Integrating Scientific Advances into Regulation: Pharmacogenomics and Pharmacogenetics Janet Woodcock, M.D. Director, Center for Drug Evaluation and Research.

Slides:



Advertisements
Similar presentations
The Drug Discovery Process
Advertisements

Regulatory Framework Leigh Shaw, Director.
Retreat Topics iPSC Opportunities in NIAMS Diseases Science Management Forum: Leveraging and Strategic Funding Collaborations Atopic Dermatitis Advancing.
FDA Pharmacogenetic Labels A Clinical Perspective David A Flockhart MD, PhD Indiana University School of Medicine Clinical Pharmacology Subcommittee of.
Safety and Extrapolation Steven Hirschfeld, MD PhD Office of Cellular, Tissue and Gene Therapy Center for Biologics Evaluation and Research FDA.
Pharmacogenetics: Global Regulatory Issues ASENT 13 th Annual Meeting Bethesda, MD February 26, 2011 Issam Zineh, PharmD, MPH Associate Director for Genomics.
1 Pharmacology/Toxicology information to submit an IND for an anticancer drug.
Mitochondrial Manipulation Technologies: Preclinical Considerations
U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES NATIONAL INSTITUTES OF HEALTH Working with FDA: Biological Products and Clinical Development Critical Path.
The Pursuit of Better Medicines through Genetic Research Terri Arledge, DVM US Department Head Drug Development Genetics.
CBER Regulatory Laboratory Planning & Preparedness for SARS-related Biologics Products Kathryn M. Carbone MD Associate Director for Research, Acting, Center.
Pharmacogenomics in Drug Development FDA Science Board April 9, 2003 Brian B. Spear, Ph.D. Director, Pharmacogenomics Abbott Laboratories.
What Do Toxicologists Do?
Importance of Pharmacogenetics in Oncology Richard Pazdur, MD. Director Office of Oncology Drug Products Center for Drug Evaluation and Research Food and.
Special Topics in IND Regulation
Guidance for Industry Establishing Pregnancy Registries Pregnancy Registry Working Group Pregnancy Labeling Taskforce March, 2000 Evelyn M. Rodriguez M.D.,
Stefan Franzén Introduction to clinical trials.
+ Drug Development and Review Process. + Objectives Learn the processes involved in drug discovery and development Define the phases involved in FDA drug.
Cellular Therapies for Cardiac Diseases – FDA Preclinical Perspective Richard D. McFarland Ph.D., M.D. CBER/OCTGT/DCEPT BRMAC # 37 March 18-19, 2004.
Neonatal/Juvenile Animal Safety Studies Kenneth L. Hastings, Dr.P.H., D.A.B.T. Office of New Drugs, CDER.
Principles of New Animal Drug Effectiveness: An Overview
Biomedical research methods. What are biomedical research methods? An integrated approach using chemical, mathematical and computer simulations, in vitro.
Food and Drug Administration Preclinical safety data for “first in human” (FIH) clinical trials in healthy volunteer subjects Oncology Drug Advisory Committee.
Howard Fillit, MD Executive Director Improving Animal Trials for Alzheimer’s Disease: Recommendations for Best Practices.
Scientific Data for Evidence- Based Drug Regulation Janet Woodcock, M.D. Director, Center for Drug Evaluation and Research Food and Drug Administration.
1 Safety Pharmacology for Oncology Pharmaceuticals at CDER John K. Leighton Associate Director for Pharmacology CDER/OND/OODP.
Richard D. Hockett, Jr. M.D. Sr. Clinical Research Physician Group Leader, Genomic Medicine FDA Clinical Pharmacology Advisory Committee Integrating Pharmacogenomics.
Stefan Franzén Introduction to clinical trials.
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,
Chapter 13. The Impact of Genomics on Antimicrobial Drug Discovery and Toxicology CBBL - Young-sik Sohn-
Pharmacogenomics. Developing drugs on the basis of individual genetic differences Tailoring therapies to genetically similar subpopulations results in.
The Accelerating Pace of Medical Development and Challenges in Evaluating Benefit and Risk Alasdair Breckenridge Medicines and Healthcare products Regulatory.
Animal Models for Porcine Xenotransplantation Products Intended to Treat Type 1 Diabetes or Acute Liver Failure CTGTAC #47 May 14, 2009.
Exploratory IND Studies
Nonclinical Perspective on Initiating Phase 1 Studies for Small Molecular Weight Compounds John K. Leighton, PH.D., DABT Supervisory Pharmacologist Division.
Clinical Trial Designs An Overview. Identify: condition(s) of interest, intended population, planned treatment protocols Recruitment of volunteers: volunteers.
Investigational Drugs in the hospital. + What is Investigational Drug? Investigational or experimental drugs are new drugs that have not yet been approved.
The Value of Tissue Banks to Drug and Dx Developers Barbara L. Handelin, Ph.D. Conflicts of Interest, Privacy/Confidentiality, and Tissue Repositories:
Pharmacogenetics & Pharmacogenomics Personalized Medicine.
Proposal for End-of-Phase 2A (EOP2A) Meetings Advisory Committee for Pharmaceutical Sciences Clinical Pharmacology Subcommittee November 17-18, 2003 Lawrence.
Update: Topics Previously Presented to the CPSC Clinical Pharmacology Subcommittee (CPSC) of the Advisory Committee for Pharmaceutical Science November.
Welcome and Introduction Lee S. Simon, MD Division Director Analgesic, Anti-inflammatory and Ophthalmologic Drug Products ODEV/CDER.
The New Drug Development Process (www. fda. gov/cder/handbook/develop
Clinical Pharmacology Subcommittee of the Advisory Committee for Pharmaceutical Science Meeting April Quantitative risk analysis using exposure-response.
The FDA: Basic Facts It takes 12 to 15 years to develop a single drug Only 1 in 10,000 potential medications makes it completely through the process Only.
History of Pediatric Labeling
Privacy Symposium / HIPAA Summit
General Regulatory Issues in the Development of Drugs Intended for Treatment of Chronic Illness Sharon Hertz, M.D. Medical Officer Division of Anesthetic,
Kathleen Giacomini, Mark J. Ratain, Michiaki Kubo, Naoyuki Kamatani, and Yusuke Nakamura NIH Pharmacogenomics Research Network III & RIKEN Center for Genomic.
Introduction to the Meeting Introduction to the Meeting Advisory Committee for Pharmaceutical Sciences Clinical Pharmacology Subcommittee November 17-18,
Integrating Pharmacogenomic Questions Into GCIG Ovarian Cancer Clinical Trials Lori Minasian, MD Chief, Community Oncology and Prevention Trials Research.
Consumer Advocate Perspective Clinical Trials Registration Sharon F. Terry, JAM Sharon F. Terry, JAM President and CEO, Genetic Alliance, Inc. Founding.
European Patients’ Academy on Therapeutic Innovation The key principles of pharmacology.
Regulatory Guidance for Genetic Testing. Three Specific Areas Laboratory tests Results of genetic testing – Clinical – Research GenomeWide Association.
Applying New Science to Drug Safety Janet Woodcock, M.D. Acting Deputy Commissioner for Operations April 15, 2005.
Ethical issues with the regulatory use of gene expression data Benjamin S Wilfond MD Medical Genetics Branch National Human Genome Research Institute Department.
NIAMS Extramural Scientific Planning Retreat 2013 April 4, 2013 Session 1: Strategies for Successful Rare Disease Clinical Trials.
Clinical Trials - PHASE II. Introduction  Important part of drug discovery process  Why important??  Therapeutic exploratory trial  First time in.
E-Clinical
Study Development and Design Suzanne Adams RN MPH Director, Clinical Operations Jefferson Clinical Research Institute.
Drug Discovery &Development
Regulatory– Terms & Definitions רגולציה - מונחים והגדרות
Mahla sattarzadeh Kerman University of Medical Sciences
Pharmacogenomics Genes and Drugs.
Drug Design and Drug Discovery
FDA Tox Working Group Which goals of the FDA Roadmap are most important to FDA stakeholders? Integration of predictive technologies Replacing animal tests.
Introduction to Pharmacogenetics
Regulatory Perspective of the Use of EHRs in RCTs
Considerations for Successful Biopharmaceutical Product Development: Discovery to Proof of Concept -A Panel Discussion Stanley C. McDermott, PharmD, MS,
Presentation transcript:

Integrating Scientific Advances into Regulation: Pharmacogenomics and Pharmacogenetics Janet Woodcock, M.D. Director, Center for Drug Evaluation and Research April 8-9, 2003

Translation of innovative science to bedside medicine

Issue: b b New science of pharmacogenomics (and increasingly, proteomics) applied extensively in drug development b b Potential to revolutionize process b b Most of the data not seen by regulatory agencies, partly out of concern for how it will be used b b Need an approach that will enable free exchange of information, help advance the science & technology & aid in the timely development of appropriate regulatory policies

Background VARIABILITY IN HUMAN RESPONSE TO DRUGS: A major barrier to effective therapeutics

Variable Effectiveness b b For many drugs (leaving aside antibiotics, antivirals, etc)… b b Size of treatment effect from randomized trials is < 10% of outcome measure b b Many conclude that effect is “small” or that “drug doesn’t work”

Mean Response PlaceboDrug

Number of subjects DrugPlacebo Response

Variability in Drug Toxicity b b Drug vs placebo: each drug has consistent pattern of side effects over placebo rate. b b Observed with common as well as rare events b b Some attributable to known pharmacologic effects; others “idiosyncratic” b b Current medical approach is at the level of organ function, or is observational

There is an Inherited (Genetic) Component to Variability in Drug Response b b Pharmacogenomics (PG): Application of genome-wide RNA or DNA analysis to study differences in drug actions b b Pharmacogenetics : Study of genetic basis for interindividual PK differences

Efficacy Response: Three Types of Genetic Variables Contribute b b Genetic diversity of disease pathogenesis: cholesterol ester transfer protein ? b b Variable drug metabolism: hypermetabolizers b b Genetically based pharmacodynamic effects  -adrenergic receptor?

Drug Toxicity: Genetic Contributions to Variability b b Genetically based interacting state Long QT syndrome b b Differences in drug metabolism Thiopurine methyltransferase b b “Toxicodynamic” interactions Abacavir?

How Important are these Differences? How much of the variability will be explained by genetic differences?

At the Level of the Individual, a Genetic Difference may: b b Determine drug response Enzyme deficiency disease b b Highly influence drug response Polymorphic drug metabolizing enzymes

Individual Drug Response b However, many responses will be--- Emergent Property of multiple gene product interactions with each other and with environmental factors b Many individual genetic differences - or even patterns of differences - may have a b Many individual genetic differences - or even patterns of differences - may have a small effect on drug response

Oltvai and Barabasi, Science, 298, 25 Oct 02.

Current Drug Development b b Satisfactory determination of efficacy--but on a population basis b b Determination of drug toxicity is observational--based on animal and then human exposures b b Carcinogenic and reproductive toxicity potential based on in vitro and animal studies

Potential Uses of PG in Drug Development b b Improve candidate drug selection b b Develop new sets of biomarkers for toxic responses in animals and humans-- eventually minimize animal studies b b Predict who will respond to a drug b b Predict who will have serious side effects b b Rationalize drug dosing

Potential Impact of PG on Drug Development b b Move from current empirical process to mechanism-based process, hypothesis driven b b Lower cost, faster process resulting in more effective, less toxic drugs for smaller population

How is PG being used now?

PG and Drug Development: Discovery and Lead Candidate b b Target identification b b Evaluating cellular or animal responses to different candidates b b Not part of regulatory submissions

PG and Drug Development: Nonclinical b b Exploratory studies: cells and animals b b Directed studies: genes of interest b b Explanatory studies: evaluate an observed toxic response b b “Toxicogenomics”: develop predictive response patterns

PG and Drug Development: Human Studies b b Sort disease syndromes into subgroups based on genetic differences in pathogenesis and evaluate differential responses to treatment b b Evaluate use of genetic/phenotypic tests for metabolizer status to predict dosing

Human Studies b b Search for genetic differences in “responders” vs “nonresponders-- markers for different PD response b b Seek genetic explanation for severe or catastrophic side effects b b STET and gene expression screening

Obligations of Drug Regulators b b Determine if drug is safe and effective b b Protect human subjects enrolled in trials

Legal requirements: FD &C Act b b Safety: evaluate reports of “all methods reasonably applicable to show whether or not such drug is safe for use under the conditions…in the proposed labeling” b b Effectiveness: “adequate and well controlled trials” to show that “the drug will have the effect it purports to have under the conditions of use”

IND Submission requirements: 21 CFR (a) (8) b Pharmacology and toxicology information “on the basis of which the sponsor has concluded that it is reasonably safe to conduct the proposed clinical investigations”

NDA Submission requirements: 21 CFR  Nonclinical studies. d (2) (I): submit “studies that are pertinent to possible adverse effects” b Clinical d (5) (iv): submit “data or information relevant to an evaluation of the safety and effectiveness of the drug product”

Question b b When/how to use developing PG information in regulatory decisions? b b When is the information “reasonably applicable” to safety?” b b Under what circumstances is submission to FDA needed?

Proposal b b FDA will establish policies on categories of PG studies b b “Submission requirement” will be included in policy Submission not required for some types Submission required for other types Some with no regulatory impact

Proposal b “Regulatory impact” will be included in policy b Results from some study categories will not have any regulatory impact b Other study results will be utilized as part of safety/efficacy evaluation

Proposal b Possible threshold determination: Does genomic information represent valid biomarker with known predictive characteristics? b Develop threshold and policies using public and transparent process with advisory committee oversight

Possible Procedures b FDA would establish Interdisciplinary PG Review Group (IPGRG) b Categorization of studies and internal procedures published in guidance b Results submitted to IND or NDA as “research information package” for review by IPGRG b Periodic public re-evaluation of decision tree

Examples: With Regulatory Impact b b Trial enrollment by genotype: enrichment of responders avoiding bad outcomes b b Selection of dose based on metabolizer genotype

Examples: With Regulatory Impact (cont.) b b Safety rationale based on animal genomic data--i.e., explaining why a toxic finding is unique to that species b b In general- results intended to influence the course of the clinical development process will be considered part of the S&E evaluation

PG results without regulatory impact b Evaluation of new transporter gene diversity vs response in clinical subjects b Genomic SNP data collection in clinical trial subjects b Gene expression microarray screen in trial subjects b Gene expression microarray screen in animal toxicology study

Questions for Discussion b Is this approach reasonable? Feasible? b Will it achieve objectives Free exchange of data? Ability of FDA scientists to begin developing framework for new findings? Advance the use of the new science ? Advance the use of the new science ?