Evolution of Translational Omics Lessons Learned and the Path Forward

Similar documents
How Can Institutions Foster OMICS Research While Protecting Patients?

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

Genomic medicine in Australia. Professor Warwick Anderson Chief Executive Officer National Health and Medical Research Council

THE SIDNEY KIMMEL COMPREHENSIVE CANCER CENTER AT JOHNS HOPKINS

Beyond the HIPAA Privacy Rule: Enhancing Privacy, Improving Health Through Research

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

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

Master of Science in Biomedical Sciences

Biobanks: DNA and Research

The University of Texas Health Science Center at San Antonio Tobacco Funds Budget Fiscal Year Ending August 31, 2015

Principles for Protecting Integrity In the Conduct and Reporting Of Clinical Trials

CALIFORNIA STATE UNIVERSITY CHANNEL ISLANDS

CAMPAIGN: Transforming MEDICINE Beyond IMAGINATION

Waiver to Allow Participation in a Food and Drug Administration Advisory Committee

The. for DUKE MEDICINE. Duke University School of Medicine. People

How To Change Medicine

Department of Health and Human Services. Final Guidance Document

Bioethics Education in Professional Science Master s Programs at California State University Channel Islands

The University of Maryland School of Medicine

Resumen Curricular de los Profesores. Jesse Boehm

Clinical Cancer Research: Alternative IRB Models and Enhancing Progress

A Biopharma/Foundation Partnering Model The Leukemia & Lymphoma Society Targets, Leads & Candidates Program

12.0 Investigator Responsibilities

2019 Healthcare That Works for All

Guideline Development The American Society of Clinical Oncology

Graduate Fellowships. Preparing the leaders of tomorrow. school of medicine. The Campaign for Washington University

Genomic Medicine Education Initiatives of the College of American Pathologists

API Overview and Informatics Training and Education J. Mark Tuthill, M. D., Henry Ford Health System. API Overview:

BOOSTING THE COMMERCIAL RETURNS FROM RESEARCH

Principal Investigator and Sub Investigator Responsibilities

American Cancer Society Extramural Grants

Groundbreaking Collaborative Clinical Trial Launched

2014 Metrics on Human Research Protection Program Performance

Challenges in the Regulation of Pediatric Clinical Trials

TRANSLATIONAL BIOINFORMATICS 101

A Closer Look at Stem Cell Treatments An ISSCR Initiative

Inside This Issue. Biostatistics and Bioinformatics Biostatistics and Bioinformatics Department Bios Buzz April 22, 2016 KUDOS

Regulatory Issues in Genetic Testing and Targeted Drug Development

This policy applies to research administrators, investigators, research staff, Human Investigation Committee (HIC) members and HIC staff.

Ranking Member Subcommittee on Labor-HHS-Education Subcommittee on Labor-HHS-Education. Washington, DC Washington, DC 20515

M The Nucleus M The Cytoskeleton M Cell Structure and Dynamics

High Performance Computing Initiatives

The Retrospective Approach To Companion Diagnostics

Accelerating Development and Approval of Targeted Cancer Therapies

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

BBSRC TECHNOLOGY STRATEGY: TECHNOLOGIES NEEDED BY RESEARCH KNOWLEDGE PROVIDERS

The Promise and Challenge of Adaptive Design in Oncology Trials

Quality by Design Concept

Clinical Trials Need More Subjects

PATHOLOGY DEPARTMENTS AT GRADUATE MEDICAL EDUCATION TEACHING INSTITUTIONS

University of Medicine and Dentistry of New Jersey (UMDNJ)

Balancing Big Data for Security, Collaboration and Performance


THE ROLE OF BIG DATA IN HEALTH AND BIOMEDICAL RESEARCH. John Quackenbush Dana-Farber Cancer Institute Harvard School of Public Health

Poster Abstracts for Innovations in Genomics and Biobanking

How To Combine The Two Companies Into A Single Company

Data and Informatics Implementation

The NIH Roadmap: Re-Engineering the Clinical Research Enterprise

Delivering the Promise of Genetic and Genomic Medicine. From Research to Clinical Care:

Clinical Trials: Questions and Answers

Guidance For Research Involving Human Embryonic Stem Cells, Germ Cells, And Cells Obtained From Cord Blood

I. INTRODUCTION DEFINITIONS AND GENERAL PRINCIPLE

A Career in Pediatric Hematology-Oncology? Think About It...

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

The National Institute of Genomic Medicine (INMEGEN) was

Part D Drugs & Medicare Coverage Rules

TRACKS GENETIC EPIDEMIOLOGY

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

The iq&a interactive Medical Intelligence Zone BRCA Gene Testing

50-State Survey of Laws Regulating the Collection, Storage, and Use of Human Tissue Specimens and Associated Data for Research

Workshop on Establishing a Central Resource of Data from Genome Sequencing Projects

The ethics of stem cell research and treatment

#Aim2Innovate. Share session insights and questions socially. Genomics, Precision Medicine and the EHR 6/17/2015

CLINICAL RESEARCH ROLES CLINICAL RESEARCH ROLESCLINICAL RESEARCH ROLES

National Framework for Excellence in

Teaching Medicine and Teaching Science: Is there a difference? From MD to PhD (Clinical Researcher) Ines Vaz Luis September 2013

The protection of individuals who volunteer to participate in research is

Genetic Counsellor. Canada

March 19, Dear Dr. Duvall, Dr. Hambrick, and Ms. Smith,

Using genetic biomarkers to pre-identify oncology patients for clinical trials

CARMEN A. PULIAFITO, M.D., M.B.A. DEAN, KECK SCHOOL OF MEDICINE OF THE UNIVERSITY OF SOUTHERN CALIFORNIA

Research Involving Human Biological Materials: Ethical Issues and Policy Guidance Executive Summary

FACULTY OF ALLIED HEALTH SCIENCES

How To Make Cancer A Clinical Sequencing

Master of Science in Vision Science and Investigative Ophthalmology MVSIO

The Healing Power of Data

THE LINCOLN INSTITUTE OF HEALTH

If you are signing for a minor child, you refers to your child throughout the consent document.

Reimagining GRADUATE EDUCATION

Sharing Data from Large-scale Biological Research Projects: A System of Tripartite Responsibility

Office of Clinical Research and the. Institute (CTSI) Patricia Emmanuel, MD

Adventist HealthCare, Inc.

BIOMEDICAL INFORMATICS Doctor of Philosophy

Summary of Advisory Council on Blood Stem Cell Transplantation: Recommendations and Status

Brad H. Pollock, MPH, PhD

GIORGIO VITTORIO SCAGLIOTTI, M.D. CURRICULUM VITAE

University of Central Florida College of Medicine Industry Relations Policy and Guidelines. Table of Contents

MEMORANDUM. The Establishment of the Center for Integrated Animal Genomics (CIAG) at Iowa State University

A guide for the patient

Transcription:

REPORT BRIEF MARCH 2012 For more information visit www.iom.edu/translationalomics Evolution of Translational Omics Lessons Learned and the Path Forward Sequencing the human genome opened a new era in biomedical science. Researchers have begun to untangle the complex roles of biology and genetics in specific diseases, and now better understand why particular therapies do or do not work in individual patients. New technologies have made it feasible to measure an enormous number of molecules within a tissue or cell; for example, genomics investigates thousands of DNA sequences, and proteomics examines large numbers of proteins. Collectively, these technologies are referred to as omics. Patients look to the scientific community to develop innovative omicsbased tests to more reliably detect disease and to predict their likelihood of responding to specific drugs. However, transforming the great promise of these new technologies into clinical laboratory tests that can help patients directly has happened more slowly than anticipated. The process to translate omics-based discoveries into clinically useful tests is much more demanding than has been widely recognized. For example, verification of the complex computational procedures used to develop omics-based tests requires adequate access to the data, computer code, and computational steps used to develop that test. Also, regulatory oversight of clinical laboratory tests differs from that of drugs. Thus far, the Food and Drug Administration (FDA) has chosen not to review most of these clinical tests. These challenges converged during a recent case involving premature use of omics-based tests in clinical trials at Duke University. Flawed gene-expression tests developed by cancer researchers at Duke were used in three clinical trials to determine which chemotherapy treatment patients with lung or breast cancer would receive. For three years, the Duke investigators rebuffed Patients look to the scientific community to develop innovative omics-based tests to more reliably detect disease and to predict their likelihood of responding to specific drugs.

external criticisms about publications describing their tests. But in July 2010, more than 30 outside scientists raised concerns about the validity of the tests and asked the National Cancer Institute (NCI) to intervene and suspend the trials until the gene-expression tests could be reviewed adequately. The episode led the NCI to request that the Institute of Medicine (IOM) establish a committee to recommend ways to strengthen omicsbased test development and evaluation. While lessons were drawn from several case studies of omics-based test development, including at Duke University, investigating allegations of scientific misconduct at Duke was outside the scope of the IOM study. Best Practices for Future Research and Development In its report Evolution of Translational Omics: Lessons Learned and the Path Forward, the IOM committee identifies best practices to enhance development, evaluation, and translation of omics-based tests while simultaneously strengthening steps to be taken to ensure that these tests are appropriately assessed for scientific validity before they are used to guide patient treatment in clinical trials. (See Figure.) The committee s recommendations speak to the many parties responsible for discovery and development of omics-based tests, including investigators, their institutions, sponsors of research, the FDA, and scientific journals. The committee s test development process guides the actions of these key players at every milestone, as omics-based tests make their way from the laboratory into clinical trials to assess their usefulness in guiding medical decisions for patients. Because omics research relies on the analysis of very large datasets, the use of rigorous methods in statistics, bioinformatics, and data management are essential. Inappropriate methods yield computational procedures that are accurate for the samples used in the initial discovery but are inaccurate for any other samples. Accordingly, the committee recommends that all information needed to verify the test discovery process be disclosed through publication or patent application. In addition, the computational procedures must be locked down (recorded and no longer changed) and then confirmed with a new set of samples not used in the initial discovery. If the locked-down test is not confirmed at this stage, it is much more likely to fail in subsequent steps. Investigators should complete this confirmation without prior knowledge of the clinical information linked to specimens that are being analyzed. At the end of the discovery phase, a candidate omics-based test should be precisely defined, including the molecular measurements, lockeddown computational procedures, and intended clinical use. Next, the candidate omics-based test moves to the validation phase so that its ability to generate accurate and consistent measurements as well as its ability to accurately and reliably predict the clinical condition of interest can be assessed. Any clinical laboratory that reports tests for use in guiding patient care falls under the purview of the Clinical Laboratory Improvement Act (CLIA), which provides oversight for the quality of laboratory operations. For this reason, test validation should be performed in a CLIA-certified laboratory, which should design, optimize, validate, and implement the omics-based test under current clinical laboratory standards. Once the performance of the clinical laboratory test is deemed acceptable, the candidate test is ready for use in a clinical trial to assess its usefulness in guiding medical decisions for patients. The committee strongly urges that investigators consult with the FDA prior to beginning a clinical trial. If decisions about patient care will be guided by omics-based test findings in a clinical trial, this consultation is a legal requirement. 2

The committee suggests that institutions lacking the infrastructure or capability to follow all elements of the recommended test development and evaluation process should consider not engaging in research to develop omics-based tests intended for clinical use. Institutional Policies and Procedures to Support Scientific Integrity The investigators involved in omics research and test development, often working in interdisciplinary teams, bear the greatest responsibility and accountability for the scientific rigor and transparency of omics research. They need to foster a culture of scientific integrity through the discovery and development process. Academic institutions, other non-profit research organizations, and for-profit companies that support development of omics-based tests also are responsible for ensuring proper oversight of the omics research conducted and reported by their respective staffs. An institution s overarching role is to develop policies and procedures that promote scientific integrity, and when circumstances warrant to investigate allegations of incorrect or improper research and reporting practices. The committee stresses the importance of institutional awareness and oversight when developing tests for use in clinical trials, in part because FDA oversight traditionally has not been a central component of clinical laboratory test development. Lack of FDA oversight places an often unrecognized demand on academic institutions to provide proper oversight for omics-based test development, validation, and clinical use. The committee suggests that institutions lacking the infrastructure or capability to follow all elements of the recommended test development and evaluation process should consider not engaging in research to develop omics-based tests intended for clinical use. To avoid adding new barriers to innovation in this promising field, the committee is not recommending creating a brand new system of oversight. Rather, its recommendations focus on existing responsibilities of institutions to ensure the integrity of the scientific process. For example, although nearly all institutions have policies and procedures to disclose and mitigate financial conflicts of interest for individuals, there is often less clarity when handling institutional conflicts of interest both financial and non-financial. In certain instances, an outside body should be asked to investigate when an institution might appear too conflicted. Lessons From the Duke Case Because many published papers describing the omics-based tests developed at Duke University now have been retracted, it is widely accepted that those particular tests were not scientifically valid. Unfortunately, the usual systems to ensure the integrity and rigor of the scientific and translational process failed to prevent those invalid tests from being used in clinical trials. This failure stemmed from problems that may exist at other institutions: unclear lines of accountability, lack of consistently strong data management, lack of independent confirmation of the omics discovery, failure to lock down the specific test methods, inadequate validation of the omics-based test prior to commencing clinical 3

Committee on the Review of Omics-Based Tests for Predicting Patient Outcomes in Clinical Trials Gilbert S. Omenn (Chair) Professor of Internal Medicine, Human Genetics and Public Health; Director, Center for Computational Medicine and Bioinformatics, University of Michigan Medical School Catherine D. DeAngelis Professor of Pediatrics and Health Policy and Management, Johns Hopkins School of Medicine and School of Public Health; Editor-in-Chief Emerita, Journal of the American Medical Association David L. DeMets Professor of Statistics and Biostatistics, University of Wisconsin-Madison Thomas R. Fleming Professor of Biostatistics, Statistics, University of Washington Gail Geller Professor of Medicine, Berman Institute of Bioethics, Johns Hopkins University Joe Gray Gordon Moore Endowed Chair, Department of Biomedical Engineering, Center for Spatial Systems Biomedicine, Oregon Health & Science University Knight Cancer Institute Daniel F. Hayes Clinical Director, Breast Oncology Program; Stuart B. Padnos Professor of Breast Cancer Research, University of Michigan Comprehensive Cancer Center I. Craig Henderson Adjunct Professor of Medicine, University of California-San Francisco Helen Diller Family Comprehensive Cancer Center Larry Kessler Professor and Chair of Health Services, University of Washington School of Public Health Stanley Lapidus Founder, President, and CEO, SynapDx Corporation Debra Leonard Professor and Vice Chair of Laboratory Medicine; Director of Clinical Laboratories, Weill Medical College of Cornell University Harold L. Moses Director Emeritus, Hortense B. Ingram Professor of Molecular Oncology; Professor of Cancer Biology, Medicine, and Pathology, Vanderbilt-Ingram Cancer Center William Pao Associate Professor of Medicine, Cancer Biology, and Pathology/Microbiology/ Immunology; Director Division of Hematology/Oncology; Director of Personalized Cancer Medicine, Vanderbilt University School of Medicine Rebecca D. Pentz Professor of Hematology and Oncology in Research Ethics, Emory School of Medicine Nathan D. Price Associate Professor, Institute for Systems Biology John Quackenbush Professor of Computational Biology and Bioinformatics, Dana-Farber Cancer Institute Elda Railey Cofounder, Research Advocacy Network David Ransohoff Professor of Medicine, Clinical Professor of Epidemiology, University of North Carolina, School of Medicine and Gillings School of Global Public E. Albert Reece Vice President for Medical Affairs, University of Maryland; John Z. and Akiko K. Bowers Distinguished Professor and Dean, University of Maryland School of Medicine Daniela M. Witten Assistant Professor of Biostatistics, University of Washington trials, and individual and institutional conflicts of interest (both financial and non-financial) that are inadequately disclosed in informed consent documents for the trials. Conclusion New omics-based tools have the potential to truly revolutionize patient care, but as with any great promise, they come with great responsibility. The recommendations outlined in this report define best practices and provide a guide along the entire pathway for developing omics-based tests from discovery to clinical trials to assist the many parties contributing to this important translational research. These guidelines, if adopted, can ensure that progress in omics-based test development is grounded in sound scientific practice and is reproducible, resulting not only in improved health care but also and perhaps equally important in continued public trust. f Study Staff Christine Micheel Study Director Sharyl Nass Director, National Cancer Policy Forum; Study Director Laura Levit Program Officer Erin Balogh Associate Program Officer Sarah Domnitz Christine Mirzayan Science and Technology Policy Graduate Fellow Julia E. Dooher Christine Mirzayan Science and Technology Policy Graduate Fellow Study Sponsors Niharika Sathe Research Assistant Michael Park Senior Program Assistant John Bailar Scholar in Residence Patrick Burke Financial Associate National Cancer Institute Food and Drug Administration Centers for Disease Control and Prevention U.S. Department of Veterans Affairs The American Society for Clinical Pathology The College of American Pathologists Roger Herdman Director, Board on Health Care Services Andrew Pope Director, Board on Health Sciences Policy 500 Fifth Street, NW Washington, DC 20001 TEL 202.334.2352 FAX 202.334.1412 www.iom.edu The Institute of Medicine serves as adviser to the nation to improve health. Established in 1970 under the charter of the National Academy of Sciences, the Institute of Medicine provides independent, objective, evidence-based advice to policy makers, health professionals, the private sector, and the public. Copyright 2012 by the National Academy of Sciences. All rights reserved.

FIGURE f MARCH 2012 For more information visit www.iom.edu/translationalomics Evolution of Translational Omics Lessons Learned and the Path Forward FIGURE: Omics-Based Test Development Process Discovery and Test Validation Stage Discovery Phase Candidate Test Developed on Training Set, Followed by Lock-Down of All Computational Procedures Confirmation of Candidate Omics-Based Test Using: 1. An Independent Sample Set If Available (Preferred); OR 2. A Subset of the Training Set NOT Used During Training (Less Preferred) Define Clinical Test Method Test Validation Phase IRB Approval and Consultation with the FDA Analytical Validation Clinical/ Biological Validation Using Blinded Sample Set Defined, Validated, and Locked Down Test (Intended Use, Assay, Computational Procedures, and Interpretation Criteria) B R I G H T L I N E Evaluation for Clinical Utility and Use Stage Three Potential Pathways (IRB Approval and FDA Consultation) Prospective/ Retrospective Study with Archived Specimens No Prospective Clinical Trial; Test Does NOT Direct Patient Management IDE Needed? No Prospective Clinical Trial; Test Directs Patient Management Yes FDA Approval/Clearance or LDT Process for Clinical Test Additional High-Quality Evidence to Evaluate Clinical Utility of the Test Practice Guidelines and Reimbursement Clinical Use NOTE: The computational procedures are locked down in the discovery phase, meaning they are recorded and no longer changed in the subsequent development steps. A clinical test should be fully defined, validated, and locked down before crossing the bright line to enter the stage in which the test undergoes evaluation for its intended clinical use. (FDA is the Food and Drug Administration, IDE is an investigational device exemption, IRB is an Institutional Review Board, and LDT is a laboratory-developed test.)