Biomarkers and Surrogates: Underpinnings and Clinical Trial Applications ASENT Annual Meeting March 2009 Marc K. Walton, M.D., Ph.D. Associate Director.

Slides:



Advertisements
Similar presentations
Fundamentals of Clinical Trials
Advertisements

Susan Boynton, VP, Global Regulatory Affairs, Shire
Marc Bailie DVM, PhD Director In Vivo Facility Michigan State University, Chief Development Officer Integrated Nonclinical Development Solutions (INDS)
Single-Patient Use of Investigational Drugs and Biologic Products for Treating Cancer Grant Williams, M.D. Medical Team Leader DODP/CDER/FDA.
The ICH E5 Guidance: An Update on Experiences with its Implementation The ICH E5 Guidance: An Update on Experiences with its Implementation Robert T. O’Neill,
The Statisticians Role in Pharmaceutical Development
U.S. Food and Drug Administration Notice: Archived Document The content in this document is provided on the FDA’s website for reference purposes only.
Safety and Extrapolation Steven Hirschfeld, MD PhD Office of Cellular, Tissue and Gene Therapy Center for Biologics Evaluation and Research FDA.
Synopsis of FDA Colorectal Cancer Endpoints Workshop Michael J. O’Connell, MD Director, Allegheny Cancer Center Associate Chairman, NSABP Pittsburgh, PA.
Substantiation of Health Claims in Advertising: Probiotics Richard L. Cleland Division of Advertising Practices Federal Trade Commission.
1 PK/PD modeling within regulatory submissions Is it used? Can it be used and if yes, where? Views from industry 24 September 2008.
U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES NATIONAL INSTITUTES OF HEALTH Working with FDA: Biological Products and Clinical Development Critical Path.
The ICH E5 Question and Answer Document Status and Content Robert T. O’Neill, Ph.D. Director, Office of Biostatistics, CDER, FDA Presented at the 4th Kitasato-Harvard.
Introduction to Research
ABCWINRisk and Statistics1 Risk and Statistics Risk Assessment in Clinical Decision Making Ulrich Mansmann Medical Statistics Branch University of Heidelberg.
Meeting Agenda Presentations on endpoints –Regulatory issues –Scientific issues Pros and cons of end points –Classical end points –Non-classical end points.
Critical Appraisal of an Article by Dr. I. Selvaraj B. SC. ,M. B. B. S
…patient reported outcome (PRO) measure for your clinical study Dr Keith Meadows, DHP Research & Consultancy Ltd.
Thoughts on Biomarker Discovery and Validation Karla Ballman, Ph.D. Division of Biostatistics October 29, 2007.
David A H Whiteman MD FAAP FACMG Global Clinical Sciences Leader Shire Pharmaceuticals.
Achieving and Demonstrating “Quality-by-Design” with Respect to Drug Release/dissolution Performance for Conventional or Immediate Release Solid Oral Dosage.
Integrating Scientific Advances into Regulation: Pharmacogenomics and Pharmacogenetics Janet Woodcock, M.D. Director, Center for Drug Evaluation and Research.
Center for Drug Evaluation and Research August 2005 Electroretinography: The FDA’s Viewpoint Wiley A. Chambers, MD Deputy Director Division of Anti-Infective.
Performance Measurement and Analysis for Health Organizations
First In Human Pediatric Trials and Safety Assessment for Rare and Orphan Diseases Andrew E. Mulberg, MD, FAAP Division Deputy Director OND/ODE3/DGIEP.
Drug Submissions: Review Process Agnes V. Klein, MD Biologics and Genetic Therapies Directorate February, 2003 www/hc-sc.gc.ca/hpb-dgps/therapeut.
Exploratory IND Studies
Nonclinical Perspective on Initiating Phase 1 Studies for Small Molecular Weight Compounds John K. Leighton, PH.D., DABT Supervisory Pharmacologist Division.
Investigational Drugs in the hospital. + What is Investigational Drug? Investigational or experimental drugs are new drugs that have not yet been approved.
The Practical Art of Endpoint Selection: Industry Perspectives A View from the Pharma Industry of the FDA Guidance on PROs Glenn A. Phillips, Ph.D. Director.
Dr K N Prasad Community Medicine
Update From FDA: Office of the Commissioner and Center for Drug Evaluation and Research Janet Woodcock, M.D. Acting Deputy Commissioner for Operations.
1 Biomarkers as Surrogates Tipping the Balance Toward Persuasiveness EveryLife Foundation Rare Disease Workshop May 2013 Washington, DC Marc K Walton MD,
Proposal for End-of-Phase 2A (EOP2A) Meetings Advisory Committee for Pharmaceutical Sciences Clinical Pharmacology Subcommittee November 17-18, 2003 Lawrence.
Regulatory Affairs and Adaptive Designs Greg Enas, PhD, RAC Director, Endocrinology/Metabolism US Regulatory Affairs Eli Lilly and Company.
DIVISION OF REPRODUCTIVE AND UROLOGIC PRODUCTS Physician Labeling Rule Lisa Soule, M.D.
Federal Institute for Drugs and Medical Devices The BfArM is a Federal Institute within the portfolio of the Federal Ministry of Health (BMG) The use of.
Clinical Pharmacology Subcommittee of the Advisory Committee for Pharmaceutical Science Meeting April Quantitative risk analysis using exposure-response.
Basic Nursing: Foundations of Skills & Concepts Chapter 9
Using Predictive Classifiers in the Design of Phase III Clinical Trials Richard Simon, D.Sc. Chief, Biometric Research Branch National Cancer Institute.
1 Nonprescription Drugs AC Meeting March 23, 2005 Efficacy of OTC Healthcare Antiseptics: Introduction to the Regulatory Issues Susan S. Johnson, Pharm.D.,
Screening of diseases Dr Zhian S Ramzi Screening 1 Dr. Zhian S Ramzi.
1 Study Design Issues and Considerations in HUS Trials Yan Wang, Ph.D. Statistical Reviewer Division of Biometrics IV OB/OTS/CDER/FDA April 12, 2007.
Current Plan for Critical Path Initiative Janet Woodcock, M.D. Acting Deputy Commissioner For Operations November 5, 2004.
+ Evidence Based Practice University of Utah Evidence-Based Treatment and Practice: New Opportunities to Bridge Clinical Research and Practice, Enhance.
Good Pharmacovigilance Practices
Enrollment and Monitoring Procedures for NCI Supported Clinical Trials Barry Anderson, MD, PhD Cancer Therapy Evaluation Program National Cancer Institute.
Introduction to Research. Purpose of Research Evidence-based practice Validate clinical practice through scientific inquiry Scientific rational must exist.
Introduction to the Meeting Introduction to the Meeting Advisory Committee for Pharmaceutical Sciences Clinical Pharmacology Subcommittee November 17-18,
October 28, F OOD AND DRUG ADMINISTRATION AMENDMENTS ACT OF 2007 (FDAAA) and Risk Evaluation and Mitigation Strategies (REMS) Presented to the Ninth.
Biotechnology Industry Organization (BIO) Risk Management Public Workshop Day 1 - April 9, 2003 Risk Assessment in Drug and Biological Development Joanna.
European Patients’ Academy on Therapeutic Innovation Ethical and practical challenges of organising clinical trials in small populations.
Principles of Assessment and Outcome Measurement for Physical Therapists ksu. edu. sa Dr. taher _ yahoo. com Mohammed TA, Omar,
Surrogate Endpoints as Measures of Efficacy: Complexities & Limitations FDA Advisory Committee November 18, 2002 Michael D. Hughes, Ph.D. Professor of.
Zometa for Prostate Cancer Bone Metastases Protocol 039 Amna Ibrahim, M.D. Oncology Drug Products FDA.
NCI Division of Cancer Prevention Ongoing Activities at Frederick Facilities Presented By: Lori Minasian, M.D. Robert Shoemaker, Ph.D. October 1, 2015.
© 2010 Jones and Bartlett Publishers, LLC. Chapter 12 Clinical Epidemiology.
Early Clinical Development Planning via Biomarkers, Clinical Endpoints, and Simulation: A Case Study to Optimize for Phase 3 Dose Selection (Musser et.
Patient Focused Drug Development An FDA Perspective
Pre-Investigational New Drug (pre-IND) Meeting with FDA
Evidence-based Medicine
Expedited Drug Approval Programs
Reasonable Assurance of Safety and Effectiveness: An FDA Division of Cardiovascular Devices Perspective Bram Zuckerman, MD, FACC Director, FDA Division.
Medical Device Regulatory Essentials: An FDA Division of Cardiovascular Devices Perspective Bram Zuckerman, MD, FACC Director, FDA Division of Cardiovascular.
Regulatory perspective
Speeding access to therapies
New FDA Guidance on Early Alzheimer’s Disease
Friends of Cancer Research
Regulatory Perspective of the Use of EHRs in RCTs
Presentation transcript:

Biomarkers and Surrogates: Underpinnings and Clinical Trial Applications ASENT Annual Meeting March 2009 Marc K. Walton, M.D., Ph.D. Associate Director Office of Translational Science CDER-FDA The views expressed are those of the author, and do not represent an official FDA position

Overview of Content & Structure  Why Bother with Biomarkers? What circumstances enable them to be useful What circumstances enable them to be useful What purposes What purposes  Why Worry with Biomarkers? How & What to worry about with biomarkers How & What to worry about with biomarkers  Qualification What is that? What is that?

Why Biomarkers/Surrogates?  Only clinical (i.e., functional) status is important to patients  BUT in some circumstances Inability to practicably identify disease prior to symptomatic clinical manifestations Inability to practicably identify disease prior to symptomatic clinical manifestations Inadequate ability to definitively diagnosis after disease onset Inadequate ability to definitively diagnosis after disease onset Poor ability to predict future disease course in an individual patient Poor ability to predict future disease course in an individual patient Poor ability assess changes in clinical status of patients Poor ability assess changes in clinical status of patients

Why Biomarkers as Outcome Assessment?  Why is clinical-only approach inadequate?  Many neurological disorders complex Pathogenesis inadequately understood Pathogenesis inadequately understood Variable manifestations from person to person Variable manifestations from person to person Dynamic – variable course within a person Dynamic – variable course within a person  Clinical outcome assessments may have flaws Difficulty in developing precise, sensitive, comprehensive assessment tools Difficulty in developing precise, sensitive, comprehensive assessment tools   Suboptimal clinical assessment of treatment effect related to the nature of the disorder  For same reasons, should not expect developing and understanding biomarkers to be simple

Potential uses of Biomarkers  Early (prior to clinically apparent) disease identification in patient or disease risk assessment  Diagnostic tool at initial clinical onset Definitive diagnosis at early stage Definitive diagnosis at early stage Identification of meaningfully distinct subgroups Identification of meaningfully distinct subgroups  Prediction of future course  Precise assessment of current status  All have relevance to therapeutic development

Biomarkers in Clinical Trials Screening or Baseline Assessment  Patient selection tool  Patient stratification tool Phase 1 Studies – Outcome Assessment  Potential to demonstrate bio-activity effect Biomarker may show early cellular effect more than a clinical outcome effect Biomarker may show early cellular effect more than a clinical outcome effect  Evidence to support devoting further resources  Assist in narrowing dose / regimen range  Biomarker vs Surrogate Endpoint A matter of terminology A matter of terminology

Terminology  Frequently used terms: Biomarker Biomarker Bioactivity endpoint Bioactivity endpoint Pharmacodynamic parameter Pharmacodynamic parameter Activity endpoint Activity endpoint Surrogate endpoint Surrogate endpoint  Exact meaning may vary with speaker and have differing implications

Terminology (cont’d)  Common themes of the varying meanings: Not a measure of clinical benefit Not a measure of clinical benefit Believed to indicate a biological effect of relevance Believed to indicate a biological effect of relevance  Important to specify to what it is relevant  Both nature and time of clinical relationship  May be thought to reflect an effect on an endpoint of direct clinical relevance  May be a phenomenon thought associated with disease process  Critical to specify the intended meaning and use

Phase 2 Studies – Outcome Assessment  Potential to evaluate dose-response relationship (efficacy or safety)  Use to select specific doses / regimens for detailed study  Aid to selection of patient population for detailed study  Aid to interpretation of observed clinical events  Aid estimation of sample size for Phase 3  May critically aid design of successful Phase 3 study Biomarkers in Clinical Trials

Phase 3 Studies – Outcome Assessment  Secondary endpoint Objective Objective Potentially precise Potentially precise Supportive of clinical efficacy endpoint Supportive of clinical efficacy endpoint Biomarkers in Clinical Trials

Phase 3 Studies – Outcome Assessment  Primary Endpoint  ‘Validated’ Surrogate Primary Endpoint Reliability of biomarker known Reliability of biomarker known Relationship to specific clinical outcome well established Relationship to specific clinical outcome well established Validly stands in place of clinical status Validly stands in place of clinical status For the specific disease/condition being studied For the specific disease/condition being studied For the specific intervention being studied For the specific intervention being studied Biomarkers in Clinical Trials

Phase 3 Studies – Outcome Assessment  Unvalidated Surrogate Primary Endpoint Requires strong, but not definitive understanding of clinical relationship Requires strong, but not definitive understanding of clinical relationship Accelerated Approval provisions of regulations Accelerated Approval provisions of regulations  Special circumstances for drug approval based on unvalidated surrogate

Potential Advantages of Activity Measures  May be more rapidly observed than clinical outcome Shorter clinical trial Shorter clinical trial  May be easier to measure Greater compliance in obtaining accurate measure Greater compliance in obtaining accurate measure  Lesser intrinsic variability than a clinical measure Smaller sample size Smaller sample size  More objective measure Accurate results (unbiased) Accurate results (unbiased)  Less expensive to measure Less costly clinical program Less costly clinical program

Potential Hazards of Activity Measures  May Mislead – Disparity with clinical outcome False impression of existence/absence of benefit False impression of existence/absence of benefit Incorrect Dose / Regimen / Population selection Incorrect Dose / Regimen / Population selection Incorrect estimate of size of benefit Incorrect estimate of size of benefit Incorrect estimate of frequency of benefit Incorrect estimate of frequency of benefit  May result in failed trial for an effective drug  Nature of risk from being mislead dependent upon how critical are the resulting decisions  Cause of divergence Alternate mechanisms of action of the intervention Alternate mechanisms of action of the intervention Shape of the surrogate-clinical relationship Shape of the surrogate-clinical relationship

Understanding the Surrogate Measure: Idealized

Understanding the Surrogate: Silent Surrogate

Understanding the Surrogate: Complexity Surrogate Endpoint Drug Intervention Clinical Outcome Sequential Order of Processes P1 P2 P3 Surrogate Endpoint Drug Intervention Clinical Outcome P1 P2 P3

Understanding the Surrogate Measure Clinical Status Biomarker

Understanding the Surrogate Measure: Idealized

Understanding the Surrogate Measure C2C2 C3C3 C4C4 C5C5 C1C1 Clinical Status Biomarker Status b1b1 b2b2

Understanding the Surrogate: Complexity Surrogate Endpoint Drug Intervention Clinical Outcome Sequential Order of Processes P1 P2 P3 Surrogate Endpoint Drug Intervention Clinical Outcome P1 P2 P3

Understanding the Surrogate Measure: Potential Consequence of Complexity C2C2 C1C1 Clinical Status Biomarker Status b1b1 b2b2

Understanding the Surrogate Measure: Precision of Knowledge Clinical Status Biomarker Status b1b1 b2b2 b3b3

Biomarker Qualification  Qualification vs. Validation  Developing FDA Program  Outgrowth of Critical Path Initiative Particularly for biomarkers expected to have repeated application in drug development & evaluation Particularly for biomarkers expected to have repeated application in drug development & evaluation

Biomarker Qualification  Definition: The conclusion that within the stated context of use, the results of biomarker measurements can be relied upon to have a stated interpretation and utility. Change in term aids awareness of need for specificity Change in term aids awareness of need for specificity  Regulatory implication: Industry can rely upon using the biomarker in the approved manner in the IND period, and in NDA and BLA submissions, without needing to request that the relevant CDER review group consider and reconfirm the biomarker.

BQ Usage  The use of a qualified biomarker can be applied in drug development and evaluation if there are NO: Serious study flaws (e.g., unverifiable data, improper performance of assays, etc.) Serious study flaws (e.g., unverifiable data, improper performance of assays, etc.) Attempts to apply the biomarker outside the qualified (i.e., approved) context of use Attempts to apply the biomarker outside the qualified (i.e., approved) context of use New and conflicting scientific facts not known at the time the qualification was granted New and conflicting scientific facts not known at the time the qualification was granted

Biomarkers at FDA  Ad hoc Context of use always drug-dependent Context of use always drug-dependent During drug development and approval During drug development and approval Labeling updates (e.g., pharmacogenomic) Labeling updates (e.g., pharmacogenomic)  Drug/Diagnostic co-development  Biomarker Qualification Process (BQP) Coordinated policy Coordinated policy

BQ Program in Spectrum of Biomarker Efforts  Early Biomarker Development  Late Biomarker Development Engaged with BQ Program Engaged with BQ Program  Other Initiatives FDA collaborates with

BQ Process  A framework for interactions between CDER and biomarker sponsors so that CDER can guide them towards compiling comprehensive evidence to support qualification of their selected biomarker(s)  A mechanism enabling CDER to have a well-organized and center-wide unified, formal review of the data supporting a biomarker, eventually leading to a CDER decision to qualify or not  Enables a publicly available and scientifically well- supported statement by CDER of biomarker qualification providing confidence that the evaluation has been comprehensive and the conclusions can be relied upon

Process within FDA  ‘Sponsor’ brings project to FDA  Interdisciplinary working team assembled within CDER & FDA  Information Package reviewed  Advice as needed on how to further progress development for intended use Ultimately development is thought complete Ultimately development is thought complete  Full review and decision on qualification  Formal statement of qualification granted if appropriate