Interpretation of effect estimates in competing risks survival models: A simulated analysis of organ-specific progression-free survival in a randomised.

Slides:



Advertisements
Similar presentations
METHODOLOGY FOR META- ANALYSIS OF TIME TO EVENT TYPE OUTCOMES TO INFORM ECONOMIC EVALUATIONS Nicola Cooper, Alex Sutton, Keith Abrams Department of Health.
Advertisements

Evidence synthesis of competing interventions when there is inconsistency in how effectiveness outcomes are measured across studies Nicola Cooper Centre.
Survival Analysis-1 In Survival Analysis the outcome of interest is time to an event In Survival Analysis the outcome of interest is time to an event The.
Foos et al, EASD, Lisbon, 13 September 2011 Comparison of ACCORD trial outcomes with outcomes estimated from modelled and meta- analysis studies Volker.
Segment 3 Introduction to Random Variables - or - You really do not know exactly what is going to happen George Howard.
Robertson JFR et al. J Clin Oncol 2009;27(27):
Meta-analysis & psychotherapy outcome research
Sample Size Determination Ziad Taib March 7, 2014.
Analysis of Complex Survey Data
Marshall University School of Medicine Department of Biochemistry and Microbiology BMS 617 Lecture 12: Multiple and Logistic Regression Marshall University.
Survival analysis Brian Healy, PhD. Previous classes Regression Regression –Linear regression –Multiple regression –Logistic regression.
Marshall University School of Medicine Department of Biochemistry and Microbiology BMS 617 Lecture 10: Survival Curves Marshall University Genomics Core.
Essentials of survival analysis How to practice evidence based oncology European School of Oncology July 2004 Antwerp, Belgium Dr. Iztok Hozo Professor.
Dr Laura Bonnett Department of Biostatistics. UNDERSTANDING SURVIVAL ANALYSIS.
Randomized Trial of Preoperative Chemoradiation Versus Surgery Alone in Patients with Locoregional Esophageal Carcinoma, Ursa et al. Statistical Methods:
What is a non-inferiority trial, and what particular challenges do such trials present? Andrew Nunn MRC Clinical Trials Unit 20th February 2012.
Lenalidomide Maintenance Therapy in Multiple Myeloma: A Meta-Analysis of Randomized Trials Singh PP et al. Proc ASH 2013;Abstract 407.
T Andre, E Quinaux, C Louvet, E Gamelin, O Bouche, E Achille, P Piedbois, N Tubiana-Mathieu, M Buyse and A de Gramont. Updated results at 6 year of the.
Biostatistics Case Studies 2005 Peter D. Christenson Biostatistician Session 6: “Number Needed to Treat” to Prevent One Case.
Introduction Sample Size Calculation for Comparing Strategies in Two-Stage Randomizations with Censored Data Zhiguo Li and Susan Murphy Institute for Social.
Final Analysis of Overall Survival for the Phase III CONFIRM Trial: Fulvestrant 500 mg versus 250 mg Di Leo A et al. Proc SABCS 2012;Abstract S1-4.
Biostatistics Case Studies 2014 Youngju Pak Biostatistician Session 5: Survival Analysis Fundamentals.
1 Using dynamic path analysis to estimate direct and indirect effects of treatment and other fixed covariates in the presence of an internal time-dependent.
Biostatistics Case Studies 2007 Peter D. Christenson Biostatistician Session 2: Aging and Survival.
Some survival basics Developments from the Kaplan-Meier method October
Continued Overall Survival Benefit After 5 Years’ Follow-Up with Bortezomib-Melphalan-Prednisone (VMP) versus Melphalan-Prednisone (MP) in Patients with.
INTRODUCTION TO CLINICAL RESEARCH Survival Analysis – Getting Started Karen Bandeen-Roche, Ph.D. July 20, 2010.
Clinical Trials 2015 Practical Session 2. Exercise I Normally Distributed Response Data H 0 :  = 0 H a :  =  a > 0 n=? α=0.05 (two-sided) β=0.20 
Marshall University School of Medicine Department of Biochemistry and Microbiology BMS 617 Lecture 13: Multiple, Logistic and Proportional Hazards Regression.
Chapter 9 Estimating a Population Proportion Created by Kathy Fritz.
Carolinas Medical Center, Charlotte, NC Website:
Results from the International, Randomized Phase 3 Study of Ibrutinib versus Chlorambucil in Patients 65 Years and Older with Treatment-Naïve CLL/SLL (RESONATE-2TM)1.
Core Research Competencies:
Geisler C et al. Proc ASH 2011;Abstract 290.
An introduction to Survival analysis and Applications to Predicting Recidivism Rebecca S. Frazier, PhD JBS International.
Palumbo A et al. Proc ASH 2012;Abstract 200.
Maury S et al. Proc ASH 2015;Abstract 1.
The Need for Competing Risks
Nivolumab in Patients (Pts) with Relapsed or Refractory Classical Hodgkin Lymphoma (R/R cHL): Clinical Outcomes from Extended Follow-up of a Phase 1 Study.
(2) - Department of Epidemiology and Population Health, and
Pooled Analysis of VA, ACAS, ACST-1 & GALA Trials
Meta-analysis of joint longitudinal and event-time outcomes
JAMA Ophthalmology Journal Club Slides: Effect of Oral Voriconazole on Fungal Keratitis Prajna NV, TKrishnan T, Rajaraman R, et al; Mycotic Ulcer Treatment.
Statistics 103 Monday, July 10, 2017.
Barrios C et al. SABCS 2009;Abstract 46.
Bergh J et al. SABCS 2009;Abstract 23.
Jones SE et al. SABCS 2009;Abstract 5082.
Effect of Obesity on Prognosis after Early Breast Cancer
Measures of Disease Occurrence
EVENT PROJECTION Minzhao Liu, 2018
The role of simultaneous resection of synchronous liver metastasis and primary colorectal cancer Samuel Lo Department of Surgery.
Martin M et al. Proc SABCS 2012;Abstract S1-7.
Clinical states of cirrhosis and competing risks
1 Verstovsek S et al. Proc ASH 2012;Abstract Cervantes F et al.
A decade after the Surgical Treatment for Ischemic Heart Failure (STICH) trial: Weaving firm clinical recommendations from lessons learned  Robert E.
Badwe RA et al. SABCS 2009;Abstract 72.
Cetuximab with chemotherapy as 1st-line treatment for metastatic colorectal cancer: a meta-analysis of the CRYSTAL and OPUS studies according to KRAS.
1Kantarjian HM et al. Lancet Oncol 2011;12:
Branford S et al. Proc ASH 2013;Abstract 254.
Impact of Diabetes Mellitus on the Association of Vascular Disease Before Transplantation With Long-term Transplant and Patient Outcomes After Kidney.
E PLURIBUS HUGO “Out of the Many, a Hugo” v0.92.
Writing a good discussion & conclusion
Measures of Disease Occurrence
Sample Size and Power Part II
Coiffier B et al. Proc ASH 2011;Abstract 265.
THE LANCET Oncology Volume 19, No. 1, p27–39, January 2018
Comparisons of Modeling Methods on Longitudinal and Survival Data: Identifying Use of Repeat Biomarker Measurements to Predict Time-to-Event Outcome in.
Use of Piecewise Weighted Log-Rank Test for Trials with Delayed Effect
Kaplan-Meier survival curves and the log rank test
Planning and analyzing clinical trials with competing risks: Recommendations for choosing appropriate statistical methodology Presented by Misun Yu.
Presentation transcript:

Interpretation of effect estimates in competing risks survival models: A simulated analysis of organ-specific progression-free survival in a randomised phase III cancer trial By: Pradeep Virdee, Peter Dutton, Sharon Love, Harpreet Wasan, Ricky A Sharma, Joanna Moschandreas The talk is a demonstration, a demonstration encouraging the reporting of all estimates from CR models. We did this in the context of a simulated study https://www.csm.ox.ac.uk/

Motivation Often papers only report certain estimates Reporting all estimates from competing risk (CR) models should be encouraged I will be doing a CR analysis in the near future and as part of my research to understand how CRs work, I noticed that papers often only report certain estimates (event of interest) One of the reasons all estimates should be reported is because it facilitates a better interpretation of the results. Also, because there is an interdependence between event-estimates but these papers are often full of equations and can get pretty confusing. This is what the presentation is about – encouraging the reporting of all CR-model estimates https://www.csm.ox.ac.uk/ 2

Contents The study design What are competing risks The types of competing risk models Interpretation of estimates from the CR models https://www.csm.ox.ac.uk/ 3

Contents The study design What are competing risks The types of competing risk models Interpretation of estimates from the CR models https://www.csm.ox.ac.uk/ 4

The Study Design A randomised, phase III cancer trial (n≈1,000) Patients have colorectal cancer with liver metastases Intervention is a liver-targeted therapy: - Experimental: Liver-targeted therapy + Chemotherapy - Control: Chemotherapy Outcome is (first site) liver-specific progression-free survival (PFS): - Hepatic progression - Extra-hepatic progression - Death The idea of this presentation came out of a clinical trial that I work on, so we wanted to keep things in context and use simulated data (with parameters from other published studies) instead of actual data for the demonstration. Because this is a liver-targeted treatment, we are interested in modelling PFS within the liver. But not just “all-round” liver-specific PFS, we are particular interested in the first progressions that occur within the liver. So in other words, a patient can have their first site of progression anywhere, but we are particularly interested in those that occur within the liver before anywhere else in the body because this is a liver-targeted treatment. https://www.csm.ox.ac.uk/ 5

Contents The study design What are competing risks The types of competing risk models Interpretation of estimates from the CR models https://www.csm.ox.ac.uk/ 6

What are Competing Risks? In survival analyses, a competing risk (CR) is a failure event that precludes the event of interest[1]. [1] Jason P. Fine, Robert J. Gray; A Proportional Hazards Model for the Subdistribution of a Competing Risk; Journal of the American Statistical Association 1999; Vol. 94, No. 446, page 496- 509 https://www.csm.ox.ac.uk/ 7

What are Competing Risks? In survival analyses, a competing risk (CR) is a failure event that precludes the event of interest[1]. Time from randomisation [1] Jason P. Fine, Robert J. Gray; A Proportional Hazards Model for the Subdistribution of a Competing Risk; Journal of the American Statistical Association 1999; Vol. 94, No. 446, page 496- 509 https://www.csm.ox.ac.uk/ 8

What are Competing Risks? In survival analyses, a competing risk (CR) is a failure event that precludes the event of interest[1]. Hepatic Progression Time from randomisation [1] Jason P. Fine, Robert J. Gray; A Proportional Hazards Model for the Subdistribution of a Competing Risk; Journal of the American Statistical Association 1999; Vol. 94, No. 446, page 496- 509 https://www.csm.ox.ac.uk/ 9

Extra-Hepatic Progression What are Competing Risks? In survival analyses, a competing risk (CR) is a failure event that precludes the event of interest[1]. Extra-Hepatic Progression Hepatic Progression Time from randomisation [1] Jason P. Fine, Robert J. Gray; A Proportional Hazards Model for the Subdistribution of a Competing Risk; Journal of the American Statistical Association 1999; Vol. 94, No. 446, page 496- 509 https://www.csm.ox.ac.uk/ 10

Extra-Hepatic Progression What are Competing Risks? In survival analyses, a competing risk (CR) is a failure event that precludes the event of interest[1]. Extra-Hepatic Progression Hepatic Progression Time from randomisation [1] Jason P. Fine, Robert J. Gray; A Proportional Hazards Model for the Subdistribution of a Competing Risk; Journal of the American Statistical Association 1999; Vol. 94, No. 446, page 496- 509 https://www.csm.ox.ac.uk/ 11

What are Competing Risks? In survival analyses, a competing risk (CR) is a failure event that precludes the event of interest[1]. Hepatic Progression Death Time from randomisation [1] Jason P. Fine, Robert J. Gray; A Proportional Hazards Model for the Subdistribution of a Competing Risk; Journal of the American Statistical Association 1999; Vol. 94, No. 446, page 496- 509 https://www.csm.ox.ac.uk/ 12

What are Competing Risks? In survival analyses, a competing risk (CR) is a failure event that precludes the event of interest[1]. Hepatic Progression Death Time from randomisation [1] Jason P. Fine, Robert J. Gray; A Proportional Hazards Model for the Subdistribution of a Competing Risk; Journal of the American Statistical Association 1999; Vol. 94, No. 446, page 496- 509 https://www.csm.ox.ac.uk/ 13

Extra-Hepatic Progression What are Competing Risks? In survival analyses, a competing risk (CR) is a failure event that precludes the event of interest[1]. Extra-Hepatic Progression Hepatic Progression Death Time from randomisation [1] Jason P. Fine, Robert J. Gray; A Proportional Hazards Model for the Subdistribution of a Competing Risk; Journal of the American Statistical Association 1999; Vol. 94, No. 446, page 496- 509 https://www.csm.ox.ac.uk/ 14

Extra-Hepatic Progression What are Competing Risks? In survival analyses, a competing risk (CR) is a failure event that precludes the event of interest[1]. Extra-Hepatic Progression Hepatic Progression For the purposes of this presentation, we are treating the two events as one composite, competing event. Death Time from randomisation [1] Jason P. Fine, Robert J. Gray; A Proportional Hazards Model for the Subdistribution of a Competing Risk; Journal of the American Statistical Association 1999; Vol. 94, No. 446, page 496- 509 https://www.csm.ox.ac.uk/ 15

Contents The study design What are competing risks The types of competing risk models Interpretation of estimates from the CR models https://www.csm.ox.ac.uk/ 16

Types of Competing Risk Models CRs are analysed using either cause-specific or subdistribution (Fine & Grey cumulative incidence) proportional hazards models. Cause-specific: models the rate of occurrence of a specific event in subjects who have not yet experienced any of the events. Subdistribution: models the risk of experiencing a specific event in subjects who have not yet experienced the same event type. There is a cause-specific and subdistribution hazard function for each event[2]. In other words, there is a HR for each event from the cause-specific model and also the subdistribution model. [2] Peter C. Austin, Douglas S. Lee, Jason P. Fine; Introduction to the Analysis of Survival Data in the Presence of Competing Risks; Circulation 2016; 133: 601–609 https://www.csm.ox.ac.uk/ 17

Contents The study design What are competing risks The types of competing risk models Interpretation of estimates from the CR models https://www.csm.ox.ac.uk/ 18

The Simulations Analyses in R 3.3.1 using “cmprisk” package 10,000 simulations with median used for estimates Event times generated using an exponential distribution – fixed 0.7 treatment hazard ratio (HR) for the event of interest Two events in the models (hepatic progression and extra-hepatic progression/death). Assumed no censoring in the study. Interpretation using 3 scenarios (HR>1, =1, <1 for the competing event) 0.7 used due to previous studies No censoring because the patients in this study have colorectal cancer with mets, so they are very ill and recent studies show that they are likely to all experience an event. Multiple competing events can be added in the models. https://www.csm.ox.ac.uk/ 19

Liver-targeted therapy What is Done… Only the results for the event of interest are often reported Subdistributon HR (95% CI) Factor Hepatic Progression Liver-targeted therapy 0.71 (0.61, 0.83) Arm=Experimental Arm=Control Having the liver-targeted therapy significantly decreases the probability of hep prog as first site of prog (SHR: 0.71). This isn’t enough because there are competing risks here. There are events that make it impossible for patients to experience the event, and this interpretation doesn’t take that into account. https://www.csm.ox.ac.uk/ 20

Extra-Hepatic Progression/Death What Should be Done… All event-results should be reported Subdistributon HR (95% CI) Cause-Specific Scenario Factor Hepatic Progression Extra-Hepatic Progression/Death Liver-targeted therapy 0.71 (0.61, 0.83) 1.55 (1.25, 1.92) 0.70 (0.60, 0.82) 1.29 (1.04, 1.59) HR > 1 Arm=Experimental Arm=Control (months) Having the liver-targeted therapy significantly decreases the risk of hep prog as first site of prog (SHR: 0.71), whereas it significantly risk that of the competing event (SHR: 1.55). Consistently to the SHR, the addition of liver-targeted therapy significantly decreases the rate of hep prog (CSHR: 0.72), whereas it significantly increases the rate of the competing event (CSHR: 1.29). You can now see that the rate of the competing event is increasing which could be why the rate of the event of interest is decreasing, i.e. as time goes on, more patients are experiencing the competing event so fewer are experiencing the event of interest. A lower rate of hep prog as first site of prog, together with a higher rate of the competing event, means that of those patients who have liver-targeted therapy, fewer will have hep progs by the end of the trial. As both the subdistribution and cause-specific analyses are consistent with each other, literature says that we can interpret the liver-targeted therapy effect on the cumulative incidence of hep-prog as an actual effect and not as an indirect effect of the competing event. https://www.csm.ox.ac.uk/ 21

Extra-Hepatic Progression/Death Interpreting Competing Risk Estimates Subdistributon HR (95% CI) Cause-Specific Scenario Factor Hepatic Progression Extra-Hepatic Progression/Death Liver-targeted therapy 0.77 (0.65, 0.90) 1.29 (1.05, 1.58) 0.70 (0.60, 0.82) 1.00 (0.82, 1.23) HR = 1 Arm=Experimental Arm=Control (months) https://www.csm.ox.ac.uk/ 25

Extra-Hepatic Progression/Death Interpreting Competing Risk Estimates Subdistributon HR (95% CI) Cause-Specific Scenario Factor Hepatic Progression Extra-Hepatic Progression/Death Liver-targeted therapy 0.82 (0.73, 0.87) 1.07 (0.87, 1.32) 0.70 (0.60, 0.82) 0.80 (0.65, 0.99) HR < 1 Arm=Experimental Arm=Control (months) A reminder that the CS-HR is fixed to 0.7 for the reasons previously stated. In essence, by fixing the number of patients who have experienced the event of interest and the competing event in the control group, the CI and rate don’t change cause-specific hep prog HR to 0.7, we have fixed the CIF for this arm. You will notice that the curve for this arm (green arm) doesn’t change. By varying ONLY the cause-specific competing event parameters, the estimates for the cumulative incidence, subdistribution models still change. This is because there is also an interrelation between the two types of models as well. So in addition to reporting the SHR for the event of interest, which is what is commonly done, we should be reporting the HRs for each competing event as well and we should be reporting these from both cause-specific and subdistribution models. By doing so, we can better understand why we have these estimates and what they really mean for clinical practice. https://www.csm.ox.ac.uk/ 26

Extra-Hepatic Progression/Death Interpreting Competing Risk Estimates Subdistributon HR (95% CI) Cause-Specific Scenario Factor Hepatic Progression Extra-Hepatic Progression/Death Liver-targeted therapy 0.71 (0.61, 0.83) 1.55 (1.25, 1.92) 0.70 (0.60, 0.82) 1.29 (1.04, 1.59) HR > 1 0.77 (0.65, 0.90) 1.29 (1.05, 1.58) 1.00 (0.82, 1.23) HR = 1 0.82 (0.73, 0.87) 1.07 (0.87, 1.32) 0.80 (0.65, 0.99) HR < 1 Scenario 1: interpretation straightforward Scenario 2 & 3: complicated interpretation: - Competing event: differences in direction of rate and risk - “This discrepancy has been identified as a major difficulty in the interpretation of competing risks and interferes with a traditional understanding of disease”[3] A reminder that the CS-HR is fixed to 0.7 for the reasons previously stated. The way we did this was by fixing the CS-hazard rate in each treatment group. Additionally, we fixed the number of patients who have experienced the competing event in the control group and modified the number of competing events in the experimental group such that we could derive our 3 scenarios: one with HR<1, =1 and >1. You will notice that the curve for this arm (green arm) doesn’t change. So the only number we change between scenarios is the number experiencing the competing event in the experimental arm. We can see that although we change only the rate in the one group, it still changes the risk (S-HR’s), i.e. there is a relationship between rate and risk, which is another reason both should be reported. I am hoping that I have shown that you should report all these CR model-estimates. However, even by having everything, interpreting the estimates can still be rather complicated. Scenario 3 is quite straightforward: Having the liver-targeted therapy significantly decreases the risk of hep prog as first site of prog, whereas it significantly that of the competing event. Consistently to the SHR, the addition of liver-targeted therapy significantly decreases the rate of hep prog, whereas it significantly increases the rate of the competing event. So a lower rate of hep prog as first site of prog, together with a higher rate of the competing event, means that of those patients who have liver-targeted therapy, fewer will have hep progs by the end of the trial. You can see that the rate of the competing event is increasing which could be why the rate of the event of interest is decreasing, i.e. as time goes on, more patients are experiencing the competing event so fewer are experiencing the event of interest. Additionally, as both the subdistribution and cause-specific analyses are consistent with each other, literature says that we can interpret the liver-targeted therapy effect on the cumulative incidence of hep-prog as an actual effect and not as an indirect effect on the competing event. Scenario 1 and 2 Difficult to interpret. It may be very difficult to provide an explanation because the cumulative incidence for the events depend on the CSHs and the 1:1 relationship b/w rate & risk has been lost [3] Wolbers et al 2014; Competing risks analyses: objectives and approaches; doi:10.1093/eurheartj/ehu131 https://www.csm.ox.ac.uk/ 27

Conclusions Studies often report the CR model-estimates for the event of interest Alone, the interpretation cannot account for CRs. To better interpret results, all event-HRs should be reported A more complete picture is better for clinical practice https://www.csm.ox.ac.uk/ 28

Meta-Analysing Competing Risk HRs Statistical feasibility of meta-analysing HRs derived from CR models and their interpretation Lack of literature on this Often, only the event of interest is reported Even with all available information, is it statistically feasible? Future work: investigate the interdependence and meta-analysis In essence, what you would be doing is taking each (interrelated) term of the equation, separating and then manipulating them (meta-analysing) but you can’t put the equation back together again because of the underlying mathematical interrelationship between the terms. So how would you interpret the HRs? https://www.csm.ox.ac.uk/ 29