FDA Risk Management Workshop – Day #3 April 11, 2003 Robert C. Nelson, Ph.D. RCN Associates, Inc Annapolis, MD, USA.

Slides:



Advertisements
Similar presentations
Overview of Risk management: A EU perspective Lincoln Tsang May 2008.
Advertisements

Agency for Healthcare Research and Quality (AHRQ)
Postgraduate Course 7. Evidence-based management: Research designs.
What is Epidemiology? (1)
Safety and Extrapolation Steven Hirschfeld, MD PhD Office of Cellular, Tissue and Gene Therapy Center for Biologics Evaluation and Research FDA.
Study Designs in Epidemiologic
© Safeguarding public health Innovation; issues for regulators, society and industry. Overview of Conference topics Alasdair Breckenridge Medicines and.
1 Proposed Pharmacovigilance Plan for H5N1 Influenza Virus Vaccine Patrick Caubel, MD, PhD Head of Pharmacovigilance North America February 27, 2007.
Reading the Dental Literature
Accutane-Psychiatric Disorders A Pharmacoepidemiological Safety Assessment Robert C. Nelson, PhD RCN Associates, Inc. Annapolis, MD.
Elements of a clinical trial research protocol
1 BMS Confidential – for internal purposes only Pharmacovigilance and Drug Safety Track AN INDUSTRY PERSPECTIVE Amrit Ray, MD, MBA, FCMI Vice President,
Assessing Disease Frequency
Pharmacoepidemiology: Goals and Methods Sean Hennessy, PharmD, PhD Assistant Professor of Epidemiology & Pharmacology Center for Clinical Epidemiology.
Safety and Vigilance (SAV)
Guidance for Industry Establishing Pregnancy Registries Pregnancy Registry Working Group Pregnancy Labeling Taskforce March, 2000 Evelyn M. Rodriguez M.D.,
Cohort Studies Hanna E. Bloomfield, MD, MPH Professor of Medicine Associate Chief of Staff, Research Minneapolis VA Medical Center.
Postmarketing Risk Assessment of Drug Products Division of Drug Risk Evaluation Office of Drug Safety Center for Drug Evaluation and Research.
Patient Safety Initiatives Kelly Cronin Director, Patient Safety and Outcomes Research Office of Policy and Planning Office of the Commissioner FDA.
David A H Whiteman MD FAAP FACMG Global Clinical Sciences Leader Shire Pharmaceuticals.
IPhVWP Polish Presidency, Warsaw October 6 th 2011 Almath Spooner Irish Medicines Board Monitoring the outcome of risk minimisation activities.
Safety data collected during clinical trials is incorporated into the product’s approved label. Regulatory reviewers monitor products’ safety profiles.
Accutane FDA (Section Name - Saver’s Initials) 3/9/00 12:50 PM 1 Introduction Russell Ellison, MD Chief Medical Officer Vice President, Medical Affairs.
Multiple Choice Questions for discussion
Copyright © 2012 Wolters Kluwer Health | Lippincott Williams & Wilkins Chapter 7: Gathering Evidence for Practice.
Research Techniques Made Simple: Databases for Clinical Research Katrina Abuabara, MD MA David Margolis, MD PhD University of Pennsylvania.
Clinical causality assessment I. Ralph Edwards R.H.B Meyboom.
Study Design. Study Designs Descriptive Studies Record events, observations or activities,documentaries No comparison group or intervention Describe.
Epidemiology The Basics Only… Adapted with permission from a class presentation developed by Dr. Charles Lynch – University of Iowa, Iowa City.
CHP400: Community Health Program- lI Research Methodology STUDY DESIGNS Observational / Analytical Studies Case Control Studies Present: Disease Past:
Preventing Surgical Complications Prevent Harm from High Alert Medication- Anticoagulants in Primary Care Insert Date here Presenter:
PHARMACOVIGILANCE AND CLINICAL TRIALS DIVISION 20 August 2015 Victoria Falls Protecting Your Right to Quality Medicines and Medical Devices.
Postmarketing Safety Assessment of Osteonecrosis of the Jaw Pamidronate & Zoledronic Acid Division of Drug Risk Evaluation Office of Drug Safety FDA Carol.
ECON ECON Health Economic Policy Lab Kem P. Krueger, Pharm.D., Ph.D. Anne Alexander, M.S., Ph.D. University of Wyoming.
Reproductive Health Drugs, Pregnancy Labeling Subcommittee Meeting March 28-29, 2000 Holli A. Hamilton, M.D., M.P.H. Pregnancy Labeling Team Office of.
Drug Submissions: Review Process Agnes V. Klein, MD Biologics and Genetic Therapies Directorate February, 2003 www/hc-sc.gc.ca/hpb-dgps/therapeut.
Julio A. Ramirez, MD, FACP Professor of Medicine Chief, Infectious Diseases Division, University of Louisville Chief, Infectious Diseases Section, Veterans.
Study Designs in Epidemiologic
Consideration for Stakeholders Regarding Hazard Identification and Risk Assessment as Part of the MSD Prevention Strategy for Ontario Richard Wells University.
Concept Paper #3: Risk Assessment of Observational Data Session 1: Good Pharmacovigilance Practices Linda S. Hostelley on behalf of PhRMA.
DEVELOPING EVIDENCE ON VACCINE SAFETY Susan S. Ellenberg, Ph.D. Center for Clinical Epidemiology and Biostatistics U Penn School of Medicine Global Vaccines.
METHODS TO STUDY DRUG SAFETY PROBLEMS animal experiments clinical trials epidemiological methods –spontaneous reporting case reports case series –Post-Marketing.
Technical Briefing Seminar September |1 | Methods to study medicine safety problems Mary R Couper Quality Assurance and Safety of Medicines.
Drug Safety and Risk Management Advisory Committee May 18-19, Overview of Drug Safety Challenges Gerald J. Dal Pan, MD, MHS Director Division of.
CAUSALITY ASSESSMENT OF SUSPECTED AEs Dr. Retesh Kumar Head, Global PhV Department 12/13/2015.
Good Pharmacovigilance Practices
Introduction.
Sifting through the evidence Sarah Fradsham. Types of Evidence Primary Literature Observational studies Case Report Case Series Case Control Study Cohort.
FDA1 Overview of Postmarketing Safety Surveillance in FDA (For Drugs and Biologics) Min Chen, M.S., R.Ph. Min Chen, M.S., R.Ph. Associate Director Division.
Signal identification and development I.Ralph Edwards.
Pharmacovigilance WHO definition
Design of Clinical Research Studies ASAP Session by: Robert McCarter, ScD Dir. Biostatistics and Informatics, CNMC
Minimum requirements for Pharmacovigilance in countries.
Postmarketing Pharmacovigilance Practice at FDA Lanh Green, Pharm.D., M.P.H. Office of Surveillance and Epidemiology June 21, 2006.
European Patients’ Academy on Therapeutic Innovation Introduction to pharmacovigilance Monitoring the safety of medicines.
Types of Studies. Aim of epidemiological studies To determine distribution of disease To examine determinants of a disease To judge whether a given exposure.
Applying New Science to Drug Safety Janet Woodcock, M.D. Acting Deputy Commissioner for Operations April 15, 2005.
Active Surveillance Using Longitudinal Data: A Pilot Project Drug Safety and Risk Management Advisory Committee Meeting Silver Spring, Maryland May 18,
Comments on FDA Concept Paper Sidney N. Kahn, MD, PhD President Pharmacovigilance & Risk Management, Inc. Risk Assessment of Observational.
1 BETTER APPROACHES TO MONITORING MEDICAL PRODUCT SAFETY ARE NEEDED.
Postmarketing Pharmacovigilance English D. Willis, MD Clinical Risk Management Merck Research Laboratories June 1, 2012.
Developing, Pilot Testing & Evaluating RiskMAP Interventions Annette Stemhagen, DrPH, FISPE Vice President UBC Epidemiology & Risk Management The FDA Regulatory.
Detection & monitoring of ADR
8. Causality assessment:
Donald E. Cutlip, MD Beth Israel Deaconess Medical Center
3. Key definitions Multi-partner training package on active TB drug safety monitoring and management (aDSM) July 2016.
9. Introduction to signal detection
Developing pharmacovigilance: new challenges and opportunities Mary Couper and Shanthi Pal Quality Assurance and Safety of Medicines.
PHARMACOVIGILANCE SYSTEM
Presentation transcript:

FDA Risk Management Workshop – Day #3 April 11, 2003 Robert C. Nelson, Ph.D. RCN Associates, Inc Annapolis, MD, USA

Definitions Postmarketing Surveillance (PMS) –Monitoring of Safety, Quality, Advertising After Market Approval Pharmacovigilance (PV) –Review and Assessment of Case (Spontaneous) Reports Pharmacoepidemiology (PE) –Use of Non-experimental Methods to Assess the Therapeutic Value of Drugs in Clinical Practice Environments Risk Assessment (RA) –PV + PE + All Needed Knowledge to Make a Global Evaluation of a Specific Risk Risk Management (RM) –Strategy that Uses All of the above, and a –Toolkit of Regulatory Interventions, and –Variety of Risk Communication Devices

FDA Questions 1.How can the quality of spontaneously reported case reports be improved? 2.What are possible advantages or disadvantages of applying datamining techniques to spontaneous reporting databases for the purpose of identifying safety signals? 3.What are possible advantages or disadvantages of performing causality assessments at the individual case level? 4.Under what circumstances would a registry be useful as a surveillance tool and when would it cease to be useful? 5.Under what circumstances would active surveillance strategies prove useful to identify as yet unreported adverse events? 6.Under what circumstances would additional pharmacoepidemiologic studies be useful?

FDA Question #1 How can the quality of spontaneously reported case reports be improved Public Health Focused Philosophy: True unless shown otherwise –“Encourages” companies to obtain full data to assess alternate explanations –“Confounding” does not mean lack of causal relationship! Focus on “important’ –Active query at point of initial contact FDA compliance on quality standards > timeliness Implement new proposed regulations ASAP after comment period closes

FDA Question #2 What are possible advantages or disadvantages of applying datamining techniques to spontaneous reporting databases for the purpose of identifying safety signals? Datamining is of value for –secondary “alerting” –Identifying previously unrecognized data relationships Most valid: Internal (denominator) based proportional analyses and comparisons within pharmacological group “Alerts” then require clinical judgment to declare a “signal” requiring subsequent epidemiological analyses

FDA Question #3 What are possible advantages or disadvantages of performing causality assessments at the individual case level? Useful: Sorts high volume by quality of evidence Without some “probable” cases there is little evidence from the remaining majority of “possible” spontaneous reports All included cases => Case series assessment “Definite” case = Myth!

FDA Question #4 Under what circumstances would a registry be useful as a surveillance tool and when would it cease to be useful? Require valid comparator group(s) SADR data needs to be collected using the same methods and instruments as used in pre-approval RCTs (i.e., “Systematic collection”) Prospective cohort studies are NOT powered to find new ADRs Best in assessing risk factors for known reactions for subsequent prevention, i.e. WHY (root cause) they occur.

FDA Question #5 Under what circumstances would active surveillance strategies prove useful to identify as yet unreported adverse events? Active case-control surveillance methodology (a la Shapiro) should be re-explored MedSun strategy extended to drugs Use of Poison Control/Drug Information Centers as dedicated data acquisition sentinels –Drug-Drug interactions –Medication errors

FDA Question #6 Under what circumstances would additional pharmacoepidemiologic studies be useful? When they can be; –Replicated –Validated –Provide Quantitative data Measure risk Measure change in risk

The Power of Myth Spontaneous Reporting –Confounders –Rates –Underreporting Secondary Data sources –ICD9-CM –Misclassification

Confounding Myth Spontaneous case reports seldom contain all history and data on concomitant conditions or drugs Absence of these data may be due to: –True absence or Poor data acquisition Presence of these confounders does not “clear’ the drug Dismissing ‘confounded cases’ en masse in complex medical conditions is an error because “unconfounded’ cases may be due to poor data acquisition Apply Public Health Focused Philosophy: True unless shown otherwise Popperian Logic: relative likelihood of alternate explanations

Reporting Rate Myth The statistical assumptions in Finney (1971) no longer valid in U.S. scheme –Non-HP reporters, nonserious, solicited, etc. Sampling scheme is unknown –Qualitative not quantitative data –Cases are therefore not true numerators Extrapolations (1%,10% etc.) are voodoo science Reporting rates can provide contributory evidence for regulatory decision making only when the outcome reaction is exceedingly rare. Otherwise, reporting rates can only serve as a confirmation bridge between the cases and the observational research More valid: Internal (denominator) based proportional analyses and comparisons within pharmacological group

The Under-Reporting Myth Voluntary spontaneous reporting schemes were designed to signal new, rare, unusual serious ADRs NOT designed to collect all reactions This is a characteristic NOT a limitation or flaw! In U.S.: Over-reporting! True underreporting: missing data in medically significant cases

Secondary Data Myth Secondary (Medical record/Billing) Data sources –ICD9-CM NOT ADRs –Misclassification blunts risk estimates Therefore, cannot be used as quantitative data sources! Limited value in true risk management!