Adjusting overall survival for treatment switch

Slides:



Advertisements
Similar presentations
Agency for Healthcare Research and Quality (AHRQ)
Advertisements

METHODOLOGY FOR META- ANALYSIS OF TIME TO EVENT TYPE OUTCOMES TO INFORM ECONOMIC EVALUATIONS Nicola Cooper, Alex Sutton, Keith Abrams Department of Health.
Survival Analysis In many medical studies, the primary endpoint is time until an event occurs (e.g. death, remission) Data are typically subject to censoring.
Phase II/III Design: Case Study
Paul Tappenden Jim Chilcott Health Economics and Decision Science (HEDS) School of Health and Related Research (ScHARR) 25 th July 2005 Consensus working.
Controlling for Time Dependent Confounding Using Marginal Structural Models in the Case of a Continuous Treatment O Wang 1, T McMullan 2 1 Amgen, Thousand.
A workshop introducing doubly robust estimation of treatment effects
Designing Clinical Research Studies An overview S.F. O’Brien.
Federal Institute for Drugs and Medical Devices | The Farm is a Federal Institute within the portfolio of the Federal Ministry of Health (Germany) How.
Treatment Switching and Overall Survival in Oncology
Which difference should we target? Alberto Sobrero Ospedale San Martino IRCCS Genova, Italy.
Treatment Switching in the VenUS IV trial Methods to manage treatment non-compliance in RCTs with time-to-event outcomes Caroline Fairhurst York Trials.
Departments of Medicine and Biostatistics
HSRP 734: Advanced Statistical Methods July 24, 2008.
Informative Censoring Addressing Bias in Effect Estimates Due to Study Drop-out Mark van der Laan and Maya Petersen Division of Biostatistics, University.
Sensitivity Analysis for Observational Comparative Effectiveness Research Prepared for: Agency for Healthcare Research and Quality (AHRQ)
Some comments on the 3 papers Robert T. O’Neill Ph.D.
Handling treatment changes in randomised trials with survival outcomes UK Stata Users' Group, September 2014 Ian White MRC Biostatistics Unit, Cambridge,
The role of economic modelling – a brief introduction Francis Ruiz NICE International © NICE 2014.
April 25 Exam April 27 (bring calculator with exp) Cox-Regression
 Confounders are usually controlled with the “standard” response regression model.  The standard model includes confounders as covariates in the response.
Model and Variable Selections for Personalized Medicine Lu Tian (Northwestern University) Hajime Uno (Kitasato University) Tianxi Cai, Els Goetghebeur,
Common Problems in Writing Statistical Plan of Clinical Trial Protocol Liying XU CCTER CUHK.
Sample Size Determination
Cohort Studies Hanna E. Bloomfield, MD, MPH Professor of Medicine Associate Chief of Staff, Research Minneapolis VA Medical Center.
Sample Size Determination Ziad Taib March 7, 2014.
Assessing Survival: Cox Proportional Hazards Model Peter T. Donnan Professor of Epidemiology and Biostatistics Statistics for Health Research.
Analysis of Complex Survey Data
1 Efficacy Results NDA (MTP-PE) Laura Lu Statistical Reviewer Office of Biostatistics FDA/CDER.
Advanced Statistics for Interventional Cardiologists.
Simple Linear Regression
Essentials of survival analysis How to practice evidence based oncology European School of Oncology July 2004 Antwerp, Belgium Dr. Iztok Hozo Professor.
Epidemiology The Basics Only… Adapted with permission from a class presentation developed by Dr. Charles Lynch – University of Iowa, Iowa City.
Statistical approaches to analyse interval-censored data in a confirmatory trial Margareta Puu, AstraZeneca Mölndal 26 April 2006.
Study design P.Olliaro Nov04. Study designs: observational vs. experimental studies What happened?  Case-control study What’s happening?  Cross-sectional.
Assessing Survival: Cox Proportional Hazards Model
Biostatistics Case Studies 2007 Peter D. Christenson Biostatistician Session 3: Incomplete Data in Longitudinal Studies.
The time to progression ratio for phase II trials of personalized medicine Marc Buyse, ScD IDDI, Louvain-la-Neuve, and I-BioStat, Hasselt University, Belgium.
Biostatistics Case Studies 2008 Peter D. Christenson Biostatistician Session 5: Choices for Longitudinal Data Analysis.
Plymouth Health Community NICE Guidance Implementation Group Workshop Two: Debriding agents and specialist wound care clinics. Pressure ulcer risk assessment.
Alec Walker September 2014 Core Characteristics of Randomized Clinical Trials.
1 1 Using Marginal Structural Model to Estimate and Adjust for Causal Effect of Post- discontinuation Chemotherapy on Survival in Cancer Trials Y. Wang,
HSRP 734: Advanced Statistical Methods July 31, 2008.
1 THE ROLE OF COVARIATES IN CLINICAL TRIALS ANALYSES Ralph B. D’Agostino, Sr., PhD Boston University FDA ODAC March 13, 2006.
Empirical Efficiency Maximization: Locally Efficient Covariate Adjustment in Randomized Experiments Daniel B. Rubin Joint work with Mark J. van der Laan.
Use of Candidate Predictive Biomarkers in the Design of Phase III Clinical Trials Richard Simon, D.Sc. Chief, Biometric Research Branch National Cancer.
Osteoarthritis Initiative Analytic Strategies for the OAI Data December 6, 2007 Charles E. McCulloch, Division of Biostatistics, Dept of Epidemiology and.
Patricia Guyot1,2, Nicky J Welton1, AE Ades1
Study designs. Kate O’Donnell General Practice & Primary Care.
1 BLA Sipuleucel-T (APC-8015) FDA Statistical Review and Findings Bo-Guang Zhen, PhD Statistical Reviewer, OBE, CBER March 29, 2007 Cellular, Tissue.
CV-1 Trial 709 The ISEL Study (IRESSA ® Survival Evaluation in Lung Cancer) Summary of Data as of December 16, 2004 Kevin Carroll, MSc Summary of Data.
1 Pulminiq™ Cyclosporine Inhalation Solution Pulmonary Drug Advisory Committee Meeting June 6, 2005 Statistical Evaluation Statistical Evaluation Jyoti.
Response, PFS or OS – what is the best endpoint in advanced colorectal cancer? Marc Buyse IDDI, Louvain-la-Neuve & Hasselt University
Approaches to quantitative data analysis Lara Traeger, PhD Methods in Supportive Oncology Research.
The parametric g-formula and inverse probability weighting
SAMBa ITT – Reducing the Size of Clinical Trials Jonathan Bartlett SAMBa ITT 4 Strictly Confidential 31 st May 2016.
J Clin Oncol 30: R2 윤경한 / Prof. 김시영 Huan Jin, Dongsheng Tu, Naiqing Zhao, Lois E. Shepherd, and Paul E. Goss.
SAMBa ITT – Portfolio Management Alun Bedding SAMBa ITT 4 Strictly Confidential 31 st May 2016.
Methods to Handle Noncompliance
Adjusting effects for treatment switching in HTA
Harvard T.H. Chan School of Public Health
Randomized Trials: A Brief Overview
Jeffrey E. Korte, PhD BMTRY 747: Foundations of Epidemiology II
Common Problems in Writing Statistical Plan of Clinical Trial Protocol
Tim Auton, Astellas September 2014
Advanced Statistical Methods for Translational Research
Use of Piecewise Weighted Log-Rank Test for Trials with Delayed Effect
Chapter 3 Hernán & Robins Observational Studies
Approaches and Challenges in Accounting for Baseline and Post-Baseline Characteristics when Comparing Two Treatments in an Observational/Non-Randomized.
How Should We Select and Define Trial Estimands
Presentation transcript:

Adjusting overall survival for treatment switch Recommendations of a cross-institutional statistical working group Claire Watkins BBS/EFSPI European Scientific Meeting Application of Methods for Health Technology Assessment 23rd June 2015 Disclosure statement: Claire Watkins is an employee of AstraZeneca UK Ltd. The views and opinions expressed herein are my own and cannot and should not necessarily be construed to represent those of AstraZeneca or its affiliates

Outline The working group Background Methods Assumptions and limitations Example Best practice – design and analysis

Treatment switch subteam of the PSI HTA SIG Remit and membership Amongst pharmaceutical industry statisticians working with trials including treatment switches: Raise awareness Current statistical methods Potential applications Strengths and limitations Promote and share best practice, in both analysis and design Encourage research into new and improved methods [Chair] Claire Watkins (AstraZeneca) Pierre Ducournau (Roche) Rachel Hodge (GSK) Xin Huang (Pfizer) Cedric Revil (Roche) Alexandros Sagkriotis (Novartis) Yiyun Tang (Pfizer) Elaine Wright (Roche) [Non-member advisory role] Nicholas Latimer (University of Sheffield)

Key publications

Standard clinical practice (inc gef) Background What do we mean by treatment switch/crossover? We might build this into the trial protocol e.g. Sunitinib GIST trial (Demetri, 2012) Or it might happen spontaneously due to clinical practice in the region, if the treatment is already on the market e.g. Gefitinib IPASS trial (Fukuoka, 2011) Patients in a parallel group RCT may switch or “crossover” to the alternative treatment at some point before an endpoint of interest occurs. Disease prog Randomise Sunitinib Survival Placebo Sunitinib Disease prog Standard clinical practice (inc gef) Randomise Gefitinib Survival Doublet chemo

Potential impact of treatment switch Correlation between PFS and OS in NSCLC Switch prohibited (n=20) R2=0.5341 Switch allowed (n=15) R2=0.0027 Interaction p=0.019 Hotta et al: Progression-free survival and overall survival in phase III trials of molecular-targeted agents in advanced non-small-cell lung cancer. Lung Cancer 79 (2013) 20– 26

Why does this matter for HTA? It all depends on the decision problem If there is switching and the new therapy is effective, ITT underestimates this difference  How to estimate long term efficacy without switch? In HTA, the decision problem is often to compare: Current clinical practice without new therapy Potential future clinical practice including new therapy vs

Commonly used methods to estimate control arm survival in absence of switch “Naive” methods Exclude switchers Censor at switch Time varying covariate “Complex” methods Inverse Probability of Censoring Weighting (IPCW; observational) Rank Preserving Structural Failure Time (RPSFT; randomisation based) Two-stage Accelerated Failure Time Others External data Simple to apply High levels of bias Harder to apply Try to reduce bias

Naive (1): Exclude switchers Control arm survival Compare to observed experimental arm survival Observed (ITT) control arm Exclude switchers Non switchers Non switchers S S Switchers Switchers S S Key Death time Censor time Switch time S Assumption: Switchers and non-switchers have the same prognosis

Naive (2): Censor switchers Control arm survival Compare to observed experimental arm survival Observed (ITT) control arm Censor switchers Non switchers Non switchers S Switchers Switchers S Key Death time Censor time Switch time S Assumption: Switchers and non-switchers have the same prognosis

Naive (3): Time varying covariate Non-switchers Switchers Confounding Covariate Covariate Switch Death Death Assumption: Switchers and non-switchers (with the same covariates) have the same prognosis (i.e. no confounders)

Complex (1): IPCW (weight non-switched times) Control arm survival Compare to observed experimental arm survival Observed (ITT) control arm IPC weighted WEIGHT Non switchers Non switchers WEIGHT WEIGHT S Switchers Switchers WEIGHT S Key Death time Censor time Switch time S Assumption: The variables in the weight calculation fully capture all reasons for switching that are also linked to survival (i.e. no unmeasured confounders) Weights represent how “switch-like” a patient is that has not yet switched Calculated using observational propensity score methods, vary by time and patient

Statistical detail: IPCW An observational data-based two stage modelling approach Modelling stage 1: Population Control arm patients Model Probability of switch over time conditional on baseline and time varying factors (propensity score) Output Time varying subject specific IPC weights Modelling stage 2: Population All patients Model Survival analysis with IPC weights applied to control arm patients (censor at switch) Output IPC weighted Hazard Ratio and KM

Statistical detail: IPCW Modelling stage 1: Obtaining IPC weights Time dependent Cox model for switch (Robins 2000) C{t|V(t), T>t} = 0(t) exp{α’V(t)} Core assumption: V includes all covariates that influence both the decision to switch and survival If not: Estimate will be biased Implication: Additional data collection Hazard of switch Actual switch time Current covariates Baseline hazard Past covariate history

Statistical detail: IPCW Modelling stage 1: Obtaining IPC weights Usually fit using pooled logistic regression (Hernan 2000, Fewell 2004) Analogous to time dependent Cox (D’Agostino, 1990) Split data into patient cycles and use as observations Time varying covariates set as constant within a patient-cycle Delete any cycles after switch Intercepts via a spline function Use patient clustering to ensure robust SE incorporating within patient correlation (e.g. SAS REPEATED statement) Logit P(switch in cycle k) = β’V(k) IPC weight for cycle k ∏ P(do not switch in cycle j|Vbase(j)) j=1..k P(do not switch in cycle j|V(j)) Baseline covariates only; stabilises weights

Statistical detail: IPCW Modelling stage 2: IPC weighted survival analysis Experimental arm: Observed data (weight 1) Control arm prior to switch: IPC weights Control arm after switch: Censor (zero weight) Often fitted using pooled logistic regression as per stage 1 (constant weight within a patient cycle) IPC weighted control arm Kaplan Meier curve can be generated with some effort (Robins 2000)

Complex (2): RPSFT (adjust post switch times) Control arm survival Compare to observed experimental arm survival Observed (ITT) control arm RPSFT adjusted Non switchers Non switchers S Switchers Switchers S Time off experimental Treatment multiplier Time on experimental x Time off experimental Time on experimental Key Death time Censor time Switch time S Assumptions: Each cycle of treatment extends survival by a constant amount. Balanced arms due to randomisation Treatment multiplier <1 means experimental treatment extends life Calculated using an accelerated failure time model assuming balanced arms

Statistical detail: RPSFTM (Robins 1991) Accelerated Failure Time model structure Works by estimating the treatment effect that balances “counterfactual” survival time in the absence of treatment [T(0)] between randomised arms T(0) = Toff + e Ton NOTE: RPSFTM does not increase power -> same p-value as ITT Survival time without experimental Time on experimental Time off experimental Treatment effect Treatment effect <1 is beneficial (time to death is slowed down on trt) Treatment effect >1 is detrimental (time to death is speeded up on trt) Core assumption: Trt effect is the same regardless of when treated

Statistical detail: RPSFTM Estimating  : The G-estimation procedure Due to randomisation, assume T(0) distribution is the same for both arms For a range of  values, calculate T(0) and test if equal (using a standard survival analysis test statistic Z) Select  that best satisfies test statistic Z()=0 Core assumption: randomised arms are balanced

Statistical detail: RPSFTM RPSFT adjusted survival analysis Use observed survival time and event flag for experimental and control arm non-switch patients Use T(0) = estimated survival time in absence of exposure to experimental for control arm switch pts Re-censoring should be applied to event flag Perform standard survival analyses to get the HR & KM SE from standard analysis too small – does not incorporate uncertainty in  Bootstrap CI, or set p-value=ITT and derive CI limits accordingly

RPSFTM (not) in SAS Vote in the SASware ballot for this to be added as a standard analysis option:   https://communities.sas.com/ideas/1325

Complex (3): 2-Stage AFT (observational study) Control arm survival Compare to observed experimental arm survival Observed (ITT) control arm 2-stage AFT adjusted Treat control arm as observational study post progression Re-baseline at progression Collect covariate data at progression Calculate effect of switch treatment adjusting for covariates Adjust switcher data and compare randomised arms P Non switchers P P S Switchers P S Assumptions: The covariates fully capture all reasons for switching that are also linked to survival No time-dependent confounding between P and S Key Death time Censor time Switch time Progression time S P (i.e. no unmeasured confounders) Not valid if switch can occur before progression Does not require covariate data collection after progression

Retrospective historical Prospective contemporaneous Use of external data Control arm without exposure to experimental Use as external validation of analyses, or quasi-control Comparability to pivotal RCT control arm is key - Patient characteristics, eligibility criteria, treatment, time, location, clinical practice, outcome RCT Retrospective historical RWE Prospective contemporaneous Ideally

Key assumptions, strengths and limitations “Naive” methods ITT Switch does not affect survival As per RCT design Assumption unlikely to hold, leading to bias Exclude switchers No confounders (only important difference between switchers and non-switchers is switch treatment) Simple Breaks randomisation Censor at switch Time varying covariate

Key assumptions, strengths and limitations “Naive” methods ITT Switch does not affect survival As per RCT design Assumption unlikely to hold, leading to bias Exclude switchers No confounders (only important difference between switchers and non-switchers is switch treatment) Simple Breaks randomisation Censor at switch Time varying covariate

Key assumptions, strengths and limitations “Complex” methods RPSFTM (Rank Preserving Structural Failure Time Model) Constant treatment effect. Counterfactual survival time is balanced between treatment groups due to randomization. Reduced selection bias. Performs well if constant treatment effect. Preserves randomisation. Preserves ITT p-value (conservative approach) Bias if treatment effect not constant (disease specific). Difficult to understand. Poor performance if survival or experimental trt amount similar on both arms Preserves ITT p-value (do not regain lost power) IPCW (Inverse Probability of Censoring Weighting) No unmeasured confounders. Only important differences between switchers and non switchers are switch treatment and variables included in weight calculation. Can recover lost power due to switch (stronger p- value than ITT) Bias if unmeasured confounders. Poor performance if most patients switch or near perfect switch predictor Can recover lost power due to switch (anti-conservative) or lose power due to loss of events

Key assumptions, strengths and limitations “Complex” methods RPSFTM (Rank Preserving Structural Failure Time Model) Constant treatment effect. Counterfactual survival time is balanced between treatment groups due to randomization. Reduced selection bias. Performs well if constant treatment effect. Preserves randomisation. Preserves ITT p-value (conservative approach) Bias if treatment effect not constant (disease specific). Difficult to understand. Poor performance if survival or experimental trt amount similar on both arms Preserves ITT p-value (do not regain lost power) IPCW (Inverse Probability of Censoring Weighting) No unmeasured confounders. Only important differences between switchers and non switchers are switch treatment and variables included in weight calculation. Can recover lost power due to switch (stronger p- value than ITT) Bias if unmeasured confounders. Poor performance if most patients switch or near perfect switch predictor Can recover lost power due to switch (anti-conservative) or lose power due to loss of events

Overall Survival (Final, 2008) Case study: Sunitinib vs Placebo, GIST Overall Survival (Final, 2008) 103 (87.3%) patients crossed over from placebo to sunitinib treatment Total deaths 176 90

Conventional Analyses Cox Model Hazard Ratio (SU/PB), 95% CI and p-value ITT (naïve) 0.876 (0.679, 1.129), p=0.306 Dropping crossover 0.315 (0.178, 0.555), p<0.0001 Censoring at crossover 0.825 (0.454, 1.499), p=0.527 Time-dependent treatment 0.934 (0.520, 1.679), p=0.820

Overall Survival (Final, 2008) Crossover Adjusted by RPSFT *Estimated by RPSFT model **Empirical 95% CI obtained using bootstrap samples.

Best practice recommendations - Trial design General Up front planning at design stage Be clear about the question, rationale, customer Describe intent to do analyses in protocol Define switch treatment (drug(s), dose(s) etc) Consider external data sources Collect the data to enable robust analysis

When (not) to build in switch Generally preferable not to Situations where rationale more compelling: Strong efficacy signal from existing data Spontaneous switch likely, wish to manage Recommended by external bodies Trial becomes infeasible without Primary question is sequencing

Best practice recommendations – Trial design Additional requirements with Built in switch Clear and unambiguous criteria for switch Blinded evaluation of criteria Identical assessments for each arm

Best practice recommendations - Analysis Take care when extrapolating switch-adjusted data! Pre-specify preferred adjustment method in protocol/ analysis plan with justification Assess plausibility of assumptions Don’t use naive as sole or primary method Don’t use IPCW/2-stage AFT if most patients switch or near perfect predictor of switch Don’t use RPSFTM if near equal exposure or survival Always present ITT Provide results from a range of sensitivity analyses External data – assess and take steps to maximise comparability

Summary and looking to the future No method is universally “best” Situation specific assessment required Best practice recommendations have been outlined Be clear on the decision problem Collect the right data Up front planning Further work and research is needed: Multiple switches Multiple switch treatments Different or less strong assumptions Binary or continuous data Software

References (1) Watkins C et al. Adjusting overall survival for treatment switches: Commonly used methods and practical application. Pharm Stats 2013 Nov-Dec;12(6):348-57 Latimer NR and Abrams KR. NICE DSU Technical Support Document 16: Adjusting survival time estimates in the presence of treatment switching. (2014). Available from http://www.nicedsu.org.uk Demetri GD et al. Complete longitudinal analyses of the randomized, placebo-controlled, Phase III trial of sunitinib in patients with gastrointestinal stromal tumor following imatinib failure. Clin Cancer Res 2012; 18:3170-3179 Fukuoka M et al. Biomarker analyses and final overall survival results from a Phase III, randomized, open label, first-line study of gefitinib versus carboplatin/paclitaxel in clinically selected patients with advanced non-small cell lung cancer in Asia (IPASS). J Clin Oncol 2011; 29(21):2866-2874 Hotta et al. Progression-free survival and overall survival in phase III trials of molecular-targeted agents in advanced non-small-cell lung cancer. Lung Cancer 2013; 79:20-26 Robins J and Finkelstein D. Correcting for Noncompliance and Dependent Censoring in an AIDS Clinical Trial with Inverse Probability of Censoring Weighted (IPCW) Log-Rank Tests. Biometrics 2000; 56(3):779-788 D’Agostino RB et al. Relation of pooled logistic regression to time dependent Cox regression analysis: The Framingham Heart Study. Stat Med 1990; 9:1501-1515 Fewell Z et al. Controlling for a time-dependent confounding using marginal structural models. The Stata Journal 2004; 4(4):402-420 Hernan MA et al. Marginal structural models to estimate the causal effect of zidovudine on the survival of HIV-positive men. Epidemiology 2000; 11(5):561-570

References (2) Robins JM and Tsiatis A. Correcting for non-compliers in randomised trials using rank-preserving structural failure time models. Communications in Statistics - Theory and Methods 1991; 20:2609-2631. White IR et al. strbee: Randomization-based efficacy estimator. The Stata Journal 2002; 2(2):140-150 Huang X and Xu Q. Adjusting the Crossover Effect in Survival Analysis Using a Rank Preserving Structural Failure Time Model: The Case of Sunitinib GIST Trial. MRC HTMR workshop, Feb 2012, available here

Backup slides

Statistical software Naive methods – standard survival analysis software IPCW (a simplified Marginal Structural Model) SAS code for MSMs available in Hernan 2000 and at http://www.hsph.harvard.edu/causal/software/ (second example) Stata code for MSMs available in Fewell 2004 No published code available for IPC weighted KMs RPSFTM Stata strbee package available (White 2002)

Data collection (may require new CRFs) Trial design features Data collection (may require new CRFs) For general understanding (optional): Built in: If and when switch criteria are met, reasons for not following criteria Spontaneous: Reason for switch Outcome of switch therapy For RPSFTM and IPCW (required): Drug, dosing, etc (to identify switch therapy) Start and stop dates of switch therapy For IPCW (required): Baseline characteristics that may influence decision to switch Time varying factors that may influence decision to switch

During ongoing study conduct Monitor switch therapy Built in switch: monitor visit dates vs schedule Take action if needed E.g. amend protocol or analysis plan if original assumptions wrong

Take care with extrapolation of adjusted data Latimer and Abrams 2014 Switch adjustment method RPSFTM 2-stage AFT IPCW Fit separate parametric models to each treatment group Fit to adjusted patient data. Re-censoring required Derive pseudo-patient data from IPC weighted KM. Fit one parametric model to all data and apply proportional treatment effect Fit model to observed experimental arm data and apply IPCW HR Extrapolation method

Confidentiality Notice This file is private and may contain confidential and proprietary information. If you have received this file in error, please notify us and remove it from your system and note that you must not copy, distribute or take any action in reliance on it. Any unauthorized use or disclosure of the contents of this file is not permitted and may be unlawful. AstraZeneca PLC, 2 Kingdom Street, London, W2 6BD, UK, T: +44(0)20 7604 8000, F: +44 (0)20 7604 8151, www.astrazeneca.com