DMC membership experience. P.Bauer Basel May 2016

Similar documents
Guidance on Data Monitoring Committee: Regulatory Perspective in Japan

Short Course: Adaptive Clinical Trials

Trial oversight SOP for HEY-sponsored CTIMPs

FDA S DRAFT GUIDANCE ON MULTIPLE ENDPOINTS IN CLINICAL TRIALS: OVERVIEW, RECEPTION AND NEXT STEPS. John Scott, Ph.D. FDA/CBER 5 October 2017

Novel multiple testing procedures for structured study objectives and families of hypotheses a case study

Data and Safety Monitoring Boards

Draft agreed by Scientific Advice Working Party 5 September Adopted by CHMP for release for consultation 19 September

Disclaimer This presentation expresses my personal views on this topic and must not be interpreted as the regulatory views or the policy of the FDA

1.4 Applicable Regulatory Requirement(s) Any law(s) and regulation(s) addressing the conduct of clinical trials of investigational products.

The Promise and Challenge of Adaptive Design in Oncology Trials

Experience with Adaptive Dose-Ranging Studies in Early Clinical Development

Adaptive Design for Medical Device Development

External IRB Review What Does it Mean for Your Institution

Some key multiplicity questions on primary and secondary endpoints of RCCTs and possible answers

FDA Drug Approval Process Vicki Seyfert-Margolis, Ph.D.

IRB-GCP and Timelines. Andrew Majewski, MSc. 1 st DOLF Meeting Washington University School of Medicine St Louis, Missouri-USA October th, 2010

APPLICATION INSTRUCTIONS INSTITUTIONAL REVIEW BOARD. Revised January 2010 Page 1

SWOG ONCOLOGY RESEARCH PROFESSIONAL (ORP) MANUAL STUDY PROTOCOL CHAPTER 14 REVISED: OCTOBER 2015

EU Clinical Trial Regulation A view from the Industry

Patient Handbook on Stem Cell Therapies

Ethics Committees/IRBs Today: Challenges for Efficiency and Quality

PHARMA CONGRESS. Pharmacovigilance and Drug Safety: Assessing Future Regulatory and Compliance Developments. October 28, 2008

Understanding clinical research trials

Roles and responsibilities of members and alternates, rapporteur and peer reviewers, experts and observers of the Paediatric Committee (PDCO)

Adaptive Trials. Raphaël Porcher. CRESS, Inserm UMR-S 1153, Université Paris Descartes

Human Research Protection Program Good Clinical Practice Guidance for Investigators Regulatory File Essential Documents

OPTINOSE, INC. CORPORATE GOVERNANCE GUIDELINES

Volunteering for Clinical Trials

Secure Interim Analysis Data Access. Management with ACES. Eric J. Silva, Cytel, Inc. Steven Ketchum, Ph.D., Sunesis Pharmaceuticals

International Transfers of Personal Data at sanofi-aventis R & D

Policy Position. Pharmacy-mediated interchangeability for Similar Biotherapeutic Products (SBPs)

Alternative Trial Designs

Twenty-five years of confirmatory adaptive designs: opportunities and pitfalls

MRC. Clinical Trials. Series MRC GUIDELINES FOR GOOD CLINICAL PRACTICE IN CLINICAL TRIALS. Medical Research Council

DEFINING BEST PRACTICE FOR ADVERSE EVENT REPORTING WITH INDUSTRY-SPONSORED CLINICAL TRIAL MANUSCRIPTS

Introduction to Clinical Research

World Medical Association Declaration of Helsinki

Data Quality and Integrity: From Clinical Monitoring to Marketing Approval

Public release of clinical information in drug submissions and medical device applications

Recommendation on elements required to support the medical plausibility and the assumption of significant benefit for an orphan designation

GCP Refresher and GCP/GCDMP Trends. in the CTN. Denise King, MS, RD, CCRA & Lauren Yesko, BS. Presented by:

Interim Analysis: What Regulatory Affairs Professionals Need to Know

About Clinical Trials

Stephanie Gentilin, CCRA

GCP Basics - refresher

IMMUNOGEN, INC. CORPORATE GOVERNANCE GUIDELINES OF THE BOARD OF DIRECTORS

BRIGHT HORIZONS FAMILY SOLUTIONS, INC. CORPORATE GOVERNANCE GUIDELINES

Equipment and preparation required for one group (2-4 students) to complete the workshop

ICH GCP E6(R2): Changes? Yes Challenges? Not as Difficult as You May Fear Lorrie D. Divers, President QRCP Solutions, Inc.

SIGMA DESIGNS, INC. CORPORATE GOVERNANCE GUIDELINES. (As adopted by the Board of Directors effective as of June 2012)

8. Clinical Trial Assessment Phase II

The EFGCP Report on The Procedure for the Ethical Review of Protocols for Clinical Research Projects in Europe (Update: April 2011) Ireland

WMA Declaration of Helsinki - Ethical

INSPECTION OF INDEPENDENT ETHICS COMMITTEES (IEC) The Italian Experience

THE FDA, THE DRUG APPROVAL PROCESS, AND THE PATIENT VOICE

An Overview of Bayesian Adaptive Clinical Trial Design

Analysis of Clinical Trials with Multiple Objectives

Crowe Critical Appraisal Tool (CCAT) User Guide

Appointment of members to Tinnitus Guideline Committee. Supporting information for applicants. November of 18

Protection of Research Participants: The IRB Process and the Winds of Change

CORPORATE GOVERNANCE King III - Compliance with Principles Assessment Year ending 31 December 2015

1201 Maryland Avenue SW, Suite 900, Washington, DC ,

Guidelines for Developing Randomization Procedures R03

REGULATORY REQUIREMENTS FOR CLINICAL TRIAL APPROVAL MEDICAL DEVICES DR D DIALE MRS P NKAMBULE. 02 December 2015

CANCER CENTER SCIENTIFIC REVIEW COMMITTEE

Quality Assurance in Clinical Trials Introduction

Designing a Disease-Specific Master Protocol

BOARD OF DIRECTORS RYDER SYSTEM, INC. CORPORATE GOVERNANCE GUIDELINES

The Science of Small Clinical Trials

What is New on the Regulatory Front?

Human Research Audit Program. Gabrielle Gaspard, MPH, CCRC Assistant Director, Human Research Compliance

The Risk Management + Design Controls Connection: What Device Makers Need to Know

REQUEST FOR AUTHORISATION TO THE COMPETENT AUTHORITY: REQUEST FOR OPINION OF THE ETHICS COMMITTEE:

NATIONAL INSTITUTE FOR HEALTH AND CARE EXCELLENCE. Health and Social Care Directorate. Indicator Process Guide. Published December 2017

APOGEE ENTERPRISES, INC. CORPORATE GOVERNANCE GUIDELINES

A GUIDE TO THIS REFLECTIONS B RESEARCH STUDY IF YOU RE FIGHTING BREAST CANCER, YOU RE NOT ALONE

Session 2 summary Designs & Methods. Pairwise comparisons approach. Dose finding approaches discussed: Guiding principles for good dose selection

DineEquity, Inc. Corporate Governance Guidelines

CPI CARD GROUP INC. CORPORATE GOVERNANCE GUIDELINES

A sequential test procedure for monitoring a singular safety and efficacy outcome

The EFGCP Report on The Procedure for the Ethical Review of Protocols for Clinical Research Projects in Europe (Update: April 2011) Bulgaria

CORPORATE NEWS EARNINGS PAION AG PUBLISHES GROUP QUARTERLY STATEMENT FOR THE FIRST NINE MONTHS OF 2016

Corporate Governance Guidelines

Standard Operating Procedures Guidelines for Good Clinical Practice

Chapter 1. Pharmaceutical Industry Overview

Adaptive Design for Clinical Trials

INTEL CORPORATION BOARD OF DIRECTORS GUIDELINES ON SIGNIFICANT CORPORATE GOVERNANCE ISSUES

Corporate Governance Statement John Bridgeman Limited

The Role of Adaptive Designs in Clinical Development Program*

SINGAPORE GUIDELINE FOR GOOD CLINICAL PRACTICE (SGGCP)

Expanded Access. to Investigational Drugs & Biologics. for Treatment Use

INTEGRATED ADDENDUM TO ICH E6(R1): GUIDELINE FOR GOOD CLINICAL PRACTICE

Florida State University Policy 7-IRB-

HOSPITAL AUTHORITY (HA) GUIDE

The primary purposes of the Corporate Governance Committee shall be to shape the corporate governance of the Company, and by doing so shall:

Audit Committee Charter

Ethical Principles in Clinical Research

Transcription:

DMC membership experience P.Bauer Basel May 2016

EMA GUIDELINE ON DATA MONITORING COMMITTEES Clinical trials frequently extend over a long period of time. Thus, for ethical reasons it is desirable to ensure that for patients participating in such trials there is no unavoidable increased risk for harm. On the other hand it is also important to ensure that a trial continues for an adequate period of time and is not stopped too early to answer its scientific questions. An independent Data Monitoring Committee (DMC) as a group of experts external to a study that reviews accumulating data from an ongoing clinical trial might serve such tasks. While in general safety monitoring should be the major task for a DMC, other aspects of a clinical trial (e.g. study integrity, design aspects) might also be assessed by a DMC. However, it should be noted that a DMC is not needed for all clinical trials.

No ethical committees No boards No infomed consent How did all that start? The medical doctor is the expert to decide whether and how an experiment should be performed as soon there is uncertainty about which treatment should be applied against a particular disease. Patient information may only have a negative impact on the course of the trial, because of noncompliance or dropping out However, in an area where so many different interests are focusing measures are needed to come to good decisions, to care for patients safety and autonomy and to avoid conflicts of interest!

A very recent experience Membership in an independent data monitoring board of a clinical trial in oncology IDMC (independent data monitoring committee) IC SC (interface committee) (steering committee) Only the IDMC has access to un-blinded data Some members of the IC are employees of the sponsor

Communication of the IDMC Recommendations At each interim analysis, the IDMC will recommend that the trial either continues or stops. The IDMC may also recommend a protocol amendment. IDMC makes recommendations to the Steering Committee through the Interface Committee (IC). The IDMC Chair will notify the IC Chair of the IDMC s recommendations,, immediately after the closed session and within twenty four (24) hours.

Communication (cont.) If the IC decides to accept the IDMC recommendation, then the IC Chair will forward the IDMC recommendation to the Chair of the SC within the timelines If the IC decides to reject the IDMC recommendation (comment: e.g. for changes or for stopping), the IC Chair will prepare an IC certificate of no objection to be forwarded to the Chair of the SC. In addition the IC Chair will prepare a communication to the IDMC Chair.

Why? The sponsor may not be convinced that the IDMC members contribute sufficient expertise for taking responsible decisions? The sponsor may be afraid of the IDMC members to make decisions not in his interest? Consequently often the communicating of any efficacy data is strictly avoided and treatment codes will only be unblinded at a special request. The sponsor may be afraid of an early termination of the trial due to foreseeable safety problems?...

Not to forget How can an assessment of harm be done without balancing against the amount of benefit to be achieved in turn? We plan large studies to get sufficiently narrow confidence on efficacy. How difficult may decisions on (rare) safety signals become if to be taken on accumulating data without formal decision rules? Don t forget that it is the IDMC to be blamed when not reacting to emerging safety issues! Also the experts in the IDMC have their share in the process and have to care for their own reputation!

Not to forget (cont.) It is perfectly acceptable that the company has its own interests. But, the team involved in the development within the sponsor is not without any conflict of interest! It may be in competition with development programs for other drugs in the pipeline. There may be a special pressure on the team in companies with few or no other drugs to come. Will the team always make sustainable decision in the overall interest of the sponsor and the public?

What to do for a member in such a case? After carefully studying the Charta of the different committees - and not being able to get any modification of the way how recommendations of the IDMC are going to be communicated - the chair and the statistician of the IDMC resigned. * E.g., we have asked for at least communicating the recommendations of the IDMC directly to the SC.

Some other actions experienced Stopping recruitement, centralizing the check of the inclusion criteria. Starting a monitoring program with additional investigations and adjudication by external experts because of an unexpected cardiac safety signal. Opposing sponsor s interpretation of the results. * * Poor and stable low quality of the data flow. * * Proposing a design modification not pre-planned and suggested from outside the committee. Debate of poor benefit-risk balance, followed by later decisons in health systems not to refund *...

Adaptive design some complications When such issues concerning committee decisions are a problem for the sponsor in a conventionally designed clinical trial, how difficult will things get if an adaptive design is run? It is the clue of such designs that the decision rule for the adaptive interim analysis are not laid down a priori in all their details. It is perfectly understandable that the sponsor wants to have his share in crucial decisions during the development of his product!

A historical example from the early courageous times of adaptive designs (you may well have seen it before) Who had and how had decisions be taken?

A typical application: Dose selection and confirmative inference (the critical issue of combining phases) Scenario k (multiple) doses, Placebo, parallel groups, balanced Many-one comparisons of doses with Placebo Individual inference at the multiple level α, e.g., by a sequential adaptive Bonferroni, Dunnett, Hochberg or strictly hierarchical test procedure BAUER & KIESER (1999) HOMMEL (2001) POSCH et al. (2005) BRETZ et al.(2006) KÖNIG et al. (2007)

A very early try - phase II study on Eniporide in acute myocardial infarction (ZEYMER et al., JACC 2001) The drug is administered after hospital admission Primary endpoint: cumulative release of the enzyme α-hbdh within 72 hours after drug administration Primary objective: investigate cardio-protective effects, safety and dose finding Multinational, double blind, randomized, placebocontrolled, and adaptive two stage dose finding study with parallel groups

Study design (Tiemann et al., Heart Drug, 2001) Product test, α=0.025 (one-sided), α 1 =0.008 (early rejection), α 0 =0.7 (stopping for futility), c α =0.0038 First stage: - placebo and 4 doses, 100 patients per group - proof of principle by a linear trend test Aim of the interim analysis - obtain some initial evidence of efficacy - select doses for stage 2 - determine sample size for stage 2

Decisions in the interim analysis Maintain all trial procedures (business as usual) Selection of double blind doses 2 and 3 and placebo for 2 nd stage (medians P: 44.2, D 1 : 45.3, D 2 : 40.2, D 3 : 34.0, D 4 : 43.4; p trend = 0.12) For the proof of principle in the 2 nd stage a one sided test for dose 3 versus placebo is planned The individual doses will be tested in a hierarchical manner To achieve a conditional power of 90 % 316 patients per group are needed for stage 2 (using the variance estimate in the interim analysis

Final analysis The t-test D 3 versus P: p =0.55 No rejection of H 0 (no effect at all) Judgement of the company biometrician (one may sympathize with): - a small dose finding study followed by a large phase III study would have needed a much higher sample size - two separate studies would have required a larger sample size and longer time - a conventional dose finding study would have required a higher sample size either

Preparation for the decisions External statisticians (Department of Medical Statistics, University of Vienna, P.B., G.S., M.P.) performed the interim analysis on an up to date data set transferred to Vienna less than a week before the meeting of the decision board The statistical analysis has been prepared extensively using test data The calculations of the main analyses have been evaluated by an independent analysis performed by the company statistician Important information on safety had been updated even later Extra monitoring capacity was required to get a real time data set A proposal for adaptations was made by the external statisticians in the interim report Altogether a bone-breaking task!

The decision board External statisticians Steering Committee Chair (P.I.) DSMC Chair Company Statistician Company project leader Company safety expert Few other people from the company including an expert for finances The decision had to be performed within two days at a neutral location (University of Vienna)

The information provided to the board The whole data base was available on computer, so that, e.g., on demand individual safety information could be retrieved online There was a phone inquiry about the form of the dose response curve to external experts for the drug in the company not sitting in the board To my remembering the decision was performed without any support or advise from outside (which, because of the adaptive design strategy, I would not have considered as a major concern anyway)

Going on The company had prepared the drug supply for several plausible selection strategies The drug batches have been replaced in the centres without creating too much white space Investigators remained blinded with regard to the selection The decision in the board was maintained when planning the second stage. - It is crucial to adhere to the design of the second stage, once chosen! - (Using the concept of preserving the conditional error even further design modifications could have been made)

It was an outstanding clinical trial experience! I am convinced, that the people involved in this pioneering study tried to do and did an honest job The clear negative result of the study and its timely publication are backing that opinion However, the way all these decisions have been made are in contradiction to existing guidance documents: Guideline on Data Monitoring Committees (EMEA, January 2006) Establishing and Operation of Clinical Trial Monitoring Committees (FDA, March 2006)

The decision board External statisticians Steering Committee Chair (P.I.) DSMC Chair (?) Company Statistician Company project leader Company safety expert Few other people from the company including an expert for finances

EMEA - Guideline or in case of complex study designs where a modification of the study design based on unblinded interim data is intended. In such a situation the use of an independent DMC gives more credibility to the process. However, major design modifications are considered exceptional and regulatory advice with respect to the acceptance of the planned procedure(s) should be sought in advance. Potential candidates for a DMC membership should have no financial interest in the outcome of the study. any person (not only employees of the sponsor) involved in the conduct of the clinical trial (e.g. investigators) should not serve on the DMC.

FDA- Guidance We therefore recommend that DMC members for a given trial not include investigators in that trial Knowledge of unblinded interim comparisons from a clinical trial is generally not necessary for those conducting or sponsoring the trial We recommend that sponsors avoid appointing to a DMC any individuals who have relationships with trial investigators and sponsor employees that could be considered reasonably likely to affect their objectivity Unblinded interim data and the results of comparative interim analyses, therefore, should generally not be accessible by anyone other than DMC members or the statistician(s) performing the analyses and presenting them to the DMC.

FDA- Guidance (cont.) Certain types of changes to the protocol, however, such as changes in the primary endpoints, could have substantial impact on the validity of the trial and/or its ability to support the desired regulatory decision if they potentially have been motivated by the interim data. We recommend that sponsors discuss proposed changes of the latter type with FDA before implementation. As a way out companies lay down independence of the DMC from the sponsor in the charter of the DMC. Also some (non-binding) guidance for the adaptation is given for the DMC. However it is also foreseen that the DMC may call in experts from the sponsor if deemed to be necessary.

Some points to consider Which type of board should adapt the design? Can the decisions in adaptive designs be made by a board independent of the sponsor? Should the principal investigator as a natural board member be involved in actually treating and assessing study patients? Up to which detail can the decision when and how to do adaptations be pre-planned in advance? Do we need regulatory people in decision boards of adaptive clinical trials? In the SAN case control study on analgesics and nephropathy regulatory authorities (D, A) nominated members of the Scientific Advisory Committee

Consort Checklist of Items To Include When Reporting Harms in Randomized, Controlled Trials (relevant items) Outcomes List addressed adverse events with definitions for each (with attention, when relevant, to grading, expected vs. unexpected events, reference to standardized and validated definitions, and description of new definitions). Clarify how harms-related information was collected (mode of data collection, timing, attribution methods, intensity of ascertainment, and harmsrelated monitoring and stopping rules, if pertinent). Statistical methods Describe plans for presenting and analyzing information on harms (including coding, handling of recurrent events, specification of timing issues, handling of continuous measures, and any statistical analyses).

Checklist (cont.) Numbers analyzed Provide the denominators for analyses on harms. Outcomes and estimation Ancillary analyses Adverse events Present the absolute risk per arm and per adverse event type, grade, and seriousness, and present appropriate metrics for recurrent events, continuous variables, and scale variables, whenever pertinent. Describe any subgroup analyses and exploratory analyses for harms.

A few for the road What are the appropriate risk sets? Use time till event analyses Should more graphical tools be used? Avoid obvious data errors * Avoid big gaps between the time limit for the analysis and the IDMC meeting Avoid different data bases with diverging information (depending on the reporting track) * The presented material may be too big and redundant (a lot of is simply skipped by the members)

A few for the road (cont.) Why not a IDMC for the whole development program of a new drug? How to integrate knowledge from prior or parallel studies into the IDMC decision? Define adverse events of special interest based on prior evidence * to improve monitoring Provide supportive decisions criteria (based on the available evidence) and their statistical properties

A few for the road (cont.) Put more emphasis on statistical methodology for safety issues There is still a wide field to be ploughed This is in the interest of the public - and should be also in the interest of the sponsor I believe that this will more and more become a quality feature in future drug development Membership in a IDMC may turn out as a statistical border line experience of decisions under uncertainty Thank you for your patience!