Estimands in oncology

Similar documents
ICH E9 (R1) Addendum on Estimands and Sensitivity Analysis

Statistical, clinical and ethical considerations when minimizing confounding for overall survival in cancer immunotherapy trials

How the ICH E9 addendum around estimands may impact our clinical trials

Treatment changes in cancer clinical trials: design and analysis

ICH E9(R1) Technical Document. Estimands and Sensitivity Analysis in Clinical Trials STEP I TECHNICAL DOCUMENT TABLE OF CONTENTS

Estimands and Their Role in Clinical Trials

Estimands. EFPIA webinar Rob Hemmings, Frank Bretz October 2017

DRAFT (Final) Concept Paper On choosing appropriate estimands and defining sensitivity analyses in confirmatory clinical trials

Transmission to CHMP July Adoption by CHMP for release for consultation 20 July Start of consultation 31 August 2017

Estimands and Sensitivity Analysis in Clinical Trials E9(R1)

Estimands in klinischen Studien: was ist das und geht mich das was an?

EU regulatory concerns about missing data: Issues of interpretation and considerations for addressing the problem. Prof. Dr.

Estimands: PSI/EFSPI Special Interest group. Alan Phillips

International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use

Estimands for time to event endpoints in oncology and beyond

Draft ICH Guidance on Estimands and Sensitivity Analyses: Why and What?

COMMITTEE FOR PROPRIETARY MEDICINAL PRODUCTS (CPMP) POINTS TO CONSIDER ON MISSING DATA

Everolimus for the Treatment of Advanced Renal Cell Carcinoma (arcc ) - Additional Analysis Using RPSFT

Statistical challenges of meta-analyses of randomised clinical trials in a regulatory setting

Estimand and analysis considerations of phase 3 clinical trials involving CAR-T A case study in lymphoma

The Logic of Causal Inference. Uwe Siebert, MD, MPH, MSc, ScD

AVOIDING BIAS AND RANDOM ERROR IN DATA ANALYSIS

Adjusting overall survival for treatment switch

Guideline on the use of minimal residual disease as a clinical endpoint in multiple myeloma studies

A (Constructive/Provocative) Critique of the ICH E9 Addendum

Estimand in China. - Consensus and Reflection from CCTS-DIA Estimand Workshop. Luyan Dai Regional Head of Biostatistics Asia Boehringer Ingelheim

Safeguarding public health Subgroup analyses scene setting from the EU regulators perspective

Beyond the intention-to treat effect: Per-protocol effects in randomized trials

Overview of requirements for surrogate endpoint adoption by CHMP: a regulatory perspective. Dr Beatriz Flores CDDF- October 2017

Recent developments for combining evidence within evidence streams: bias-adjusted meta-analysis

A journey towards estimand specification in pain: motivation and challenges

Implementation of estimands in Novo Nordisk

NICE DSU TECHNICAL SUPPORT DOCUMENT 16: ADJUSTING SURVIVAL TIME ESTIMATES IN THE PRESENCE OF TREATMENT SWITCHING

Safeguarding public health Subgroup Analyses: Important, Infuriating and Intractable

Guideline on the evaluation of medicinal products indicated for treatment of bacterial infections (CPMP/EWP/558/95 rev 2)

Strategies for handling missing data in randomised trials

Inverse Probability of Censoring Weighting for Selective Crossover in Oncology Clinical Trials.

ICH E9(R1): terminology, taxonomy, systematic approach. Reflections on trimmed means and undilution. Jay P. Siegel, MD April, 2018 Philadelphia, PA

Adjusting the Crossover Effect in Overall Survival Analysis Using a Rank Preserving Structural Failure Time Model: The Case of Sunitinib GIST Trial

Missing data in clinical trials: making the best of what we haven t got.

Analysis Strategies for Clinical Trials with Treatment Non-Adherence Bohdana Ratitch, PhD

The RoB 2.0 tool (individually randomized, cross-over trials)

Interested parties (organisations or individuals) that commented on the draft document as released for consultation.

WATCHMAN PROTECT AF Study Rev. 6

Missing data in clinical trials: a data interpretation problem with statistical solutions?

Assessing methods for dealing with treatment switching in clinical trials: A follow-up simulation study

Online Supplementary Material

Possible causes of difference among regions How to look at the results from MRCT Non-compliance with GCP and/or protocol Apparent Differences (Play of

Estimands, Missing Data and Sensitivity Analysis: some overview remarks. Roderick Little

Delfini Evidence Tool Kit

Reflection paper on assessment of cardiovascular safety profile of medicinal products

Guidance for Industry Clinical Trial Endpoints for the Approval of Cancer Drugs and Biologics

Kaspar Rufibach (F. Hoffmann-La Roche) Joint work with Mouna Akacha (Novartis)

Some Thoughts on Estimands in a Chronic Pain Indication

Statistics for Clinical Trials: Basics of Phase III Trial Design

Histology independent indications in Oncology

Standards for Analyzable Submission Datasets for Directed Safety Analyses

Cancer Immunotherapy from the Health Technology Assessment (HTA) and Payer Perspectives

Estimands in Chronic Symptomatic Diseases. Martin Jenkins Statistical Science Director, AstraZeneca, Cambridge September 2017

arxiv: v1 [stat.ot] 4 May 2018

EMEA experience with endpoints for Oncology drug approval

Statistical Analysis Plan (SAP)

Estimands in clinical trials

Technology appraisal guidance Published: 27 July 2016 nice.org.uk/guidance/ta399

confirmatory clinical trials - The PMDA Perspective -

(Regulatory) views on Biomarker defined Subgroups

Bevacizumab for the treatment of recurrent advanced ovarian cancer

BEST PRACTICES FOR IMPLEMENTATION AND ANALYSIS OF PAIN SCALE PATIENT REPORTED OUTCOMES IN CLINICAL TRIALS

A response by Servier to the Statement of Reasons provided by NICE

Synopsis (C1034T02) CNTO 95 Module 5.3 Clinical Study Report C1034T02

Benefit - Risk Analysis for Oncology Clinical Trials

The (mis)use of treatment switching adjustment methods in health technology assessment busting some myths!

WORK PLAN FOR THE EFFICACY WORKING PARTY (EWP) CHAIRPERSON: Barbara van Zwieten-Boot

Background Comparative effectiveness of nivolumab

Health authorities are asking for PRO assessment in dossiers From rejection to recognition of PRO

CHL 5225 H Advanced Statistical Methods for Clinical Trials. CHL 5225 H The Language of Clinical Trials

Statistical Analysis Plan

PSI Missing Data Expert Group

Application of Marginal Structural Models in Pharmacoepidemiologic Studies

1 st Appraisal Committee meeting Background & Clinical Effectiveness Gillian Ells & Malcolm Oswald 24/11/2016

Sustained benefits for women with HER2-positive early breast cancer JORGE MADRID BIG GOCCHI PROTOCOLO HERA

Systematic reviews and meta-analyses of observational studies (MOOSE): Checklist.

PATIENTS REPORTED OUTCOMES IN ONCOLOGY PATIENT- CENTERED DRUG DEVELOPMENT: OPPORTUNITIES & CHALLENGES

Fattori predittivi di efficacia ed interpretazione della risposta all immunoterapia. Dott. Matteo Brighenti Oncologia Cremona

Possibilities and risks associated with directly moving from Phase I to Phase III: Adaptive designs and Co

Summary. Table 1 Blinatumomab administration, as per European marketing authorisation

Scottish Medicines Consortium

Title: Lung cancer (non-small-cell, anaplastic lymphoma kinase positive, previously treated) ceritinib

Accounting for treatment switching/discontinuation in comparative effectiveness studies

tenofovir disoproxil (as fumarate), 245mg, film-coated tablet (Viread ) SMC No. (720/11) Gilead Sciences Ltd

SUPPLEMENTARY MATERIAL

Clinical Trial Study Synopsis

Evidence- and Value-based Solutions for Health Care Clinical Improvement Consults, Content Development, Training & Seminars, Tools

When a Threshold Crossing approach may and may not be appropriate: A Case Study in SMA

Glossary. Ó 2010 John Wiley & Sons, Ltd

Technology appraisal guidance Published: 21 December 2016 nice.org.uk/guidance/ta422

Randomized Controlled Trial

Kaspar Rufibach Methods, Collaboration, and Outreach Group (MCO) Department of Biostatistics, Roche Basel

Trial designs fully integrating biomarker information for the evaluation of treatment-effect mechanisms in stratified medicine

Transcription:

Estimands in oncology Steven Teerenstra 1,2 1 Radboud university Nijmegen medical center, NL 2 Statistical assessor at Medicines Evaluation Board (MEB), NL

Disclaimer 2 Views expressed my own, based on my experience at MEB as statistical assessor not to be quoted or interpreted as those of MEB

estimand 3 Estimand Origin: gerundive from the Latin verb estimare estimandum= (the property) that is to be estimated

Intercurrent events 4 Patient 1 Patient 2 Patient 3 Patient 4 Patient 5 Patient 6 Patient 7? Rescue medication Study discontinuation Death Treatment discontinuation due to lack of efficacy Rescue medication Treatment discontinuation due to adverse events Study discontinuation? Treatment complete Randomisation TIMELINE Primary endpoint e.g. treatment discontinuation, switching, rescue/additional medication, competing/terminal event, deviations from protocol (measurements),

Intercurrent events 5 Patient 1 Patient 2 Patient 3 Patient 4 Patient 5 Patient 6 Patient 7? Rescue medication Study discontinuation Death Treatment discontinuation due to lack of efficacy Rescue medication Treatment discontinuation due to adverse events Study discontinuation? Treatment complete Randomisation e.g. treatment discontinuation, switching, rescue/additional medication, competing/terminal event, deviations from protocol (measurements), often causing missing information as to the situation when patients would have adhered to * randomised treatment and * (assessment as per) protocol until end of trial TIMELINE Primary endpoint

Intercurrent events 6 Patient 1 Patient 2 Patient 3 Patient 4 Patient 5 Patient 6 Patient 7? Rescue medication Study discontinuation Death Treatment discontinuation due to lack of efficacy Rescue medication Treatment discontinuation due to adverse events Study discontinuation? Treatment complete Randomisation e.g. treatment discontinuation, switching, rescue/additional medication, competing/terminal event, deviations from protocol (measurements), often causing missing information as to the situation when patients would have adhered to * randomised treatment and * (assessment as per) protocol until end of trial TIMELINE Primary endpoint Relevance, definition, existence endpoint questionable

7 Oncology: intercurrent events->time-to-event endpoints No different from continuous, binary, rate outcomes in principle Deviation from protocol treatment discontinuation of treatment lack of efficacy: e.g. progression AE (substantial) resolution of disease other Especially treatment switching (often no rescue medication) change in concomittant therapy (additional medication)

8 Oncology: intercurrent events->time-to-event endpoints No different from continuous, binary, rate outcomes in principle Deviation from protocol treatment discontinuation of treatment lack of efficacy: e.g. progression AE (substantial) resolution of disease other Especially treatment switching (often no rescue medication) change in concomittant therapy (additional medication) Deviation from protocol assessment undocumented (spontaneous) assessments e.g. undocumented progression altering treatment of disease (intermittent) missed assessments visits leave the trial (permanently): withdrawal by patient or by physician (for reasons as above) Lost-to-followup Terminal/competing event (e.g. death when looking at time to progression)

9 Handling intercurrent events partly different Intercurrent events: Continuous /binary/rate outcomes: currently often hidden in missing data Discussions about non-missing at random time-to-event outcomes: currently often hidden in censored data e.g. PFS/DFS/EFS censoring for start new anti-cancer therapy Censoring for > 1 missed visits Censored for undocumented progression Discussion about informative censoring

Current practice 10 Set rules regarding data collection wrt intercurrent events (in design) handling of intercurrent design (in analysis) Example: Censor control patients who switch to experimental treatment No data collection (on e.g. new progressions) after switch

Caveat 11 However: handling of intercurrent events in design and analysis dictates which effect is estimated (which question is answered) under which assumptions Example: handling of intercurrent event= censor when switched One may think this address the ITT question, because it enables to include everyone starting the trial Effect when everyone would have adhered to treatment: no control patient would have switched to experimental treatment (Hypothetical estimand) Kaplan-Meier/Cox: unbiased for this effect under the assumption: patients in control group that do not switch are representative for those that do switch (censoring due to switching is non-informative wrt outcome)

Usual practice 12 Handling of intercurrent events in design/analysis Effect estimated / Question answered (estimand)

Aim of ICH-E9 addendum 13 Handling of intercurrent events in design/analysis Effect to be estimated / Question to be answered (estimand)

Aim of ICH-E9 addendum 14 Handling of intercurrent events in design/analysis Effect to be estimated / Question to be answered (estimand) Discussion with stakeholders (regulators, industry, HTA, patients,..) about which estimand is primary and why how design and analysis will estimate it (estimator) under which assumptions Sensitivity analyses per estimand i.e. different assumptions (different estimators) for the same question Possibly: which secondary/other estimands For other stake holders / broader view

15 Survival outcomes in oncology estimands Examples PFS (DFS, EFS,..) primary endpoint Censoring rules / data collection after censoring: what is the implied estimand? Which analyses address the same question (estimand) and which different estimands? OS primary endpoint cross-over of control patients to experimental treatment Switch to treatment that changes course of disease substantially e.g. (completely) different mechanism of action (completely) different efficacy (safety) For example: stem cell transplantation (underlying consideration: some stakeholders consider it unfair to use an treatment policy estimand then) Late survival / tolerators effects

PFS: EMA guideline 16 EMA: Appendix 1 to GL on evaluation of anticancer medicinal products in man (CHMP/EWP/205/95 rev.3) progression date= first evidence of objective progression Regardless of violations, discontinuation, change of therapy. If, for a particular study, a different approach is considered to be more appropriate, a justification is expected and CHMP Scientific Advice agreement is recommended at the planning stage. => no censoring for violations, discontinuation, change therapy => treatment policy estimand is preferred by EMA guideline

PFS: FDA guideline 17 FDA: GfI: Clin. trial endpoints for the approval of cancer drugs and biologics progression date = earliest time of observing any progression without prior missing assessments and censoring at the date when the last radiological assessment determined a lack of progression. so: when missing visits: censoring For instance, for the primary analysis, patients going off-study for undocumented clinical progression, change of cancer treatment, or decreasing performance status can be censored at the last adequate tumor assessment. The secondary sensitivity analysis would include these dropouts as progression events.

PFS: FDA guideline 18 censoring when going off study for missed visits undocumented clinical progression, change of cancer treatment, or decreasing performance status going of study: no PFS data collection implied after censoring hypothetical estimand effect if no patient would switch to another treatment and stay in the study until documented clinical progression (measured at prespecified assessments) (Kaplan-Meier/Cox regression) unbiased for this effect under the assumption: patients that do *not* switch/undocu.pd/ps /miss visits are representative for those that do switch/undocu.pd/..

PFS: sensitivity analyses 19 Often many sensitivity analyses presented

PFS: sensitivity analyses 20 Often many sensitivity analyses presented May give a false sense of consistency as to the primary estimand (question at hand) if these sensitivity analyses give similar answers but actually address different estimands (questions) May give a false sense of inconsistency if these do not give similar answers because they are addressing different estimands (questions)

PFS 21 Important: Clarify which estimand / question is addressed Especially for sensititivity analyses Present per estimand (question) the primary analysis (estimator) The sensitivity analyses (estimators) explaining that the same estimand (question) is addressed which assumptions are varied

OS 22 Many intercurrent events: always subsequent therapy: may argue whether this is intercurrent or not If started after protocol specified treatment regimen Some acknowledgement of part of difference in survival due to difference in subsequent therapies hypothetical estimands (e.g. what would be the effect if no subsequent therapy, what would be the effect is all arms had similar subsequent subsequent therapy ) in principle may be interesting for some stakeholders, but difficult to estimate Often a treatment policy estimand (implicity) chosen so intercurrent events and subsequent therapy no issue

OS 23 Situations where not only trt policy estimand could be used: switch to experimental treatment ( cross-over ) switch to therapies that substantially change course of disease Early / late survival effects

Switch to experimental therapy 24 Two estimands (implicitly) used Treatment policy effect Effect if no cross-over would have been available (hypothetical estimand) Issues: Latter often proposed by Sponsor for pure efficacy of randomised trt if hypothetical estimand post-hoc proposed when treatment policy estimand fails Estimators for this hypothetical estimand typically require strong assumptions IPCW: no unmeasured confounders RPSFT: Main assumption: (relative) treatment effect when switching is the same as time of randomisation Two-stage: switch at designated secondary baseline (e.g. progression) and no unmeasured confounders at this secondary baseline

Switch to experimental therapy 25 Important issues: Preplan (and discuss) which estimand is primary and why Preplan to collect extra data to motivate that assumptions of estimators are plausible Avoid switch to experimental therapy (?!)

OS (switch to disease altering therapy) 26 Example: leukaemia EU trial, but different standard of care in regions west-eu: stem cell transplantation part of standard of care east-eu: stem cell transplantation much much less available Preplanned analysis: OS including transplant No censoring for transplant => treatment policy estimand Treatment policy OS: not clinically relevant difference, not statistically significant

OS (switch to disease altering therapy) 27 Post hoc proposed: hypothetical estimand: OS when no transplant would have been available Censoring for transplant with inverse probability weighting on certain characteristics to replace a patient having transplant by a patient not having transplant with those characteristics similar (IPCW). No unmeasured confounders assumption: prognosis, switch, and time of switch are predictable by (time dependent) characteristics used in IPCW But: data-collection (timing & type of variables) often not preplanned and not (fully) convincing that no unmeasured confounders assumption is plausible Even if clin. relevant & stat.sign. effect for hypothetical estimand: may be relevant for east-eu, but not for west-eu (?!)

OS 28 Important issue: A priori discussion which estimand primary, for which stakeholder Preplan data-collection/design accordingly Here: A priori data collection to make no unmeasured confounders assumption needed by IPCW stronger

Early / Late survival effect 29 Based on Fattori predittivi di efficacia ed interpretazione della risposta all immunoterapia by Matteo Brighenti, March 13-14 March, 2017 Borghaei, et al. Presented at the American Society of Clinical Oncology 2016 Annual Meeting; June 3 7, 2016; Chicago, IL, USA. Abstract 9025.

30 February 18, 2016 database lock; min.follow-up: 2 years What is the effect for those able to survive at least 3 months on nivolumab ( late effect, tolerator effect ) Principal stratum estimand Interesting for patients

31 nivolumab docetaxel estimator 1: Landmark analysis with 3-month landmark But groups based on selection on post-baseline covariate => those able to survive 3 months on nivolumab may be prognostically different from those able to survive 3 month on docetaxel => non-randomised comparison => bias

32 Identify those who can survive 3 months on docetaxel estimator 2 based on identification of those who can survive at least 3 months on nivolumab via predictive modeling convincing predictive model for nivolumab survival at 3 months possible? (e.g. because only 44 deaths available for modeling) principal strata estimand often difficult because of selecting the stratum (without bias) Can also look at those who tolerate docetaxel at least 3 months

Estimands: 33 different way to look at questions that were relevant before what question are we trying to answer, under which assumptions and how to translate this in good design and analysis Reframing good practice we (at least partially) did before (R)enforcing clarity in discussions (planning & interpretation) (Re)empowering stake holders

34 Thank you for your attention