FDA Perspective on the Preclinical Development of Cancer Vaccines

Similar documents
Beth Hutchins, PhD PhRMA ICH Gene Therapy Discussion Group

Regulatory Approval of Modern Gene-Based Cancer Immunotherapies CAR T Cells A product perspective

CQAs for C&GT Products to Enable Comparability Assessment. Ben Thompson Snr Director, Biopharmaceutical CMC RA GlaxoSmithKline

Regulatory considerations for manufacturing and testing of investigational chimeric antigen receptor (CAR) T-cell products

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

COMMITTEE FOR THE MEDICINAL PRODUCT FOR HUMAN USE (CHMP)

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP)

TECHNOLOGIES & SERVICES FOR THERAPEUTIC ANTIBODY DEVELOPMENT

FDA Oversight of Gene Therapy

FDA Perspectives On Viral & Vector Shedding Studies. Daniel Takefman, Ph.D. Chief, Gene Therapy Branch FDA/CBER ICH Workshop: October 31, 2007

Manufacturing Challenges with Cell and Gene Therapy Products: A Health Canada Perspective

S9 Implementation Working Group ICH S9 Guideline: Nonclinical Evaluation for Anticancer Pharmaceuticals Questions and Answers

Regulatory Pathways for Rare Diseases

Current situation on nonclinical safety evaluation of regenerative medical products in Japan

Mitochondrial Manipulation Technologies: Preclinical Considerations

Guidelines on the nonclinical evaluation of vaccine adjuvants and adjuvanted vaccines

Assessing and Controlling Potency of Vector and Drug Product for Chimeric Antigen Receptor T Cells

Defining Clinical Benefit in Clinical Trials: FDA Perspective

What are ATMP s: Why do they Require Special Scientific and Regulatory Attention? Christopher A Bravery.

Biomarkers, Early Prediction of Vaccine Efficacy and Safety

Drug Development: Why Does it Cost so Much? Lewis J. Smith, MD Professor of Medicine Director, Center for Clinical Research Associate VP for Research

GREAT STRIDES IN SCIENCE WHAT ABOUT ACCESS? Marlene E. Haffner, MD, MPH 2nd IRDiRC Conference Shenzhen, China November 8, 2014

Overview of Biologics Testing and Evaluation: Regulatory Requirements and Expectations. Audrey Chang, PhD, Senior Director Development Services

Preclinical Drug Development

HSV-2 therapeutic vaccine program. Subunit vaccine candidates

Ancillary Materials for Cell & Tissue Therapies Definitions, US Regulatory Approach, and USP s Risk-Tiered Approach

Strategies for Assessment of Immunotoxicology in Preclinical Drug Development

Pharmacology. Chatchai Chinpaisal, Ph.D. Department of Pharmacology and Toxicology, Faculty of Pharmacy, Silpakorn University.

Preclinical Development Drugs. Darrin Cowley PhD Executive Director Amgen BioBoot Camp 2015

OSWG s Role in Shaping the Regulatory Development of Oligos - A Potential Model for Moving Forward -

Regulation of Microbiota- Based Products

"From Bedside to Bench and Back: regulatory requirements for collaborations between pharma industry and academia

COMMITTEE FOR VETERINARY MEDICINAL PRODUCTS NOTE FOR GUIDANCE 1 : DNA VACCINES NON-AMPLIFIABLE IN EUKARYOTIC CELLS FOR VETERINARY USE

Baek, Kyung-min. Recombinant Protein Products Division. Ministry of Food and Drug Safety

Outcomes in Mesenchymal Stem Cell Manufacturing. Athena Russell, MT(AAB) Human Cellular Therapy Laboratory Mayo Clinic Jacksonville, FL

Optimisation de votre programme de développement

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

Long-Term Follow-Up in Gene Transfer Clinical Research

Guidance for Industry

US FDA: CMC Issues for INDs

FDA Perspectives on Biomarkers. Steven Kozlowski, M.D. Office of Biotechnology Products OPS/CDER/FDA

1 Name. 1. (3 pts) What is apoptosis and how does it differ from necrosis? Which is more likely to trigger inflammation?

Oncology Biopharmaceuticals and Preclinical Development: Evolving Regulatory Challenges

Global Regulatory Perspective Workshop

PACT. PACT Program. Production Assistance for Cellular Therapies

Guidance for Industry

Regulatory Perspective

Course Descriptions. BIOL: Biology. MICB: Microbiology. [1]

Immunological Techniques in Research and Clinical Medicine. Philip L. Cohen, M.D. Chief of Rheumatology, LKSOM 10 March 2016

First Annual Biomarker Symposium Quest Diagnostics Clinical Trials

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP) GUIDELINE ON THE NON-CLINICAL DEVELOPMENT OF FIXED COMBINATIONS OF MEDICINAL PRODUCTS

Development of Multiplex Sensitive Anti-Drug Antibody Assays for CRISPR/Cas9 Gene Therapies

SUBMISSION OF COMMENTS ON The Requirements for First-in-Human Clinical Trials for Potential High Risk Medicinal Products EMEA/CHMP/SWP/28367/2007

Applied Protein Services

Industry Academic Collaboration: A Key to Successful Involvement of Patients Early in Clinical Development

1 R01 AI VMD HALFORD, W

Safety Assessment of Biotechnology-Derived Pharmaceuticals:

IBC protocol Risk Assessment and Determination of NIH Guidelines

DNA Microarray Technology

Measurement of Hematopoietic Stem Cell Potency Prior to Transplantation

Individualised medicine: regulatory challenges

BIOPHARMACEUTICAL PROCESS EVALUATED FOR VIRAL CLEARANCE

An Overview of Chimeric Antigen Receptor T-cells: CAR-T-ing Away Cancer

KFDA Regulatory Framework on Biopharmaceuticals - Focus on Biosimilar

The science behind Betalutin : why is it unique? Roy H. Larsen PhD Sciencons AS, Oslo, Norway

Guidance for Industry

How can innovation in regulatory science inform the regulatory process to facilitate the development of new vaccines?

Chapter 17: Immunization & Immune Testing. 1. Immunization 2. Diagnostic Immunology

Updates to the NIH Guidelines for Research Involving Recombinant DNA Molecules (NIH Guidelines)

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

UNCLASSIFIED. FY 2016 Base

Nanotechnology: A Brief History and Its Convergence with Medicine. Weston Daniel, PhD Director of Program Management

Platelet Refractoriness: The Basics. Martin H. Bluth, MD, PhD

Revised Immunogenicity Guideline: Assays and methods- Presentation of the draft guideline and introduction of the topics for discussion

February 28, Churchill Place Canary Wharf London E14 5EU United Kingdom

Global Leader in Viral Vector Technologies

Development of non-substantially manipulated cell-based ATMPs 1 : flexibility introduced via the application of the risk-based approach

Update from the Center for Biologics Evaluation and Research (CBER): Advancing the Development of Complex Biologic Products

Managing Specimen Stability for Robust Flow Cytometric Clinical Biomarker Assays

Reflection paper on co-development of pharmacogenomic biomarkers and Assays in the context of drug development

HCT/P Regulation vs 361 Products

Issues in production of viral gene transfer vectors. Stefan Kochanek Department of Gene Therapy Ulm University

Peptide libraries: applications, design options and considerations. Laura Geuss, PhD May 5, 2015, 2:00-3:00 pm EST

The Road to Functional Bioanalysis: Development and Validation of a Cell-Based Assay for Neutralizing Anti-Drug Antibody Analysis

Janice Soreth, M.D. Deputy Director FDA Europe Office Liaison to EMA. Making Gene and Cell Therapy a Reality July 10-11, 2012 Dublin

The Role of Mass Spectrometry for Developing Biotherapeutics: Regulatory Perspectives

Guidelines: c. Providing the investigational drug for the requested use will not interfere with the initiation, conduct, or completion of clinical

Integrating an exploratory BM in an early clinical stage in Pharma R&D

8. Clinical Trial Assessment Phase II

Cytomics in Action: Cytokine Network Cytometry

Strategy for Selecting NAb Assay Format

INDUSTRY PERSPECTIVE: REGULATORY ASPECTS OF DEVELOPING PERSONALIZED CANCER VACCINES

Flow CAST : Testing Potency and Efficacy of Inhibitors of PI3K δ, PI3Kγ, BTK and SYK Activity

PRECLINICAL DRUG DISCOVERY AND DEVELOPMENT BIOBOOT CAMP 2016

De-risking Vaccine Formulation Design

Regulatory Requirements for CRISPR Therapeutics. Bill Lundberg, MD February 2017

The Laboratory-based Physician Scientist

CHANGES TO A GENE THERAPY MEDICINAL PRODUCT: REGULATORY VIEWS

Transcription:

FDA Perspective on the Preclinical Development of Cancer Vaccines Richard D. McFarland Ph.D., M.D. Medical Officer CBER/OCTGT/DCEPT mcfarlandr@cber.fda.gov Cancer Vaccine Clinical Trials Workshop Alexandria, VA November 10, 2005 The views expressed in this document do not necessarily reflect the policies of the USFDA and USHHS.

Safety is Always Primary FDA s primary objectives in reviewing an IND are, in all phases of the investigation, to assure the safety and rights of subjects, and, in Phase 2 and 3, to help assure that the quality of the scientific evaluation of drugs is adequate to permit an evaluation of the drug s effectiveness and safety. IND Regulations [21 CFR 312.22 (a)]

Safety of a Biological Product is Relative The relative freedom from harmful effects of the recipient when a product is prudently administered, taking into consideration the characteristics of the product in the relationship to the condition of the recipient at the time; thus the property of safety is relative. 21 CFR, Subchapter F

FDA Review is Product-based Parallels prudent product development Dependent on characteristics of specific product Preclinical studies designed to support use of specific products Clinical trial design supported by manufacturing, preclinical data Supported by science, framed by regulations

Classes of Tumor Vaccines Currently in Development Conventional Antigen-based vaccines (Ag) Purified Ag Synthetic peptides Conjugated Ag Tumor cell lysates Cell-based vaccines Manipulated tumor cells Activated peripheral blood or BM-derived lymphocytes DCs or other APCs [modified (i.e. cell fusion)] Gene-modified tumor cells Cells engineered to express cytokines, growth factors, or tumor Ags

Classes of Tumor Vaccines Currently in Development Gene Therapy-based vaccines Plasmid Human tropic viruses (adenovirus) Retroviral based vaccine vectors Non-human tropic based vaccine vectors (fowlpox) Bacterial based vectors (Listeria sp.)

Potential Modifying Factors Adjuvants Immunomodulation Concomitant Conventional Chemotherapy Formulation (liposomes) Route of Administration Site Delivery Device

Phase 1 Preclinical Expectations Scientific basis for conducting clinical trial Feasibility/establishment of rationale Establish pharmacologically effective dose(s) Optimize ROA/dosing regimen Rationale for species/model selection for further tests Recommend initial safe dose & dose escalation scheme in humans Identification of potential target tissue(s) of toxicity/ activity Identification of parameters to monitor clinically Identification of patient eligibility criteria

Phase 1 Preclinical Expectations Proof of concept and sufficient toxicity data Final product (Vaccine and adjuvant, etc) Each component separately Vaccine Adjuvant or immunomodulating agent Liposome components Concomitant conventional chemotherapies

Potential Sources of Data to Support Initiation of Clinical Trials Preclinical studies specifically designed to support a clinical trial Other potential sources Existing animal studies designed to answer other questions In vitro studies Clinical trials using the same product

Using Published Animal or Human Studies as Sole Support for Initiation of Clinical Trials Often they were not designed to answer a toxicologic question, and therefore, adequate toxicology endpoints may not have been incorporated into the design Published reports must provide sufficient information for independent review

Flexible Animal Study Designs Proof of concept activity studies in animal model(s) of disease Toxicology/biodistribution studies in healthy animals (traditional approach) Hybrid pharmacology-toxicology study design in animal model(s) of disease Activity endpoints Tissue localization Toxicology endpoints

Proof of Concept (Activity) Models From FDA s perspective, the specific choice of the activity model(s) is less important than the underlying rationale for therapy in humans supporting the choice. This generally involves understanding the therapy s putative mechanism of action and demonstrating proof-of-efficacy in an appropriate preclinical model.

Examples of Proof-of-Concept Models Tumor cell survival (cell lines and clonogenic assays) Human tumor xenograft murine models Metastasis models Transgenic and knockout murine models Carcinogen-induced tumor models Transplantable syngeneic rodent tumors

Bioactivity Endpoints of Potential Relevance to Tumor Vaccine Development MHC-peptide tetramer assays (Ag-specific T subsets) ELISPOT to assess T cell subset # s, cytokine profiles (γ IFN production) Antibody titers (humoral response) Quantitative RT-PCR assays TCR dysfunction (α-cd3 stimulation of T cells, TCR ζ chain expression) Limiting dilution assays (Ag-specific CD8, 51 Cr release, thymidine incorporation) Cytokine flow cytometry

Bioactivity Endpoints FDA cannot comment as to which immunoassays represent the best measures of immune function or correlation with survival; as these data are not yet available. The decision to choose one immune response parameter over another as the appropriate surrogate endpoint has more to do with the scientific question(s) being asked than correlation with a relevant clinical endpoint in a given malignancy.

Preclinical Toxicology Assessment For tumor vaccines often traditional animal models for toxicity evaluation often yield limited information, especially where human-specific Ags are being targeted. No one animal model can address all concerns or fulfill all necessary criteria to mimic humans. In such situations (e.g. autologous DC vaccines, human HLAspecific therapies), information about the antigen, construct, etc. involved and its tissue distribution and expression in humans is very important (e.g. mrna expression, tissue crossreactivity studies where mab against the Ag have been tested, genomic database searches).

Tissue Cross-Reactivity Studies Should be performed prior to human exposure for novel antigens with potential for expression in normal tissues. Tissue binding should be determined using appropriate immunohistochemical procedures. A range of human tissues should be tested. Analogous study in primary pre-clinical toxicity species may help substantiate species relevance of the animal model.

Choice of Animal Species for Toxicity Testing Should be Based on Expression and Homology of the Targeted Antigen Animal models expressing the targeted Ag or epitope of interest, when available, are more likely to reveal the effects of Ag sinks or tissues with unexpected Ag expression on biodistribution tissue localization and/or toxicity. Critically important for assessing risk of autoimmunity. (+) signals in in vitro testing: incorporate relevant safety monitoring into preclinical, clinical studies. (labs: ANA, RF, CH50, physical exam monitoring: cytopenias, arthalgias, rash).

Animal Models: GT Vector-based Vaccines May need to evaluate tissue biodistribution and persistence; especially in germ-line tissues. When aberrant or unexpected localization is observed, studies should be conducted to determine whether the gene is expressed, and whether its presence is associated with pathologic changes. May need assessment of replication competent virus Potential for an overly robust inflammatory response to viral, bacterial vectors (e.g. adenovirus). Evaluation for adventitious viruses. May require long-term animal studies.

Transgenic Animal Models: Pros and Cons Pros: May express the human protein of interest and/or may express the appropriate human HLA molecule (e.g. HLA-A2.1) necessary for immune responsiveness. With conditional transgenics can activate a gene in a cell-specific manner. Easier to handle than non-human primates

Transgenic Animal Models: Pros and Cons Cons: Transgenics appear to respond to most, but not all human HLA-restricted epitopes to which humans respond. Genetic drift and non-uniformity can be a problem. Significant inter-experiment variability in response magnitude has been described.

Animal Models: Which Are Best? Depends on the question asked: For solid tumor malignancies: solid tumor xenograft models may mimic human disease more than tumor cell lines administered to animals. Animal model may not express the relevant human receptor or protein, hence might need to rely on a transgenic model or evaluate the effect of therapy on the animal homologue of the human protein.

Role of Genomics, Proteomics in Tumor Vaccine Development: The Pros Believed useful in drug discovery/identification of targets, understanding disease pathways and a new therapy s mechanism of action. In principle, can monitor complete biochemical pathways rather than single biomarkers. Identification of bridging biomarkers to monitor key damage responses in lab animal models and humans. Identification of polymorphisms that may modify sensitivity to disease or treatments. Potential to reduce animal use in vaccine development (3Rs)

Role of Genomics, Proteomics in Tumor Vaccine Development: Cons Non-quantitative and of unknown predictive value. Need to establish the relationship of chosen microarray endpoints with accepted preclinical and/or clinical endpoints and will need to validate these. In order to be clinically useful, patterns of correlation will need to exist, that are reproducible (heretofore a problem with array platforms).

Early Communication ( pre-pre- IND ) Non-binding, informal scientific discussion between FDA and sponsor Via telecons Via scientific meetings/workshops Via outreach presentations (i.e., this meeting) Provide pre-read materials to FDA Discuss specific issue (s) of interest A two-way communication to allow for information exchange

Pre-IND Meeting Submit a pre-ind package to include: Product development/characterization Chemistry, Manufacturing and Controls (CMC) Summary of device information Bench and/or in vivo Summary of preclinical information Pharmacology/Toxicology - in vitro and/or in vivo Proposed clinical protocol

Concluding Remarks

Concluding Remarks The animal model and extent of primary data required for any particular product should be science-based, but framed by regulatory requirements to protect the safety of human subjects in all phases of clinical investigations. The decision to allow a therapy to go forward into a phase 1 trial involves a risk:benefit assessment of safety concerns vs. the likely clinical response based on supportive preclinical & clinical data (similar products).

Contact Information Richard McFarland Ph.D., M.D. E-mail- mcfarlandr@cber.fda.gov Voice- 01-301-827-5102 Fax- 01-301-827-9796 Food and Drug Administration Center for Biologics Evaluation and Research 1401 Rockville Pike, Woodmont I, HFM-760 Rockville, Maryland 20852 USA