Chrissie Fletcher Amgen Ltd HTA 1-day scientific meeting 25 th Sept 2014 Bayer, Berlin Subgroups for Regulatory vs HTA – Methods and Perspectives.

Slides:



Advertisements
Similar presentations
6th European Patients’ Rights Day The EMA Geriatric Medicines Strategy and the empowered aging patient Francesca Cerreta EMA (European Medicines Agency)
Advertisements

Robert T. O’Neill, Ph.D. Director, Office of Biostatistics CDER, FDA
1 WORKSHOP 4: KEY COMMENTS FROM THE PANEL DISCUSSION The 3rd Kitasato University - Harvard School of Public Health Symposium Wednesday October 2nd - Thursday.
Study Objectives and Questions for Observational Comparative Effectiveness Research Prepared for: Agency for Healthcare Research and Quality (AHRQ)
Protocol Development.
Introduction to the User’s Guide for Developing a Protocol for Observational Comparative Effectiveness Research Prepared for: Agency for Healthcare Research.
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
Safety and Extrapolation Steven Hirschfeld, MD PhD Office of Cellular, Tissue and Gene Therapy Center for Biologics Evaluation and Research FDA.
Federal Institute for Drugs and Medical Devices | The Farm is a Federal Institute within the portfolio of the Federal Ministry of Health (Germany) How.
Strengthening the Medical Device Clinical Trial Enterprise
Many Important Issues Covered Current status of ICH E5 and implementation in individual Asian countries Implementation at a regional level (EU) and practical.
Exploring uncertainty in cost effectiveness analysis NICE International and HITAP copyright © 2013 Francis Ruiz NICE International (acknowledgements to:
Clinical Trial Designs for the Evaluation of Prognostic & Predictive Classifiers Richard Simon, D.Sc. Chief, Biometric Research Branch National Cancer.
Sensitivity Analysis for Observational Comparative Effectiveness Research Prepared for: Agency for Healthcare Research and Quality (AHRQ)
ODAC May 3, Subgroup Analyses in Clinical Trials Stephen L George, PhD Department of Biostatistics and Bioinformatics Duke University Medical Center.
New or existing slides are easily formatted using built-in layouts that can be applied via the Home tab EMA DRAFT GUIDELINE ON SUBGROUPS DISCUSSION April.
Estimation and Reporting of Heterogeneity of Treatment Effects in Observational Comparative Effectiveness Research Prepared for: Agency for Healthcare.
Incorporating considerations about equity in policy briefs What factors are likely to be associated with disadvantage? Are there plausible reasons for.
U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES NATIONAL INSTITUTES OF HEALTH Working with FDA: Biological Products and Clinical Development Critical Path.
Journal Club Alcohol and Health: Current Evidence July-August 2006.
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.
Interpreting Adverse Signals in Diabetes Drug Development Programs Featured Article: Clifford J. Bailey, Ph.D. Diabetes Care Volume 36: 1-9 July, 2013.
Good Clinical Practice GCP
David A H Whiteman MD FAAP FACMG Global Clinical Sciences Leader Shire Pharmaceuticals.
3rd Baltic Conference on Medicines Economic Evaluation, Reimbursement and Rational Use of Pharmaceuticals Pricing and Reimbursement of Pharmaceuticals.
IPhVWP Polish Presidency, Warsaw October 6 th 2011 Almath Spooner Irish Medicines Board Monitoring the outcome of risk minimisation activities.
Norisuke Kawai Clinical Statistics, Pfizer Japan Inc.
Systematic Reviews.
Drug Submissions: Review Process Agnes V. Klein, MD Biologics and Genetic Therapies Directorate February, 2003 www/hc-sc.gc.ca/hpb-dgps/therapeut.
Delivering Robust Outcomes from Multinational Clinical Trials: Principles and Strategies Andreas Sashegyi, PhD Eli Lilly and Company.
1 Statistical Review Dr. Shan Sun-Mitchell. 2 ENT Primary endpoint: Time to treatment failure by day 50 Placebo BDP Patients randomized Number.
Impact of E9 Addendum to Industry
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.
Adaptive Licensing UK. Content What is adaptive licensing? Draft criteria for pilot candidate selection EMA Road Map 2015 and work programme 2012 UK perspective.
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.
1 Updates on Regulatory Requirements for Missing Data Ferran Torres, MD, PhD Hospital Clinic Barcelona Universitat Autònoma de Barcelona.
META-ANALYSIS, RESEARCH SYNTHESES AND SYSTEMATIC REVIEWS © LOUIS COHEN, LAWRENCE MANION & KEITH MORRISON.
The Diabetic Retinopathy Clinical Research Network Effect of Diabetes Education During Retinal Ophthalmology Visits on Diabetes Control (Protocol M) 11.
EBM --- Journal Reading Presenter :呂宥達 Date : 2005/10/27.
General Regulatory Issues in the Development of Drugs Intended for Treatment of Chronic Illness Sharon Hertz, M.D. Medical Officer Division of Anesthetic,
Introduction to the Meeting Introduction to the Meeting Advisory Committee for Pharmaceutical Sciences Clinical Pharmacology Subcommittee November 17-18,
Guidelines Recommandations. Role Ideal mediator for bridging between research findings and actual clinical practice Ideal tool for professionals, managers,
Division of Oncology Drug Products 1 AREAS OF MAJOR STATISTICAL CONCERNS IN THE M01 STUDY Overall (ITT Population) Finding Liver Metastasis Subgroup Finding.
NIHR Themed Call Prevention and treatment of obesity Writing a good application and the role of the RDS 19 th January 2016.
European Patients’ Academy on Therapeutic Innovation Ethical and practical challenges of organising clinical trials in small populations.
Is a meta-analysis right for me? Jaime Peters June 2014.
Health Technology Assessment for Pharmaceuticals and New Medical Technologies - Where are we now? The industry perspective Jenny Hughes, Director, Vaccines.
Regulatory considerations for Subgroup Analysis in Clinical Trials SFDS Journée nationale B&S 2014 Gautier Paux27 November 2014.
Regulatory and Reimbursement Harmonization An Industry Perspective Adrian Griffin | April 2016.
CONFIDENTIAL © 2012 | 1 Writing a Statistical Analysis Plan DIA Medical Writing SIAC July 12, 2012 Peter Riebling, MS, RAC Associate Director, Regulatory.
Off-label Use.
CLINICAL PROTOCOL DEVELOPMENT
Industry Perspective: Expanded Access Programs
The FDA Early Feasibility Study Pilot and the Innovation Pathway
Supplementary Table 1. PRISMA checklist
Deputy Director, Division of Biostatistics No Conflict of Interest
9/17/2018 Meeting local HTA requirements Challenges for the Pharma HTA Statistician Marie-Ange PAGET Project Statistician – Lilly France EFSPI meeting.
Reading Research Papers-A Basic Guide to Critical Analysis
Regulatory perspective
Issues in Hypothesis Testing in the Context of Extrapolation
Tim Auton, Astellas September 2014
Joint BES/BBS Seminar Patient Preference Studies – Introduction
An introduction to EMA’s support for medicines development
BioCapital Europe 2019, Amsterdam
Regulatory Perspective of the Use of EHRs in RCTs
Meta-analysis, systematic reviews and research syntheses
Gregory Levin, FDA/CDER/OTS/OB/DBIII
David Manner JSM Presentation July 29, 2019
Presentation transcript:

Chrissie Fletcher Amgen Ltd HTA 1-day scientific meeting 25 th Sept 2014 Bayer, Berlin Subgroups for Regulatory vs HTA – Methods and Perspectives

Disclaimer (Chrissie Fletcher)  The views expressed herein represent those of the presenter and do not necessarily represent the views or practices of Amgen or the views of the general Pharmaceutical Industry. 2

Outline  Who is interested in subgroup analyses?  Challenges with subgroup analyses  Guidance on methods and perspectives from regulators  Guidance on methods and perspectives from HTA agencies (effectiveness and cost-effectiveness)  Trends in R&D and market access influencing use of subgroup analyses  Recommendations for optimising use of subgroups in drug development 3

Who is interested in subgroup analyses  Pharmaceutical Industry –Demonstrating the benefit-risk profile supports the proposed population  Regulatory agencies –Is the evidence supporting benefit-risk acceptable to grant approval for the proposed population  Reimbursement agencies –Is the incremental balance of benefit-risk better than existing standard of care (e.g. IQWiG) –Is the incremental balance of benefit-risk worth paying for (e.g. NICE)  Patient –Will I benefit from the treatment and what potential side effects may I experience? 4

What questions can be addressed by subgroup analyses?  Does the drug work in a particular subset of patients?  Is the drug effect consistent across different patient subsets?  Is the drug effect (or balance of benefit-risk) more pronounced in a particular subset of patients? 5

Subgroup analyses have numerous challenges  Subgroup analysis definition and pre-specification  Consistency of effects and subgroup by treatment interactions  Multiplicity and replication  Presenting and interpreting subgroup results  Meeting needs for regulators and reimbursement agencies 6

Guidance on methods and perspectives from regulators  Subgroup analyses are covered in numerous regulatory guidance documents –ICH –EMA draft ‘Guideline on the investigation of subgroups in confirmatory clinical trials –FDA –+ other countries (e.g. Switzerland, Australia, Canada) 7

ICH relevant guidelines  ICH E9 Statistical Principles for Clinical Trials  ICH E5 Ethnic Factors in the Acceptability of Foreign Clinical Data  ICH Gender Considerations in the conduct of clinical trials 8

ICH E9 Statistical Principles for Clinical Trials  Subgroup effects should be pre-specified in the protocol as part of the planned analyses  In most cases, subgroup analyses are exploratory, e.g. explore uniformity of treatment effects  When exploratory, results should be interpreted cautiously  A conclusion of treatment efficacy/safety (or lack of) based solely on exploratory subgroups unlikely to be accepted  Dangers of over-interpretation of unplanned subgroup analyses are well known 9

EMA draft guideline on subgroups in confirmatory clinical trials 10  Pre-agreement with regulatory authorities on important subgroups prior to starting trials (key vs exploratory)  Subgroup characteristics should be easy to measure and scale is important  Defining how to assess consistency of effect difficult  Interaction tests are a possible way of approaching subgroup analyses and should be presented with estimates of size of effect in addition to p-values.  Replication across >1 trial can help with interpretation  Analyses depend on heterogeneity in target population  Forest plots are useful for visual display  Bayesian approaches may be potentially useful in some situations

EMA draft guideline on subgroups in confirmatory clinical trials – Industry views 11  EFSPI/PSI submitted comments (~ 18 pages), for example further clarification regarding: −Rare diseases −Subgroups for reimbursement −Dose adjustment for different subgroups… and the impact on benefit/risk −Use of subgroups in adaptive designs −Role of Bayesian methods −Confirmatory subgroups vs exploratory subgroups  EFPIA submitted comments (~ 42 pages), for example further clarification regarding: −Assessing safety in subgroups (+ benefit-risk) −Pre-specification and labelling −Multiplicity, credibility, replication…. −Guideline for assessors and Industry or just assessors?

FDA  Guideline for the Format and Content of the Clinical and Statistical Sections of New Drug Applications emphasized the importance of conducting subset analyses on data from clinical studies submitted in new drug applications (NDAs) – focus on race and ethnicity  Guideline for the Study and Evaluation of Gender Differences in the Clinical Evaluation of Drugs. The guidance specifically called for analyzing trials by gender and for evaluating pharmacokinetics in women. 12

FDA Guideline for the Format and Content of the Clinical and Statistical Sections of New Drug Applications  Evidence to support labeling for specific subgroups (for example, pediatrics, geriatrics, patients with renal failure)  Subgroup hypotheses should be stated explicitly. It should be noted whether the objectives were pre-planned or formulated during or after completion of the study. –Not pre-planned, usually not considered adequate for definite conclusions  If the size of the study permits, relevant demographic or baseline value-defined subgroups should be examined for unusually large or small responses and the results presented, e.g., comparison of effects by severity groups, by age, sex, or race, or by history of prior treatment with a drug of the same class. –not intended to “salvage” an otherwise non-supportive study –may suggest hypotheses worth examining in other studies or –refining labelling information, patient selection, dose selection, etc. 13

Guidance on methods and perspectives from HTA agencies  Subgroup analyses are covered in numerous HTA agency guidance documents –EUnetHTA (network of HTA agencies across Europe) –NICE (England/Wales) –IQWiG (Germany) –+ other countries (e.g. France, Australia, Canada) 14

EUnetHTA Applicability of evidence in the context of a relative effectiveness assessment of pharmaceuticals  “Metaregression, subgroup analysis, and/or separate applicability summary tables may help reviewers, and those using the reports see how well the body of evidence applies to the question at hand.”  “In large clinical trials it is possible to have reliable subgroup analyses which may help prescribers to relate the trial’s findings more closely to patients for whom they are trying to select appropriate therapies”  “Moderators: Are there any analyses of moderator effects—including different subgroups of participants and types of intervention — to assess robustness versus specificity of effects? ” 15

NICE  Require estimates of clinical and cost effectiveness by subgroups  Clearly defined subgroups ideally identified based on expected differential clinical or cost effectiveness because of known biological/other justified factors –Biological plausibility for why subgroups may differ  Ideally pre-defined (e.g. at scoping stage) with rationale for expected subgroup effects, –subgroups could be identified later (post-scoping) –‘relevant subgroups may be identified in terms of differences in 1 or more contributors to absolute treatment effects’ –‘post-hoc data dredging in search of subgroup effects should be avoided and will be viewed sceptically’ 16

NICE  Careful consideration for choice of scale  Statistical precision of all subgroup effects reflected in analysis of parameter uncertainty  Differences in relative effects between subgroups due to chance could be high when multiple subgroups reported  Credibility will be enhanced when expected subgroup effect has pre-specified rationale and consistent across studies) –Quality of analysis, representativeness of evidence and relevance to decision problem important  Subgroups not considered based solely on differential treatment costs 17

IQWiG  Pre-specification –Subgroup analyses rarely planned a priori –Post-hoc results cannot be regarded as confirmatory  Multiplicity –Caution with interpreting results from several subgroups  Lack of power –Subgroup sizes often too small to detect moderate differences (unless included in sample size calculations)  Testing for homogeneity  Despite limitations, subgroups may represent best scientific evidence  Written into law “show a therapeutically relevant added benefit” in patient subgroups –Gender, age, disease severity and disease state required 18

Comparing regulatory and HTA agency method guidelines Similarities  Prospectively defined and statistically powered (ideal)  Biologic rationale  Small number of subgroups tested  Assessment of heterogeneity  Replication Differences  Important subgroups could be identified post-design (HTA)  Advice on required subgroups from regulators and HTA agencies available at different stages  Robustness (sensitivity) and levels of uncertainty (HTA) 19

Trends in R&D and market access influencing use of subgroup analyses  Personalised/stratified medicine –Biomarkers  Adaptive licensing –Accumulating evidence  Increased data transparency –Evaluating new subgroups  Real world data –Evaluating effectiveness  Economic pressures in healthcare systems –Rationalising treatment decisions 20

Principles and best practices for subgroup analyses (Paget et al) 21  Subgroups pre-specification & definition  Subgroup by treatment interaction  Multiplicity issues  Sensitivity analyses  Replication  Source of evidence  Presenting and reporting subgroup results

Relevance for cost-effectiveness subgroup analyses (Fletcher et al) Clinical effectiveness  Sensitivity analyses  Presenting and reporting  Replication  Source of evidence  Subgroups pre- specifications  Multiplicity issues  Subgroup by trt interaction Cost-effectiveness Desirable Less important Extremely important Important All data sources Transparency trt: treatment 22

Recommendations for optimal use of subgroups in drug development Subgroup analyses remains a difficult area Plan for subgroups in design and analysis −Individual RCTs −Across product development program −Benefit-risk assessments Present and interpret subgroup results appropriately Discuss subgroup analysis strategies with regulatory agencies and reimbursement agencies −Assess if different strategies are needed in different regions/countries −Understand how each stakeholder will view subgroup results 23

Conclusions  Subgroup analyses are important for regulatory and HTA decision making  Variety of guidance on methods for subgroup analyses from regulatory and HTA agencies  Whilst there is agreement on key principles, there are differences in perspectives between these stakeholders  Discussing subgroup strategies with regulators and HTA agencies should be a priority (via scientific advice and early dialogue)  Statisticians add strategic value in optimising the use of subgroup analyses for regulatory and HTA decision making 24

References  ICH E9 Statistical principles for clinical trials eline.pdf eline.pdf  ICH E5 Ethnic Factors in the Acceptability of Foreign Clinical Data 1__Guideline.pdf 1__Guideline.pdf  ICH E7 Studies in support of special populations: geriatrics eline.pdf eline.pdf  ICH E11 Clinical investigation of medicinal products in the pediatric population ideline.pdf ideline.pdf  ICH Gender considerations in the conduct of clinical trials: 87.pdf 87.pdf  E17: General principle on planning/designing Multi-Regional Clinical Trials ncept_Paper_July_2014.pdf ncept_Paper_July_2014.pdf  EMA draft ‘Guideline on the investigation of subgroups in confirmatory clinical trials’ 23.pdf) pdf 25

References  FDA Guideline for the Format and Content of the Clinical and Statistical Sections of New Drug Applications  FDA Guideline for the Study and Evaluation of Gender Differences in the Clinical Evaluation of Drugs.  EUnetHTA (network of HTA agencies across Europe) Guidelines for Rapid Relative Effectiveness Assessment of Pharmaceuticals:  NICE (England/Wales) Methods guide to Health Technology Assessment: appraisal-2013-pdf appraisal-2013-pdf  IQWiG (Germany) General Methods (Version 4.1)  Questioning Patient Subgroups for Benefit Assessment: Challenging the German Gemeinsamer Bundesausschuss Approach :  Paget, Chuang-Stein, Fletcher, Reid. Subgroup analyses of clinical effectiveness to support health technology assessments. Pharmaceut. Statist. 2011, –538  Fletcher C, Chuang-Stein C, Paget MA, Reid C, Hawkins N. Subgroup analyses in cost- effectiveness analyses to support health technology assessments. Pharm.Stat Jul- Aug;13(4):