point. To support potency specification: Characterize and assess the immune responses globally of each dosing group, covering both cellular and humoral immunity. If both cellular and humoral immunity induced by the vaccine dose intended for commercial product is similar to that induced by vaccine with lower antigen content, the potency result of the vaccine with lower antigen may be used to set lower potency limit (assuming that efficacy of the vaccine candidate is demonstrated in Phase III clinical study). Preserve clinical samples for further studies, as correlate of protection may be established in future efficacy trials, or through advances in basic research. 29

Conclusion Developing a new vaccine is technically challenging and financially risky. Regulators must facilitate vaccine development process, and continue to enhance benefit-risk assessment in regulatory decision-making. promote science-based regulatory standards. Remove outdated requirements Implement improved quality control methods support innovation in vaccine development, manufacturing and quality control. 30

Thank you!? 31