Applying New Science to Drug Safety Janet Woodcock, M.D. Acting Deputy Commissioner for Operations April 15, 2005.

Slides:



Advertisements
Similar presentations
Mary Ellen Turner MD, MPH Vice-President
Advertisements

ACCF/AHA Clopidogrel Clinical Alert: Approaches to the FDA “Boxed Warning” A Report of the American College of Cardiology Foundation Task Force on Clinical.
Office of New Animal Drug Evaluation Laura L. Hungerford, DVM, MPH, PhD Senior Advisor, Science and Policy, ONADE Professor, University of Maryland School.
Susan Boynton, VP, Global Regulatory Affairs, Shire
Importance of Vaccine Safety Decreases in disease risks and increased attention on vaccine risks Public confidence in vaccine safety is critical Low tolerance.
Analysis of Electronic Medical Record Systems Jonathan S. Schildcrout.
Study Designs in Epidemiologic
1 Proposed Pharmacovigilance Plan for H5N1 Influenza Virus Vaccine Patrick Caubel, MD, PhD Head of Pharmacovigilance North America February 27, 2007.
Assessing the Safety of Marketed Drugs Current Issues and Controversies Captain Paul J. Seligman, M.D., MPH Associate Director, Safety Policy and Communication.
Capturing and Reporting Adverse Events in Clinical Research
U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES NATIONAL INSTITUTES OF HEALTH Working with FDA: Biological Products and Clinical Development Critical Path.
Special Topics in IND Regulation
Objectives Why we need DHCPL Situations that call for a DHCPL Definitions DHCPL itself–content, presentation, process Target audience Current and future.
U of Arizona Innovation Conference 20 September 2011 Marlene E. Haffner, MD, MPH Haffner Associates, LLC.
Office of Drug Evaluation IV, CDER FDA/IDSA/ISAP Workshop 4/16/04 Overview of PK-PD in Drug Development Programs: FDA Perspective FDA/IDSA/ISAP Workshop.
Postmarketing Risk Assessment of Drug Products Division of Drug Risk Evaluation Office of Drug Safety Center for Drug Evaluation and Research.
Pilot Risk-Ranking Model to Prioritize Manufacturing Sites for GMP Inspections Advisory Committee for Pharmaceutical Science Manufacturing Subcommittee.
Biomedical research methods. What are biomedical research methods? An integrated approach using chemical, mathematical and computer simulations, in vitro.
Clinical Pharmacy’s Role in Research Trials Sheree Miller Pharm.D. Investigational Drug Service University of Washington Medical Center.
1 The Chemoprevention of Sporadic Colorectal Cancer Issues Surrounding a Benefit/Risk Analysis in Clinical Trials Mark Avigan MD CM Medical Officer Division.
Vaccine Safety Epidemiology and Prevention of Vaccine- Preventable Diseases National Immunization Program Centers for Disease Control and Prevention Revised.
Patient Safety Initiatives Kelly Cronin Director, Patient Safety and Outcomes Research Office of Policy and Planning Office of the Commissioner FDA.
1 Lotronex ® (alosetron HCl) Tablets Risk-Benefit Issues Victor F. C. Raczkowski, M.D. Director, Division of Gastrointestinal and Coagulation Drug Products.
IPhVWP Polish Presidency, Warsaw October 6 th 2011 Almath Spooner Irish Medicines Board Monitoring the outcome of risk minimisation activities.
Eureka Pre-Clinical Investigation Animal toxicology Animal pharmacokinetics/ pharmacodynamics Clinical Investigation Phase I Safety and pharmacology Phase.
Integrating Scientific Advances into Regulation: Pharmacogenomics and Pharmacogenetics Janet Woodcock, M.D. Director, Center for Drug Evaluation and Research.
Center for Drug Evaluation and Research August 2005 Electroretinography: The FDA’s Viewpoint Wiley A. Chambers, MD Deputy Director Division of Anti-Infective.
Copyright © CRF Box, Ltd. All rights reserved. 1 Introduction to Electronic Data Collection Methods; Improving Data Quality and Integrity in Epidemiological.
Stefan Franzén Introduction to clinical trials.
1 Post Marketing Plan Earl Sands, M.D. Vice President and Chief Medical Officer US Research and Development Solvay Pharmaceuticals Earl Sands, M.D. Vice.
First In Human Pediatric Trials and Safety Assessment for Rare and Orphan Diseases Andrew E. Mulberg, MD, FAAP Division Deputy Director OND/ODE3/DGIEP.
PHARMACOVIGILANCE AND CLINICAL TRIALS DIVISION 20 August 2015 Victoria Falls Protecting Your Right to Quality Medicines and Medical Devices.
Reproductive Health Drugs, Pregnancy Labeling Subcommittee Meeting March 28-29, 2000 Holli A. Hamilton, M.D., M.P.H. Pregnancy Labeling Team Office of.
From the Lab to Market Unit 3.04 Understanding Biotechnology research & Development.
Biomedical Research.
Investigational Drugs in the hospital. + What is Investigational Drug? Investigational or experimental drugs are new drugs that have not yet been approved.
Study Designs in Epidemiologic
Update From FDA: Office of the Commissioner and Center for Drug Evaluation and Research Janet Woodcock, M.D. Acting Deputy Commissioner for Operations.
1Presentation Name Pre-Marketing Safety Assessment: The Safety Review Guidance Armando Oliva, M.D. Associate Director for Policy Office of New Drugs.
Risk Management in premarketing phase Anshu Vashishtha MD PhD (in individual capacity employer : Watson Pharmaceuticals)
DEVELOPING EVIDENCE ON VACCINE SAFETY Susan S. Ellenberg, Ph.D. Center for Clinical Epidemiology and Biostatistics U Penn School of Medicine Global Vaccines.
1 Evaluating & Applying What Works Best Leaders’ Project Policy Forum Kathy Buto, VP Health Policy April 24, 2008.
Developing medicines for the future and why it is challenging Angela Milne.
Drug Safety and Risk Management Advisory Committee May 18-19, Overview of Drug Safety Challenges Gerald J. Dal Pan, MD, MHS Director Division of.
DIVISION OF REPRODUCTIVE AND UROLOGIC PRODUCTS Physician Labeling Rule Lisa Soule, M.D.
Causal relationships, bias, and research designs Professor Anthony DiGirolamo.
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.
FDA Risk Management Workshop – Day #3 April 11, 2003 Robert C. Nelson, Ph.D. RCN Associates, Inc Annapolis, MD, USA.
1 Operation of the Prescription Drug User Fee Program Janet Woodcock, M.D. Deputy Commissioner for Operations November 14, 2005.
FDA Science Board CDER Drug Safety Update November 6, 2005 Rockville, MD Steven Galson, MD, MPH Director, Center for Drug Evaluation and Research Food.
Pharmacogenetics.
1 Presentation of the Pharmaceutical Research and Manufacturers of America at FDA Part 15 Hearing on Communication of Drug Safety Information December.
UNIT-II CLINICAL DATA. UNIT-II CLINICAL DATA: Clinical Data, Application, Challenges, Solutions, Clinical Data Management System.
European Patients’ Academy on Therapeutic Innovation Introduction to pharmacovigilance Monitoring the safety of medicines.
Questions to Committee about Potential Cancer Risk with Use of Topical Immunosuppressants (Calcineurin Inhibitors) Question 1: Messages about Risk A. Based.
1 BETTER APPROACHES TO MONITORING MEDICAL PRODUCT SAFETY ARE NEEDED.
Developing, Pilot Testing & Evaluating RiskMAP Interventions Annette Stemhagen, DrPH, FISPE Vice President UBC Epidemiology & Risk Management The FDA Regulatory.
27 June 2000Victor F. C. Raczkowski, M.D.1 Risk-Management Options Victor F. C. Raczkowski, M.D., M.S. Gastrointestinal Drugs Advisory Committee 27 June.
Section I General principle of Pharmacology. Where can you get information about general principle of Pharmacology?  Text books:  Katzung, Basic and.
Drug Utilization Review & Drug Utilization Evaluation: An Overview
Clinical Trials.
FDA’s Role in the Risk Management of Opiate Analgesics Steven Galson, M.D., M.P.H. Deputy Center Director, Center for Drug Evaluation and Research Food.
Drug Development Process Stages involved in Regulating Drugs
Patient Focused Drug Development An FDA Perspective
Reasonable Assurance of Safety and Effectiveness: An FDA Division of Cardiovascular Devices Perspective Bram Zuckerman, MD, FACC Director, FDA Division.
Clinical Trials in STS Shreyaskumar Patel, M.D.
Pharmacovigilance (PV)
Pharmacogenomics Genes and Drugs.
Patient Involvement in the Development and Safe Use of
Presentation transcript:

Applying New Science to Drug Safety Janet Woodcock, M.D. Acting Deputy Commissioner for Operations April 15, 2005

Current Drug Safety System n n Extensive premarket testing with rigorous FDA review, including evaluating of remaining uncertainties n n (Proposed) user-friendly communication via drug label, compatible with e-prescribing & electronic decision support n n Proactive risk management strategies prior to and post approval n n Voluntary adverse event reporting system with additional population-based information

Capacities of Current System: n n Generate profile of common adverse events in tested populations during drug development n n Understand drug metabolism and common metabolism-based drug-drug interactions n n Develop plans for managing/evaluating certain anticipated risks after marketing n n Identify rare serious adverse events after marketing

What Current System May Not Elucidate: n n Increases in frequency of events that occur commonly-to-uncommonly in the population without the drug n n Time dependent events n n Events occurring more frequently in populations not tested in trials: the very sick, those on polypharmacy, multiple medical problems, etc. n n Events that occur much more frequently with off- label use – –Tendency toward medical errors or abuse

Approaches to Resolution of the Knowledge Gaps n n Utilization of emerging electronic medical record systems for surveillance n n Randomized trials or registries conducted in practice settings after marketing n n More surveillance systems in specialized settings: e.g., ER, nursing homes, etc.

These Approaches Should Be Implemented But They are Not Sufficient n n Traditional focus on detection, communications, (warnings, precautions), management n n Need to add where possible: prediction; prevention; monitoring; mitigation n n Avoid treatment of individuals at high risk for event: serious side effects occur in only a small fraction of patients n n Develop new ways of monitoring for emerging toxicity before it becomes severe

Imperatives for Change n n Drug development (e.g., animal & human testing) is largely empirical in nature n n This tradition focuses on population means & observations of outliers n n Directly translated into “trial and error” approach in clinical medicine n n Major loss of information

Imperatives for Change n n There are significant limitations on the number of questions that can be answered via empirical testing (imposed by # of patients, changing practice patterns, cost, etc) n n Despite hundreds of millions of dollars invested in a development program – we often lack key information at approval. We know that the drug is effective in a defined population, and can describe the common side effects n n Many of the patients subsequently exposed will not benefit from the Rx

Imperatives for Change n n Success rate for Phase I entries below 15% n n No other major part of the technological infrastructure (e.g. civil engineering, aeronautics, electrical engineering, electronics, etc) operates on nearly this level of risk n n The same evaluative tools that will improve predictability will also support markedly improved decision making in clinical medicine n n Much of this information can be developed now

Example: Drug Metabolism n n Drugs that are CYP450 2D6 substitutes may have up to 10 fold higher blood levels from “normal” dosing in “poor metabolizers” n n Distribution of alleles varies with ethnicity n n These drugs frequently implicated in “unavoidable” side effects n n Role of variable exposure not well elucidated n n FDA recently approved a commercial test to determine an individual’s 2D6 status

Drug Metabolism in Drug Development n n Development of in vitro human cell models and animal models over last 15 years have enabled manufacturers to predict human metabolism n n Avoid candidates with problematic metabolism/drug- drug interactions n n These have dropped in same timeframe from leading cause of late clinical failures to minor cause n n Critical path focused on development of additional tools

Example: New Technologies n n Genomic, proteomic, metabolomic markers n n Status in patients with serious side effects vs those without? n n Study in prospective trials and from MedWatch reports n n Develop ability to avoid high risk patients or monitor for development before overt toxicity occurs

Development of Targeted Therapeutics n n Identify subgroups of patients with high response rates n n Avoid treatment of people with low probability of benefit n n Can remarkably decrease required power of trials in drug development n n Can significantly improve benefit/risk assessment

Future of Drug Safety n n Continue to improve current system but also– n n Deliver right dose to right patient and – n n Monitor for emergence of side effects n n Prevent serious side effects

Issues n n How to get this applied science done? n n No entity charged with task n n Public-private partnerships may be ideal n n Need venues to pool anonymized proprietary data n n Need ability to conduct studies or add-on modules to existing trials