National Academies Press: OpenBook
« Previous: Front Matter
Suggested Citation:"1 Introduction." Institute of Medicine. 2011. Generating Evidence for Genomic Diagnostic Test Development: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/13133.
×

1
Introduction

The field of genomics has expanded greatly since the first sequence of the human genome was published a decade ago. According to workshop chair Debra Leonard of Weill Cornell Medical College, the hoped-for outcomes of the human genome project were to understand human genetic variations and their relationship to health and disease; to predict disease risks for prevention and earlier treatment; to refine disease diagnosis by understanding the underlying genetic variance and molecular mechanisms; and to use that information to create better treatments and improve the health and health outcomes of the U.S. population. Over the past 10 years, scientists have developed genomic tests based on identified gene-disease associations that can predict the response of an individual patient to a drug intervention or the risk of developing Alzheimer’s disease. However, much of the evidence surrounding the clinical value and utility of these tests has not been sufficient enough for clinical practitioners to broadly embrace many of these in practice.

A major impediment to the integration of these genomic tests into routine health care is the lack of an adequate evidence base linking the use of genomic diagnostic tests to health outcomes. Since these new technologies are beginning to play an increased role in clinical decision-making and the management of disease, the Institute of Medicine’s Roundtable on Translating Genomic-Based Research for Health hosted a public workshop on

Suggested Citation:"1 Introduction." Institute of Medicine. 2011. Generating Evidence for Genomic Diagnostic Test Development: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/13133.
×

November 17, 2010, to explore issues related to this lack of evidence.1 Various stakeholders, including regulators and policymakers, payers, healthcare providers, researchers, funders, and evidence-based review groups, were invited to share their perspectives on the strengths and limitations of the evidence being generated to assess the clinical validity and utility of genomic diagnostic tests. Specifically, panelists were asked to address the following:

  • What evidence is required by stakeholders (e.g., for decisions regarding clearance, use, and reimbursement)?

  • How is evidence currently being generated?

  • Are there innovative and efficient ways to generate high-quality evidence?

  • How can the barriers to generating this evidence be overcome?

Early genetic tests, Leonard explained, were focused on single genes. The market was limited and reimbursement was poor, and the in vitro diagnostics industry was therefore not very interested in developing genetic tests. Instead, genetic tests for the diagnosis of disease were generally developed as needed by clinical laboratories. These were based on published genotype–phenotype correlations and were developed using standard molecular biology methods and sets of patient and control samples. The Clinical Laboratory Improvement Amendments (CLIA) (42 U.S.C. 263a) allows such practices without the need for receiving device clearance from the U.S. Food and Drug Administration (FDA). However, Leonard said, there were concerns about the quality of these tests, the potential for harm to patients, the clinical validity and utility of the tests, and the relatively expensive cost.

Genetic tests are still in use today, Leonard said, but the focus has shifted to genomic tests, which are complex testing algorithms of multiple genetic variants, multiple genes, or gene expression patterns. Genomic tests are used for diagnosis as well as for therapeutic selection, dosing, prognosis, and residual disease detection. However, the majority of these tests have insufficient clinical validity and utility data, and there is currently little evidence of improved health outcomes from their use (Table 1-1). The increasing role of genomic tests in clinical decision-making has led to

1

This workshop was organized by an independent planning committee whose role was limited to developing the meeting agenda. This summary has been prepared by the rapporteurs as a factual summary of the discussion that took place at the workshop. All views presented in the report are those of the individual workshop participants and should not be construed as reflecting any group consensus.

Suggested Citation:"1 Introduction." Institute of Medicine. 2011. Generating Evidence for Genomic Diagnostic Test Development: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/13133.
×

TABLE 1-1 Evidence-Based Review of Select Genomic Tests

Genetic Test

Reviewed by

Conclusion

Thrombophilia tests

AHRQ/EGAPP

No direct evidence for improved outcomes

HER2 testing in breast cancer

AHRQ

Weak evidence relating test result to treatment outcomes

Gene expression profiles for breast cancer

AHRQ

High quality retrospective clinical utility data for Oncotype DX

UGT1A1 genotyping for colorectal cancer patients

EGAPP

Insufficient evidence for or against testing

Genetic testing for hereditary nonpolyposis colorectal cancer (HNPCC)

EGAPP

Limited evidence that mismatch repair gene mutations cause family members to have increased screening

CYP450 for non-psychotic depression

EGAPP

Paucity of good quality evidence that testing is useful for selective serotonin reuptake inhibitor (SSRI) outcomes

Genomic tests for ovarian cancer

EGAPP

No evidence that tests affect outcomes in asymptomatic women

Abbreviations: AHRQ (Agency for Healthcare Research and Quality), EGAPP (Evaluation of Genomic Applications in Practice and Prevention).

SOURCE: Adapted from Leonard, IOM workshop presentation on November 17, 2010.

ongoing discussions at FDA regarding the appropriate level of regulatory oversight for genetic and genomic tests.2

The barriers to evidence generation have been discussed in many venues (summarized in Box 1-1). The goal of this workshop, Leonard said, is to look beyond these barriers to define the evidence needed and the mecha-

2

As further discussed in Chapter 2, other stakeholder groups including payers, evidence-based review groups, providers, professional societies, and patient groups have also initiated discussions on the utility of genetic and genomic tests in clinical decision making.

Suggested Citation:"1 Introduction." Institute of Medicine. 2011. Generating Evidence for Genomic Diagnostic Test Development: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/13133.
×

BOX 1-1

Speaker’s Perspectives on Barriers to the Collection of Clinical Validity and Utility Data for Genomic Diagnostic Tests

  • Various stakeholders require different types and levels of evidence (e.g., doc­tors, patients, FDA, payers, evidence-based review groups)

  • Limited or nonexistent funding for randomized controlled trials of genomic tests

  • Length of time needed for clinical trials to be completed

  • High cost of archiving specimens from therapeutic clinical trials

  • Lack of access to annotated clinical specimens

nisms to obtain it so that the promise of the human genome project and genomic diagnostic testing can be fully realized.

The report that follows summarizes the presentations and discussions by the expert panelists. Chapter 2 provides the different stakeholder perspectives on the type and level of evidence needed for decision making. Approaches for evidence generation are discussed in Chapter 3. Chapters 4 and 5 examine ways to overcome the barriers to evidence generation and strategies for moving forward. Final remarks are provided in Chapter 6, and the workshop agenda, biographical sketches of the panelists, and list of attendees are included in the appendixes.

Suggested Citation:"1 Introduction." Institute of Medicine. 2011. Generating Evidence for Genomic Diagnostic Test Development: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/13133.
×
Page 1
Suggested Citation:"1 Introduction." Institute of Medicine. 2011. Generating Evidence for Genomic Diagnostic Test Development: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/13133.
×
Page 2
Suggested Citation:"1 Introduction." Institute of Medicine. 2011. Generating Evidence for Genomic Diagnostic Test Development: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/13133.
×
Page 3
Suggested Citation:"1 Introduction." Institute of Medicine. 2011. Generating Evidence for Genomic Diagnostic Test Development: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/13133.
×
Page 4
Next: 2 Stakeholder Perspectives on Evidence »
Generating Evidence for Genomic Diagnostic Test Development: Workshop Summary Get This Book
×
Buy Paperback | $29.00 Buy Ebook | $23.99
MyNAP members save 10% online.
Login or Register to save!
Download Free PDF

Ten years after the sequencing of the human genome, scientists have developed genetic tests that can predict a person's response to certain drugs, estimate the risk of developing Alzheimer's disease, and make other predictions based on known links between genes and diseases. However, genetic tests have yet to become a routine part of medical care, in part because there is not enough evidence to show they help improve patients' health.

The Institute of Medicine (IOM) held a workshop to explore how researchers can gather better evidence more efficiently on the clinical utility of genetic tests. Generating Evidence for Genomic Diagnostic Test Development compares the evidence that is required for decisions regarding clearance, use, and reimbursement, to the evidence that is currently generated. The report also addresses innovative and efficient ways to generate high-quality evidence, as well as barriers to generating this evidence.

Generating Evidence for Genomic Diagnostic Test Development contains information that will be of great value to regulators and policymakers, payers, health-care providers, researchers, funders, and evidence-based review groups.

  1. ×

    Welcome to OpenBook!

    You're looking at OpenBook, NAP.edu's online reading room since 1999. Based on feedback from you, our users, we've made some improvements that make it easier than ever to read thousands of publications on our website.

    Do you want to take a quick tour of the OpenBook's features?

    No Thanks Take a Tour »
  2. ×

    Show this book's table of contents, where you can jump to any chapter by name.

    « Back Next »
  3. ×

    ...or use these buttons to go back to the previous chapter or skip to the next one.

    « Back Next »
  4. ×

    Jump up to the previous page or down to the next one. Also, you can type in a page number and press Enter to go directly to that page in the book.

    « Back Next »
  5. ×

    Switch between the Original Pages, where you can read the report as it appeared in print, and Text Pages for the web version, where you can highlight and search the text.

    « Back Next »
  6. ×

    To search the entire text of this book, type in your search term here and press Enter.

    « Back Next »
  7. ×

    Share a link to this book page on your preferred social network or via email.

    « Back Next »
  8. ×

    View our suggested citation for this chapter.

    « Back Next »
  9. ×

    Ready to take your reading offline? Click here to buy this book in print or download it as a free PDF, if available.

    « Back Next »
Stay Connected!