Beyond Discovery: The Critical Role of Bioengineering Innovation in Enhancing Productivity and Value - PowerPoint PPT Presentation

1 / 42
About This Presentation
Title:

Beyond Discovery: The Critical Role of Bioengineering Innovation in Enhancing Productivity and Value

Description:

Beyond Discovery: The Critical Role of Bioengineering Innovation in Enhancing ... Janet Woodcock, M.D. Deputy Commissioner. for Operations, FDA ... – PowerPoint PPT presentation

Number of Views:55
Avg rating:3.0/5.0
Slides: 43
Provided by: cde489
Category:

less

Transcript and Presenter's Notes

Title: Beyond Discovery: The Critical Role of Bioengineering Innovation in Enhancing Productivity and Value


1
Beyond Discovery The Critical Role of
Bioengineering Innovation in Enhancing
Productivity and Value in the Biomedical Sector
  • Janet Woodcock, M.D.
  • Deputy Commissioner
  • for Operations, FDA

2
This is a Golden Age for Biomedical Discovery
  • Sequencing of human genome reveals new candidate
    targets
  • Combinatorial chemistry, high throughput
    screening, biosynthesis provide thousands of
    candidate drugs
  • Electronics innovations, nanotechnology,
    materials science drive device innovation
  • Transgenic animals, new technologies (e.g., RNAi)
    for evaluating activity

3
Problem 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

4
Ten Year Investment in U.S. Biomedical Research
  • Increased from 37B in 1994 to 94B in 2003
    (doubling when inflation-adjusted)
  • 57 of funding from industrial sector
  • 33 of funding from government (28 NIH)
  • 10 private sources

5
(No Transcript)
6
  • Matching Acceleration of Product Development Has
    Been Expected

7
(No Transcript)
8
Ten Year Trends Worldwide
  • 2004 marked a 20-year low in introduction of new
    medical therapies into worldwide markets
  • DiMasi, et al. (2003) estimated that the
    capitalized cost for self-originated NMEs
    developed by multinational pharma approved in
    2001 would be about 1.1 B per NME.
  • Disincentive for investment in less common
    diseases or risky, innovative approaches

9
Higher Development Costs
10
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.

11
Problem with Pipeline?
  • Multi-Factorial Cause
  • 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

12
Beyond Discovery Root Cause of Problem?
  • Science used to predict and evaluate product
    performance has not advanced at the same pace as
    basic science
  • Continuing to use the tools and methods of 19th
    and 20th century to evaluate 21st century
    technology development is now the bottleneck
  • Huge opportunity to improve product development
    with new science
  • Requires major paradigm shifts

13
The Critical Path for Medical Product Development
Is Now the Bottleneck
14
Evaluative Science Underlying The Critical Path
of Drug Development
Science to predict and evaluate safety efficacy
performance of new products, and enable
manufacture, is different from basic discovery
science
15
"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?

16
Working in Three Dimensions on the Critical Path
17
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.

18
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

19
Policy Implications of Critical Path Initiative
  • Statutory requirements for product approval do
    not specify assessment technologies
  • Regulations and guidance describe evaluative
    tools, e.g., tests in animals or humans
  • Although Critical Path is a science initiative,
    advances in evaluative science will result in
    regulatory changes
  • FDA will change policies and regulation as
    science progresses

20
Role of Biomedical Engineering
  • Medical product development, and consequently,
    medical care, must move from a cottage
    industry, artisan-based model to a more
    rigorous, science and engineering based set of
    processes and procedures need a bioindustrial
    revolution
  • New science and technology will provide the
    tools cross-disciplinary teams must devise new
    processes

21
Role of Biomedical Engineering
  • Biomedical engineers can span the requisite
    disciplines
  • Relate to the biological scientists
  • Understand the underlying physical science
  • Understand utility and application of
    quantitative models
  • Effectively utilize bioinformatics
  • Apply process design to product development
  • Bring a conceptual model of successive
    approximation rather than pass/fail

22
Beyond Discovery Current Post-Discovery
Development
  • Product development (e.g., animal human
    testing) is currently largely empirical in nature
  • This approach focuses on population means
    observations of outliers, is observational and
    probabilistic rather than explanatory or
    mechanistic
  • Directly translated into trial and error
    approach in clinical medicine
  • Major loss of information and ability to improve
    outcomes

23
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

24
Major Areas of Opportunity
  • New mechanistically-linked biomarkers
  • Qualifying for various uses
  • Development of surrogate EPs
  • Incorporation of bioinformatics
  • New clinical trial designs
  • Improved manufacturing technologies and
    regulatory oversight

25
New Biomarkers Example
  • Pharmacogenomic markers
  • Drug metabolism polymorphisms avoiding serious
    side effectsfirst tests have been approved
  • Predictors of drug response or nonresponse
    (narrow population)
  • Genetic basis of adverse eventsavoid treating
    those at riskprevention is preferable to
    warnings

26
New Biomarkers
  • Advanced Imaging Technologies
  • Distinguish disease subgroups for therapy
  • Rapidly evaluate response to treatment
  • Use as response measure in clinical trials

27
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

28
Manufacturing
  • Large pharmaceutical firms spend more on
    manufacturing than on RD
  • Device problems with product consistency
  • Major opportunities for cost reduction with
    increased quality (defined as less variability)
  • Requires application of modern manufacturing
    quality science to pharmaceutical sector
  • FDAs Product Quality for the 21st Century
    Initiative

29
Worked Example Animal Toxicology Testing
  • Tried and true method, fairly predictive for
    initial starting dose
  • Thousands of animal protocols run yearly with
    candidate drugs and devices strictly empirical
  • Often correlation with clinical toxicity
    available
  • No systematic generalization of results little
    improvement of testing protocol

30
Animal Toxicology Testing
  • Opportunity to evaluate new predictive toxicology
    (genomic, proteomic) within existing testing
    protocols
  • Imaging techniques may provide key distribution
    data
  • Move towards mechanistic understanding computer
    models of toxicity
  • Requires collaboration across sectors and
    companies

31
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

32
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

33
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

34
Limitations of Controlled Trials as Currently
Conducted
  • Binary outcomesuccess or failure--determined by
    p valuelimits information gain and often results
    in misinterpretation of data (e.g., estrogen
    trials)
  • Large time expenditureand may find out at the
    end that the wrong question was being asked
  • Little flexibility

35
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

36
Example Treatment Comparisons Controversy
  • Need for comparative trials for drug
    interventions safety, effectiveness,
    tolerability etc.
  • This is a standard approach, e.g., oncology,
    chronic diseases
  • Large, expensive, empirical studies that may
    loose relevance as practice evolves
  • Difficult to study more than a few interventions
    at once Congress calling for more comparative
    trials

37
Example Treatment Comparisons
  • A more fundamental problem is the following
  • superiority on a population basis may not
    reflect best choice for any given individual.
  • A treatment with a 10 advantage over a
    comparator may still be the wrong drug for a lot
    of people
  • Need better ways to distinguish response subsets
    and at-risk groups pharmacogenomics,
    proteomics, etc

38
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

39
The Development Process Needs More Engineering
Presence
  • Despite, and perhaps because of, the incredible
    complexity of biology, we must move to
    quantitative models of physiology and disease
  • We cannot be deterred by the knowledge gaps,
    approximations are better than guessing
  • We must incorporate new processes and evaluation
    technologies

40
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

41
Payoff for Patients
  • Larger treatment effects via more targeted
    therapy
  • Avoidance of side effects and injury through
    prevention
  • Better/earlier product availability
  • Higher quality healthcare

42
Strategies for Improving Health Care
Automation New Surveillance Mechanisms
Registries Outcomes Research Education and
Guidelines
Improve Evaluative Science
Better Health Outcomes
Biomedical Engineering Innovation
Medical Product Development Environment
Healthcare Environment
Write a Comment
User Comments (0)
About PowerShow.com