FDAs Critical Path Initiative - PowerPoint PPT Presentation

1 / 31
About This Presentation
Title:

FDAs Critical Path Initiative

Description:

FDA's Critical Path Initiative. Janet Woodcock, M.D. Deputy Commissioner. for Operations, FDA ... Lack of corresponding new products available to patients ... – PowerPoint PPT presentation

Number of Views:59
Avg rating:3.0/5.0
Slides: 32
Provided by: cde498
Category:

less

Transcript and Presenter's Notes

Title: FDAs Critical Path Initiative


1
FDAs Critical Path Initiative
  • Janet Woodcock, M.D.
  • Deputy Commissioner
  • for Operations, FDA
  • May 11, 2006

2
Biomedical Discoveries are Not Effectively
Translated
  • Huge Investment in U.S. Biomedical Research
  • Lack of corresponding new products available to
    patients
  • Major increases in medical product development
    costs
  • Major rise in healthcare costs

3
Predictability Problem
  • Product development success rate has declined
  • New compounds entering Phase I development today
    have 8 chance of reaching market, vs. 14 chance
    15 years ago.
  • Phase III failure rate now reported to be 50,
    vs. 20 in Phase III, 10 years ago.

4
Speculation on Causes of Translational Problems
  • Genomics other new science not at full
    potential (10-15 yrs)
  • Easy targets taken chronic disease harder to
    study
  • Rapidly escalating costs complexity decrease
    willingness and ability to bring many candidates
    forward into the clinic
  • Mergers and other business arrangements
  • Some people blame FDA

5
Whats the Diagnosis?
  • Investment and progress in basic biomedical
    science has far surpassed investment and progress
    in the medical product development process
  • The development process the critical path to
    patients becoming a serious bottleneck to
    delivery of new products
  • We are using the evaluation tools and
    infrastructure of the last century to develop
    this centurys advances

6
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
  • FDAs Critical Path Initiative focuses on the
    science used for these evaluations

7
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
8
"Critical PathDimensions
  • 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?

9
Guiding Principles of FDA Initiative
  • Collaborative efforts among government, academia,
    industry and patient groups
  • Infrastructure and toolkit development, not
    product development
  • Build support for academic science bases in
    relevant disciplines
  • Build opportunities to share existing knowledge
    database
  • Develop enabling standards

10
Steps to Date
  • Published Initial Report 5/04
  • Opened Docket for public comment and discussed
    with FDA Science Board and other Advisory
    Committees
  • Initiating multiple public-private partnership
    consortia with non-profit conveners
  • Publication of Critical Path Opportunities
    Report and List in March 06 project report
    soon
  • Organizational structures being established at
    FDA CDER Office of Translational Science
    Office of Commissioner

11
Major Opportunities for Modernization per March
Report
  • Biomarker Qualification
  • In-vitro diagnostics
  • Imaging
  • Preclinical toxicogenomics
  • Clinical Trial Modernization
  • Bioinformatics
  • Modernizing Manufacturing
  • Pediatric Treatments
  • Public Health Emergencies

12
Biomarker Qualification
  • Qualification of a biomarker means developing
    the correlative information that lets us
    understand its clinical meaning in a given
    situation
  • Examples
  • Drug metabolizing enzyme genotypes
  • Metabolic imaging of tumors
  • Acute risk of cardiac event serum assays

13
Biomarker Qualification
  • Amount of correlative information needed depends
    on use of biomarker
  • In drug development (examples)
  • Candidate activity
  • Dose-response information
  • Selection for trial enrollment
  • Surrogate endpoint

14
Biomarker Qualification Two Major Opportunities
  • Develop conceptual framework
  • Reach general consensus on amount/type of data
    needed for various uses
  • Publish FDA guidance
  • Develop consortia for qualification of specific
    biomarkers

15
Biomarker Qualification Framework Progress
  • Multiple FDA and Advisory Committee Discussions
  • Deliberations by academia and industry
  • Center for Drugs Endpoint Survey
  • Research collaborations with private sector to
    study process in specific biomarker qualification
    efforts

16
Development of Consortia for Biomarker
Qualification
  • OBQI (Oncology Biomarker Qualification
    Initiative) Announced in January
  • FDA-NCI-CMS consortium to qualify new cancer
    biomarkers
  • First project involves FDG-PET in non-Hodgkins
    Lymphoma
  • Other major consortia in development

17
Why Public-Private Biomarker Consortia?
  • Successful biomarker qualification is quite
    uncommon
  • New biomarkers are critical to clinical medicine
    and efficient product development
  • No single entity charged with accomplishing
    qualification
  • All parties (government, industry, insurers,
    academia, patients) have a big stake however

18
New Biomarkers Example
  • Pharmacogenomic markers
  • Drug metabolism polymorphisms avoiding serious
    side effectsfirst tests have been approved
    (project with C-Path and University of Utah
    proposed)
  • Predictors of drug response or nonresponse
    (narrow population)consortia being explored
  • Genetic basis of adverse eventsavoid treating
    those at riskprevention is preferable to
    warningsconsortium under construction

19
New Biomarkers
  • Advanced Imaging Technologies
  • Distinguish disease subgroups for therapy
  • Rapidly evaluate response to treatment
  • Use as response measure in clinical trials
  • OBQI exploring additional imaging biomarker
    qualification

20
Modernizing Clinical Trials Opportunities
  • Move to automated environment
  • Develop new methodological approaches to
    evaluation
  • Move towards greater mechanistic understanding,
    incorporating biomarkers

21
Modernizing Clinical Trials Automation
  • E-clinical trials initiative trial conduct and
    regulatory submission
  • Clinical trial networks (Ca-BIG)
  • FDA e-submission standards (ICH)
  • NIH roadmap projects on disease area terminology
  • Continue development of standards
  • CDISC--trial standards organizationto lead on--
  • Case Report Form standards
  • BiMo Initiative (FDA project)
  • Modernize FDA oversight of clinical trials and
    IRBs
  • Will require industry cooperation as per Product
    Quality for the 21st Century

22
Modernizing Clinical Trials Move Away from Trial
and Error Evaluation
  • Employ rigorous, informative assessments in
    preclinical and early clinical studies build
    generalized knowledge from results
  • Will require new processes and pathways
  • Will require development and regulatory
    acceptance of new evaluative tools
  • Final trials would be confirmatory-however,
    confirmatory trial process also needs to be
    redesigned

23
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
  • Basis for many of the advances of modern medicine

24
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

25
Limitations of Controlled Trials
  • Fact at the end of most drug development
    programs, after huge expenditures of time and
    resources, we dont know a great deal about the
    drug
  • Were 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

26
Healthcare Consequences of Current Development
  • Health care cost controversy Debates about value
    of products we cant quantify
  • Health care policy community believes that
    increased technologygreater 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

27
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
    questionsi.e, what dose is correct for which
    group

28
New Trial Designs for Personalized Therapy
  • Hope that industry, FDA and NIH can collaborate
    on new types of trials through public-private
    partnership arrangements
  • Plans for such trials, using new biomarkers,
    being explored
  • Use genetic, genomic, proteomic or imaging
    information to direct or modulate therapy

29
Bioinformatics
  • Develop publicly available quantitative disease
    models (natural history and response to
    intervention)
  • Perform modeling and simulation of trials of new
    interventions
  • Assess in silico the impact of device design
    modifications
  • Assemble databases necessary for qualification of
    biomarkers

30
Critical Path Payoff for Patients
  • More predictable development process for new
    drugs lower development costs more choices
  • More and better information about risks and
    benefits
  • Personalized treatments who should use the
    product and how, who should not use it!
  • More positive outcomes of therapy, avoidance of
    side effects

31
Summary
  • Critical Path Initiative moving into
    implementation phase
  • Structures being set up within agency and as
    consortia
  • Pace is defined by (limited) available resources
  • Large payoffs for patients if critical path
    initiative is successful
Write a Comment
User Comments (0)
About PowerShow.com