Regulatory Issues in Genetic Testing and Targeted Drug Development



Similar documents
Guidance for Sponsors, Institutional Review Boards, Clinical Investigators and FDA Staff

FDA Regulation of Genetic Tests. Steven Gutman, M.D. Office of In Vitro Diagnostics

Voluntary Genomic Data Submissions at the U.S. FDA

A leader in the development and application of information technology to prevent and treat disease.

Overview of Phase 1 Oncology Trials of Biologic Therapeutics

Personalized Medicine Issues affecting adoption of personalized medicine

Guidance for Industry and FDA Staff Commercially Distributed Analyte Specific Reagents (ASRs): Frequently Asked Questions

Genomic Medicine The Future of Cancer Care. Shayma Master Kazmi, M.D. Medical Oncology/Hematology Cancer Treatment Centers of America

exactly. The need for efficiency in developing effective new therapeutics has never been greater.

The Clinical Trials Process an educated patient s guide

A Cost Effective Way to De Risk Biomarker Clinical Trials: Early Development Considerations

Is it time for a new drug development paradigm?

The Retrospective Approach To Companion Diagnostics

Achieving Regulatory Success: Areas of focus for biotechnology companies. Michael J. Schlosser, PhD, DABT April 21, 2013

Assessing the Clinical Utility of Diagnostics Used in Drug Therapy

The Evolving Role of Nurses in Early Phase Research

Pharmacology skills for drug discovery. Why is pharmacology important?

Division of Bioinformatics and Biostatistics

Not All Clinical Trials Are Created Equal Understanding the Different Phases

RESEARCH INVOLVING DATA AND/OR BIOLOGICAL SPECIMENS

Cancer Clinical Trials: In-Depth Information

Regulatory Submission: Applying GLP in Surgical Efficacy Studies

Roche Position on Human Stem Cells

Call 2014: High throughput screening of therapeutic molecules and rare diseases

Draft Guidance for Industry, Food and Drug Administration Staff, and Clinical Laboratories

Human Research Protection Program University of California, San Diego ISSUES ON DNA AND INFORMED CONSENT

Implementation of Pharmacogenomics in Clinical Practice: Barriers and Potential Solutions

Clinical Trial Designs for Incorporating Multiple Biomarkers in Combination Studies with Targeted Agents

CTC Technology Readiness Levels

Future Directions in Clinical Research. Karen Kelly, MD Associate Director for Clinical Research UC Davis Cancer Center

STATE OF MICHIGAN DEPARTMENT OF INSURANCE AND FINANCIAL SERVICES Before the Director of Insurance and Financial Services

Diagnostic Tests. Brad Spring Director, Regulatory Affairs

Guidance for Industry and FDA Staff

IMPLEMENTING BIG DATA IN TODAY S HEALTH CARE PRAXIS: A CONUNDRUM TO PATIENTS, CAREGIVERS AND OTHER STAKEHOLDERS - WHAT IS THE VALUE AND WHO PAYS

INTERNATIONAL CONFERENCE ON HARMONISATION OF TECHNICAL REQUIREMENTS FOR REGISTRATION OF PHARMACEUTICALS FOR HUMAN USE E15

CLINICAL TRIALS SHOULD YOU PARTICIPATE? by Gwen L. Nichols, MD

Guidance for Clinical Investigators, Sponsors, and IRBs

Accelerating Development and Approval of Targeted Cancer Therapies

Guidance for Sponsors, Investigators, and Institutional Review Boards. Questions and Answers on Informed Consent Elements, 21 CFR 50.

The Value of A Qualified Outcome Measure Janet Woodcock, M.D.

EXHIBIT COORDINATING PROVISIONS-STATE/FEDERAL LAW, ACCREDITATION STANDARDS AND GEOGRAPHIC EXCEPTIONS MARYLAND

MOBILE MEDICAL APPLICATIONS

Participating in Alzheimer s Disease Clinical Trials and Studies

Ask Us About Clinical Trials

Memorandum. Factual Background

Clinical Trial Design. Sponsored by Center for Cancer Research National Cancer Institute

Essential Nursing Competencies and Curricula Guidelines for Genetics and Genomics: Outcome Indicators

Quality by Design Concept

Subject: No. Page PROTOCOL AND CASE REPORT FORM DEVELOPMENT AND REVIEW Standard Operating Procedure

How Public-Private Partnerships Benefit All Stakeholders Douglas C. Throckmorton, MD Deputy Director for Regulatory Programs, CDER, FDA

National Cancer Institute

How Can Institutions Foster OMICS Research While Protecting Patients?

TIBCO Spotfire Helps Organon Bridge the Data Gap Between Basic Research and Clinical Trials

Genetic testing. The difference diagnostics can make. The British In Vitro Diagnostics Association

Technology funding opportunities at the National Cancer Institute

Clinical Trials: The Crux of Cancer Innovation

SMF Awareness Seminar 2014

Regulatory Pathways for Licensure and Use of Ebola Virus Vaccines During the Current Outbreak FDA Perspective

Proposed Definition for Clinical Decision Support Software

BIOTECHNOLOGY OPERATIONS

Guidance for Industry

What Lies Ahead? Trends to Watch: Health Care Product Development in North America

EMA and Progressive Multifocal Leukoencephalopathy.

Guidance for Industry

Guidance for Industry

Glossary of Clinical Trial Terms

TERM SHEET EXAMPLE. 1 P age

Payers those financially responsible for the cost of

Mobile Medical Application Development: FDA Regulation

Personalized Predictive Medicine and Genomic Clinical Trials

The MSCR Curriculum and Its Advantages

FDA Regulation of Whole Slide Imaging (WSI) Devices: Current Thoughts

US Perspective on the Regulatory Assessment of Benefit-Risk of Vaccines

Course Course Name # Summer Courses DCS Clinical Research 5103 Questions & Methods CORE. Credit Hours. Course Description

Roche Diagnostics Driving Personalized Healthcare

Clinical Trials: Questions and Answers

Revision of the Directive 98/79/EC on In Vitro Diagnostic Medical Devices. Response from Cancer Research UK to the Commission August 2010

Kentucky Lung Cancer Research Program Strategic Plan Update

SMALL BUSINESS INNOVATION RESEARCH (SBIR) SMALL BUSINESS TECHNOLOGY TRANSFER (STTR) GRANT APPLICATIONS

The Promise and Challenge of Adaptive Design in Oncology Trials

Integrating Bioinformatics, Medical Sciences and Drug Discovery

Factors Influencing LC/MS/MS Moving into Clinical and Research Laboratories

LCFA/IASLC LORI MONROE SCHOLARSHIP IN TRANSLATIONAL LUNG CANCER RESEARCH

Principles for Responsible Clinical Trial Data Sharing

Guidance for Industry

Use of Electronic Health Record Data in Clinical Investigations

How To Know If A Mobile App Is A Medical Device

Guidance on Withdrawal of Subjects from Research: Data Retention and Other Related Issues

> Semantic Web Use Cases and Case Studies

Non-clinical development of biologics

Expanded Access Programs. Richard Klein Office of Special Health issues Food and Drug Administration

TECHNOLOGIES, PRODUCTS & SERVICES for MOLECULAR DIAGNOSTICS, MDx ABA 298

Patient Handbook on Stem Cell Therapies

Pfizer Oncology. Guest Expert: Louis Denis, MD Director of Clinical Research, Pfizer Oncology.

Guidance for Industry FDA Approval of New Cancer Treatment Uses for Marketed Drug and Biological Products

Clinical Trials: Improving the Care of People Living With Cancer

Challenges in the Regulation of Pediatric Clinical Trials

Transcription:

Regulatory Issues in Genetic Testing and Targeted Drug Development Janet Woodcock, M.D. Deputy Commissioner for Operations Food and Drug Administration October 12, 2006

Genetic and Genomic Tests are Types of Biomarkers Biomarker: a measurable characteristic that reflects physiologic or pathologic processes or states, or responses to pharmacologic interventions Genetic or genomic biomarkers: characteristics of an individual s genes or gene expression

Biomarker Measurement is the Foundation of Medicine Early biomarkers were physical signs and symptoms that could be measured by observation Later biomarkers (e.g., cholesterol) could provide rough prognostic information for the individual or diagnose serious organ malfunction (e.g., ECG, serum creatinine) A new generation of biomarkers that can transform diagnosis is under development: these include genetic-based assays but the universe is much broader

Challenges in Biomarker Development Many candidate biomarkers are discovered Few are evaluated rigorously for their clinical implications i.e., what they really mean in terms of human health A number enter clinical use but fewer receive regulatory acceptance (e.g., for use in drug development) and reimbursement is uneven

Reasons for Lagging Development of Clinical Biomarkers Lack of good business models for diagnostics Difficulty in establishing clinical correlation: what is needed? Clinical community focus on intervention vs diagnosis e.g., reimbursement practices Until recently: not so many good candidates

FDA's Critical Path Initiative A serious attempt to bring attention and focus to the need for targeted scientific efforts to modernize the processes and methods used to evaluate the safety, efficacy and quality of medical products as they move from product selection and design to mass manufacture.

What is the Critical Path? There is a critical path stretching from candidate identification to commercial product Involves serial evaluation of product performance through preclinical testing and clinical evaluation The science underlying the critical path has lagged behind the progress of other biomedical science FDA s Critical Path Initiative focuses on improving the science used for these evaluations

The Critical Path for Medical Product Development Is Now the Bottleneck

Evaluative Science Underlying The Critical Path Science to predict and evaluate safety & efficacy performance of new products, and enable manufacture, is different from basic discovery science

"Critical Path Dimensions Evaluative science to address 3 key product performance dimensions: Assessment of Safety how to predict and asses the risks of a potential product? Proof of Efficacy -- how to predict and demonstrate that a potential product will have medical benefit? Industrialization how to manufacture a product at commercial scale with consistently high quality?

Steps to Date Published Initial Report: Innovation or Stagnation, 5/04 Opened Docket; numerous public discussions about drug, device and biologic development challenges Initiating multiple public-private partnership consortia with non-profit conveners Published Critical Path Opportunities Report and List 3/06

Major Opportunities for Modernization per Critical Path Opportunities List Biomarker Qualification In-vitro diagnostics Imaging Preclinical toxicogenomics Clinical Trial Modernization Bioinformatics Modernizing Manufacturing Pediatric Treatments Public Health Emergencies

Genetic Tests and FDA s Critical Path Initiative Major opportunity: Utilize new biomarkers to modernize product development Genetic/genomic markers prominent Proposal: Qualify biomarkers for regulatory use via public-private partnership consortia to overcome the identified obstacles

Progress in Biomarker Qualification Predictive Safety Consortium: Sponsored by C-Path Institute, Tucson, AZ. Developing safety biomarkers for organ toxicity using animal models hope to move into human evaluations after animal qualification C-Path also evaluating pharmacogeneticdirected dosing of warfarin Oncology Biomarker Qualification Initiative (FDA/NCI/CMS): Qualifying imaging, genetic, genomic and other biomarkers for use in cancer treatment

The Biomarker Consortium Announced last week Convened by FNIH: NIH/FDA/CMS; PhRMA/BIO; Nonprofit patient groups Goal: Development and Qualification of new human biomarkers Announced projects include imaging; serum protein and genetic biomarkers

Genetic Biomarkers and the FDA: Current Issues FDA regulation of genetic and genomic tests Use of genetic and genomic testing in therapeutic product development

FDA Regulation of Genetic and Genomic Tests New FDA Guidances: Testing of leftover specimens Regulation of home brew and ASRs New draft guidance on IVDMIA s

New FDA Guidance Documents Guidance on Informed Consent for In Vitro Diagnostic Device Studies Using Leftover Human Specimens that are Not Individually Identifiable http://www.fda.gov/cdrh/oivd/guidance/1588.pdf Commercially Distributed Analyte Specific Reagents (ASRs): Frequently Asked Questions (Draft) http://www.fda.gov/cdrh/oivd/guidance/1590.pdf In Vitro Diagnostic Multivariate Index Assays (Draft)

Leftover Specimens FDA law and regulations make patient samples subject to human subject protections and requirement for informed consent Differs from NIH Common Rule Means unidentified left-over discard samples and specimens cannot be used for development or research on new tests for FDA submissions

Leftover Specimens FDA intends to exercise enforcement discretion, under certain circumstances, with respect to requiring informed consent when human specimens are used in FDAregulated IVD Device investigations Guidance is first step regulation to follow

Leftover Specimens: Enforcement Discretion Investigation meets the IDE exemption criteria at 21 CFR 812.2 (c)(3) The study uses leftover specimens Specimens may be coded but not individually identifiable Specimen may be accompanied by clinical information if source not identifiable Individuals caring for patients are different from those conducting the study Supplier of specimen has established policies and procedures to prevent release of identifiable information Study protocol must be reviewed by an IRB

ASR Draft Guidance Intended to address confusion in the marketplace and to clarify the regulations regarding commercially distributed ASRs and the role and responsibilities of ASR manufacturers Guidance was adapted from a document submitted to FDA by AdvaMed

ASR Guidance Intended to clarify the definition of an ASR Intended to communicate limitations on marketing of ASRs Not intended to eliminate legitimate home brew testing 90 day comment period

ASR Guidance Examples of ASRs: a single antibody e.g., an anti-troponin I antibody a single nucleotide primer e.g., a forward primer for amplification of the ΔF508 locus of the CFTR gene a single purified protein or peptide e.g., purified estrogen receptor protein

Multivariate Draft Guidance Identifies a class of laboratory developed devices subject to FDA regulation In Vitro Diagnostic Multivariate Index Assays: IVDMIAs

Multivariate Guidance IVDMIAs: Use clinical data (from one or more IVDs assays and sometimes demographic data) to empirically identify an algorithm Employ the algorithm to integrate these different data points in order to calculate a patient-specific result (e.g., a classification, score, or index ) The result cannot be interpreted by clinicians using prior knowledge of medicine without information from the test developer regarding its clinical performance and effectiveness

Multivariate Guidance IVDMIAs are a narrow niche of laboratory developed devices that are not subject to enforcement discretion Guidance indicates regulation will be risk-based by intended use (same as all other medical devices) 90 day comment period: draft has generated some confusion

Genetic Tests and Therapeutic Product Development Voluntary Genomic Data Submission Use in targeted therapy Drug-Diagnostic Co-development Guidance

Voluntary Genomic Data Submission Process FDA procedure to evaluate emerging genetic or genomic data relevant to investigational products outside the regulatory arena Enthusiastic participation by industry multiple types of genetic data shared (e.g., prognostic, patient stratification, adverse events etc) FDA able to develop expertise to evaluate proposals for standard drug development FDA also working on microarray standards via MAQC consortium (FDA s NCTR)

Use of Genomic Markers to Target Therapy Pharmacogenomic markers Proper dosing: Drug metabolism polymorphisms: avoiding serious side effects first tests have been approved Improving efficacy: Predictors of drug response or nonresponse (to target treated population) Improving safety: Genetic basis of adverse events avoid treating those at risk prevention is preferable to warnings

The Current Clinical Development Model The randomized controlled clinical trial represented a scientific triumph over anecdotal medicine in the 1960s Used to control for bias and the impact of random (unexplainable) variability but this variability is a the heart of personalization Basis for many of the advances of modern medicine

Limitations of Controlled Trials Theoretically can answer any and all questions via controlled experiments Can answer one or a few questions per trial There are an unlimited number of questions about the appropriate use of medical products and the outcomes of such use, and these questions evolve over time There is a decidedly limited universe of funding, patients, investigators, time and resources to conduct trials to answer these questions

Limitations of Controlled Trials Fact: at the end of most drug development programs, after huge expenditures of time and resources, we don t know a great deal about the drug We re quite confident it has a measurable beneficial effect in a described population-but the overall treatment effect is often small. Did few people respond a lot or did a lot of people respond a bit? Often many of the people who take the drug do not benefit

Limitations of Controlled Trials as Currently Conducted Binary outcome success or failure-- determined by p value limits information gain and often results in misinterpretation of data (e.g., estrogen trials) Large time expenditure and may find out at the end that the wrong question was being asked Little flexibility

Healthcare Consequences of Current Development Health care cost controversy: Debates about value of products: we can t quantify Health care policy community believes that increased technology=greater expense, and usually lower productivity Safety controversies: Products are Safe or Unsafe Health care quality: Confusing results and conflicting reports lead to anecdotal approach to care

More Informative Clinical Trial Designs Pair diagnostic(s) with therapeutic in development to identify responsive subgroup(s), or prevent toxicity Adaptive designs to answer series of questions i.e, what dose is correct for which group

Critical Path Payoff for Development Process More predictable process; higher success rate, lower development costs More information about product performance Continuous improvement of development science and processes

Critical Path: Payoff for Patients: More Personalized Treatment Much larger treatment effects via more targeted therapy Stopping ineffective therapy faster Avoidance of side effects and injury through prevention Better/earlier product availability Higher quality healthcare

Drug-Diagnostic Co-Development Guidance Need clear regulatory pathway for developing a investigational genetic or genomic test along with an investigational therapy Issues of sequencing, trial design, and statistical analysis important Also must clarify claims that would be allowed under various scenarios Much progress made although guidance not out yet

Summary FDA regulation of genetic tests remains a somewhat controversial area New draft guidances to provide greater clarity FDA is intensifying involvement in development of genetic/genomic markers Standardization of platforms for analysis is a key issue that is currently being addressed Critical Path Initiative brings intense focus on genetic/genomic biomarkers that may be used in drug development