General Considerations for Clinical Trial Design

Similar documents
Challenges in the Regulation of Pediatric Clinical Trials

U.S. Food and Drug Administration

Glossary of Clinical Trial Terms

Overview of Phase 1 Oncology Trials of Biologic Therapeutics

The Clinical Trials Process an educated patient s guide

Rare Diseases: Common Issues in Drug Development Guidance for Industry

Not All Clinical Trials Are Created Equal Understanding the Different Phases

Guidance for Industry

NONCLINICAL EVALUATION FOR ANTICANCER PHARMACEUTICALS

Reflection paper on clinical aspects related to tissue engineered products

University of Hawai i Human Studies Program. Guidelines for Developing a Clinical Research Protocol

Guidance for Industry Determining the Extent of Safety Data Collection Needed in Late Stage Premarket and Postapproval Clinical Investigations

Clinical Trial Design. Sponsored by Center for Cancer Research National Cancer Institute

Guidance for Industry

Guidance for Industry

GENERAL CONSIDERATIONS FOR CLINICAL TRIALS E8

CTC Technology Readiness Levels

First In Human Pediatric Trials and Safety Assessment for Rare and Orphan Diseases

ICH Topic E 8 General Considerations for Clinical Trials. Step 5 NOTE FOR GUIDANCE ON GENERAL CONSIDERATIONS FOR CLINICAL TRIALS (CPMP/ICH/291/95)

Regulatory Pathways for Licensure and Use of Ebola Virus Vaccines During the Current Outbreak FDA Perspective

Guidance for Industry

Guidance for Industry

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

Adoption by CHMP for release for consultation November End of consultation (deadline for comments) 31 March 2011

Session 6 Clinical Trial Assessment Phase I Clinical Trial

Clinical trials for medical devices: FDA and the IDE process

4.1 Objectives of Clinical Trial Assessment

Overview of Drug Development: the Regulatory Process

Subject: No. Page PROTOCOL AND CASE REPORT FORM DEVELOPMENT AND REVIEW Standard Operating Procedure

The Promise and Challenge of Adaptive Design in Oncology Trials

Guideline for Industry

Guidance for Industry

Guidance for Industry Diabetes Mellitus Evaluating Cardiovascular Risk in New Antidiabetic Therapies to Treat Type 2 Diabetes

Pharmacology skills for drug discovery. Why is pharmacology important?

INTERNATIONAL CONFERENCE ON HARMONISATION OF TECHNICAL REQUIREMENTS FOR REGISTRATION OF PHARMACEUTICALS FOR HUMAN USE. Current Step 4 version

Guidance for Industry

Participating in Alzheimer s Disease Clinical Trials and Studies

GT-020 Phase 1 Clinical Trial: Results of Second Cohort

Basic Overview of Preclinical Toxicology Animal Models

Safety Assessment for IND Safety Reporting Guidance for Industry

INTERNATIONAL CONFERENCE ON HARMONISATION OF TECHNICAL REQUIREMENTS FOR REGISTRATION OF PHARMACEUTICALS FOR HUMAN USE

Guidance for Industry FDA Approval of New Cancer Treatment Uses for Marketed Drug and Biological Products

Regulatory Issues in Genetic Testing and Targeted Drug Development

Guidance for Industry

Anticancer Drug Clinical Trial Guideline. (version 2.0)

Guidance for Industry Expedited Programs for Serious Conditions Drugs and Biologics

Cancer Clinical Trials

COMMITTEE FOR HUMAN MEDICINAL PRODUCTS (CHMP) DRAFT GUIDELINE ON THE EVALUATION OF MEDICINAL PRODUCTS FOR CARDIOVASCULAR DISEASE PREVENTION

Clinical Trial Designs for Incorporating Multiple Biomarkers in Combination Studies with Targeted Agents

Laurie Shaker-Irwin, Ph.D., M.S. Co-Leader, Regulatory Knowledge and Research Ethics UCLA Clinical and Translational Science Institute

Achieving Regulatory Success: Areas of focus for biotechnology companies. Michael J. Schlosser, PhD, DABT April 21, 2013

Guidance for Industry

Biosimilars: Additional Questions and Answers Regarding Implementation of the Biologics Price Competition and Innovation Act of 2009

BIOTECHNOLOGY OPERATIONS

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP)

Guidance for Industry

Expanded Access Programs. Richard Klein Office of Special Health issues Food and Drug Administration

INTERNATIONAL CONFERENCE ON HARMONISATION OF TECHNICAL REQUIREMENTS FOR REGISTRATION OF PHARMACEUTICALS FOR HUMAN USE S1A. Current Step 4 version

Die Zulassung von Ultra Orphans und die Rolle des COMP und CAT 14. DGRA Jahreskongress , Bonn

EMA EFPIA workshop Break-out session no. 4

Goals & Objectives. Drug Development & the FDA Pharmacy 309. Outline. An History of Disasters. Be able to describe

How To Weigh Data From A Study

Clinical Trial Protocol Development. Developed by Center for Cancer Research, National Cancer Institute Endorsed by the CTN SIG Leadership Group

Sheffield Kidney Institute. Planning a Clinical Trial

Chander Sehgal, MD, MBA Director, Common Drug Review (CDR) Taipei, Taiwan July 23 26, 2012

Newsletter. WntResearch AB, Medeon Science Park, Per Albin Hanssons väg 41, Malmö, Sweden. Primary Objective:

Nova Southeastern University Institutional Review Board Policies and Procedures

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP) GUIDELINE ON THE EVALUATION OF MEDICINAL PRODUCTS FOR CARDIOVASCULAR DISEASE PREVENTION

Medicine Safety Glossary

Research on Research: Learning about Phase 1 Trials

Atrial Fibrillation in the ICU: Attempting to defend 4 controversial statements

Guidance for Clinical Investigators, Sponsors, and IRBs

Role of the Investigational Drug Services (IDS) in the Management of Investigational Drugs

Study Design and Statistical Analysis

Selected Ethical and Legal Issues in Pediatric Clinical Research

Causality Assessment in Practice Pharmaceutical Industry Perspective. Lachlan MacGregor Senior Safety Scientist F. Hoffmann-La Roche Ltd.

ICH Topic S 1 A The Need for Carcinogenicity Studies of Pharmaceuticals. Step 5

CLINICAL RESEARCH PROTOCOL CHECKLIST

FDA Issues Final Guidance on Patient-Reported Outcome Measures Used to Support Labeling Claims

Medical management of CHF: A New Class of Medication. Al Timothy, M.D. Cardiovascular Institute of the South

CENTER FOR DRUG EVALUATION AND RESEARCH

Non-clinical development of biologics

ESCMID Online Lecture Library. by author

Omega-3 fatty acids improve the diagnosis-related clinical outcome. Critical Care Medicine April 2006;34(4):972-9

19. Drug Treatment Trials

Journal Club: Niacin in Patients with Low HDL Cholesterol Levels Receiving Intensive Statin Therapy by the AIM-HIGH Investigators

Guidance for Industry

Ethics and Scientific Oversight for Phase 1 Clinical Trials in Hong Kong. Sydney TANG Chairman, HKU/HA HKW IRB November 21, 2015

EXHIBIT COORDINATING PROVISIONS-STATE/FEDERAL LAW, ACCREDITATION STANDARDS AND GEOGRAPHIC EXCEPTIONS MARYLAND

Guidance for Industry Influenza: Developing Drugs for Treatment and/or Prophylaxis

Clinical Trials and YOU

Clinical Study Synopsis

Effective Outsourcing of Clinical Pharmacology Studies in Europe. John Horkulak Executive Director, Eurasian External Clinical Study Operations

Advancing research: a physician s guide to clinical trials

Clinical Trials: The Crux of Cancer Innovation

EMEA RM DRAFT GUIDANCE ISPE RESPONSE 1

1. Overview of Clinical Trials

Introduction to r andomized c linical t rials in c ardiovascular d isease

Summary and general discussion

Transcription:

General Considerations for Clinical Trial Design Steve Winitsky, M.D. Division of Clinical Evaluation and Pharmacology/Toxicology Office of Cellular, Tissue, and Gene Therapies Center for Biologics Evaluation and Research

Disclosure No financial relationships to disclose

Phases of Development: Phase 1 Includes the initial introduction of an investigational new drug into humans. Typically closely monitored and may be conducted in patients or normal volunteer subjects. Designed to determine: The metabolism and pharmacologic actions of the drug in humans (Note: the PK considerations are different among drugs, cell therapies, and gene therapies, as discussed elsewhere in this training course). The side effects associated with increasing doses, and, if possible, to gain early evidence of biologic activity and perhaps efficacy. Information about the drug's safety profile pharmacokinetics and pharmacological effects is obtained in order to help design subsequent Phase 2 studies. The total number of subjects is generally in the range of 20 to 80.

Phases of Development: Phase 2 Includes the controlled clinical studies conducted to evaluate: Preliminary evidence of efficacy of the drug for a particular indication or indications in patients with the disease or condition under study. The evidence may rely on biomarkers, surrogates, or clinical outcomes The common short-term side effects and risks associated with the drug Phase 2 drug studies are typically well controlled, closely monitored, and usually conducted in no more than several hundred subjects. For cell and gene therapies, the size of Phase 2 studies is often limited by practical concerns, but Phase 2 is important for planning subsequent Phase 3 pivotal trials.

Phases of Development: Phase 3 Expanded controlled and uncontrolled trials (e.g., long-term open-label extensions of controlled trials) Performed after preliminary evidence suggesting effectiveness of the drug has been obtained Intended to gather sufficient information about efficacy and safety needed: To evaluate the overall benefit-risk relationship of the drug, and To provide an adequate basis for labeling. Phase 3 studies usually include from several hundred to several thousand subjects.

Product Development Plan A Phase 1 study should be one component of a larger development plan Designing a trial that has the potential to effectively test a hypotheses relies on: Knowledge of disease pathophysiology (e.g., if a protein is noted to be expressed at low levels in the disease state, is it merely a marker, is it an adaptive response that may be beneficial, or is it part of the disease pathophysiology?)

Product Development Plan Designing a trial that has the potential to effectively test a hypotheses also relies on: Knowledge of your product What is the pattern of expression, and how does this fit with the scheme for treatment of the disease (e.g., does the time of peak expression correlate with the window for injury in the case of prevention of post-operative ischemia using a vector-delivered transgene?)? For a chronic disease, what is the long-term scheme for treatment if effects are transitory? Is only a single dose required to modify the disease? If multiple dosing is required, how will this be addressed in the development plan? Is multiple dosing even feasible (e.g., tissue trauma due to repeated injection of a small muscle, toxicity, immunogenicity)?

Overview of Required Content of Phase 1 Protocols Phase 1 protocols should be directed primarily at providing an outline of the investigation: An estimate of the number of patients to be involved General enrollment criteria, with a detailed description of safety exclusions A description of the dosing plan including duration, dose, or method to be used in determining dose

Phase 1 Protocol Elements Background Pre-clinical studies In vitro testing Bench testing (delivery logistics) Animal studies Previous experience from human studies Rationale What is the mechanism of action What is the basis for the hypothesis that the proposed treatment will have some activity against the disease? For example, if cell engraftment is felt to be critical, have you assessed whether injected cells are retained in the target tissue for any appreciable amount of time?

Objectives Phase 1 Trial Design Phase 1 studies are primarily focused on the evaluation of safety Preliminary evidence of efficacy For cell and gene therapies, the clinical studies should provide some evidence of biological activity of the product.

Study population Selection of the study population Depends on pre-clinical and clinical safety and preliminary efficacy data Consider enrolling subjects whose disease falls within a limited range of severity When obtaining preliminary evidence of efficacy, if severity of disease in the study group is too high, it may ultimately be difficult to demonstrate an impact on disease outcome in later trials Results may be confounded by inclusion of multiple subgroups that differ in natural course of disease Later-phase studies may enroll a more general population to address the issue of generalizability Exclude subjects who are at high risk of complications Exclude vulnerable populations if feasible

Dosing Dose Starting dose should be selected based on pre-clinical studies Dosing Regimen Single dose for first human trial; multiple doses for later Phase 1 trials Usually 1 route of administration per trial Dose escalation should be conservative for first-inhuman trial Other Considerations Are there issues (e.g., cell clumping) that affect reproducibility of a consistent dose What should be done for subjects for whom collected/expanded cells do not meet lot release criteria (# of cells)

Controls and Blinding Control comparator arms None (open-label study) Standard of care comparator Placebo Sham (e.g., catheter-based delivery of cells to the myocardium) Blinding Single-blinded (subjects only) Double-blinded (subjects and investigators/assessors) Open-label trial: no blinding

Sample Size In general, cohorts should be fairly small (less than 20 subjects in a single cohort)

Enrollment Rate Staggered intra-cohort and inter-cohort enrollment of subjects Interval between subjects and between cohorts should be determined by the time course of expected and unexpected adverse events, as well as an estimate of the time required to assess these events. Once a safety signal arises, the trial can be halted prior to enrollment of subsequent subjects.

Study Period Duration of the study should be appropriate for the disease (e.g., studies of acute versus chronic disease have different considerations). Duration should also take into account the mechanism of action of the product (e.g., what is the time by which the intervention will be expected to have had a measurable effect?).

Safety Monitoring Plan Should take into account product characteristics (e.g., vector biodistribution and potential for insertional mutagenesis; transgene s potential for promotion of cancer) when determining duration of long-term monitoring (see CBER guidance document on gene therapy monitoring) Should be based on pre-clinical study data and also on theoretical concerns Should be presented as a table of scheduled assessments Types of assessments and time points for data collection should be sufficient to capture expected and unexpected adverse events. Stopping rules should be strongly considered for all studies under IND, and may be required for higher risk studies.

Endpoints Events or outcomes that can be measured to evaluate the safety or efficacy of a treatment An efficacy endpoint should be clinically relevant. Phase 1 studies often utilize multiple endpoints (it is important to collect as much relevant data as possible, however, an improper analysis of data from multiple endpoints may lead to a misinterpretation of product activity)

Biomarkers in Phase 1 Studies Biomarker: A characteristic that is objectively measured and evaluated as an indicator of normal biological processes, pathogenic processes, or pharmacologic responses to a therapeutic intervention. (Biomarker Definitions Working Group, 2001, Clin Pharmacol Ther 2001;69:89-95) Use of biomarkers may help in early development (both safety and efficacy). A biomarker is not necessarily a surrogate.

Surrogates in Phase 1 Studies Surrogate A biomarker that is intended to substitute for a clinical endpoint. A surrogate endpoint is expected to predict clinical benefit (or harm or lack of benefit or harm) based on epidemiologic, therapeutic, pathophysiologic, or other scientific evidence. (Biomarker Definitions Working Group, 2001, Clin Pharmacol Ther 2001;69:89-95) May allow for faster collection of data; however that should be weighed against the potential for incorrect assumptions regarding preliminary efficacy Examples of biomarkers that are not surrogates for outcome: Left ventricular ejection fraction as a surrogate for mortality in cardiovascular trials Carotid intimal medial thickness as a surrogate for improvement of atherosclerotic disease

Statistical Analysis Plan Analysis is exploratory. Phase 1 studies are not generally powered to demonstrate statistically significant differences in outcome but can still provide some preliminary evidence of efficacy. Pre-specification of the analytical plan improves the interpretability of data.

Common Reasons for Clinical Hold 21 CFR 312.42(b)(i): Human subjects are or would be exposed to an unreasonable and significant risk of illness or injury: Risk-benefit ratio unfavorable Eligibility criteria issues Inadequate justification of dosing scheme Insufficient justification for starting dose/volume What may appear to be a safe volume (e.g., intramyocardial injection) administered to a normal organ may not be safe when administered to a diseased organ (e.g., may cause arrhythmias and diastolic dysfunction). Dose escalation not sufficiently conservative for a first-in-human study Inadequate safety monitoring plan Failure to include stopping rules in the protocol

Contact Information Steven Winitsky, MD Steve.winitsky@fda.hhs.gov General Information: CBER Web page http://www.fda.gov/cber CBER - Office of Communication, Outreach and Development (formerly OCTMA) 1-800-835-4709 or 301-827-1800 matt@fda.hhs.gov (manufacturers or regulated industry) octma@fda.hhs.gov (consumers, health care professionals) combination@fda.gov (Office of Combination Products)