1
|
Rippin G, Salmasi S, Sanz H, Largent J. Core Concepts in Pharmacoepidemiology: Time-To-Event Analysis Approaches in Pharmacoepidemiology. Pharmacoepidemiol Drug Saf 2024; 33:e5886. [PMID: 39444098 DOI: 10.1002/pds.5886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 07/11/2024] [Accepted: 07/19/2024] [Indexed: 10/25/2024]
Abstract
AIM This article provides an overview of time-to-event (TTE) analysis in pharmacoepidemiology. MATERIALS & METHODS The key concept of censoring is reviewed, including right-, left-, interval- and informative censoring. Simple descriptive statistics are explained, including the nonparametric estimation of the TTE distribution as per Kaplan-Meier method, as well as more complex TTE regression approaches, including the parametric Accelerated Failure Time (AFT) model and the semi-parametric Cox Proportional Hazards and Restricted Mean Survival Time (RMST) models. Additional approaches and various TTE model extensions are presented as well. Finally, causal inference for TTE outcomes is addressed. RESULTS A thorough review of the available concepts and methods outlines the immense variety of available and useful TTE models. DISCUSSION There may be underused TTE concepts and methods, which are highlighted to raise awareness for researchers who aim to apply the most appropriate TTE approach for their study. CONCLUSION This paper constitutes a modern summary of TTE analysis concepts and methods. A curated list of references is provided.
Collapse
Affiliation(s)
- Gerd Rippin
- Statistical Services, IQVIA Commercial GmbH and Co OHG, Frankfurt, Germany
| | | | - Héctor Sanz
- Statistical Services, IQVIA Espana Barcelona, Barcelona, Spain
| | - Joan Largent
- Epidemiology, IQVIA Deerfield, Deerfield, Illinois, USA
| |
Collapse
|
2
|
Chen R, Wang H. Time-to-Event Endpoints in Imaging Biomarker Studies. J Magn Reson Imaging 2024. [PMID: 38739014 DOI: 10.1002/jmri.29446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 05/01/2024] [Accepted: 05/01/2024] [Indexed: 05/14/2024] Open
Abstract
Time-to-event endpoints are widely used as measures of patients' well-being and indicators of prognosis. In imaging-based biomarker studies, there are increasingly more studies that focus on examining imaging biomarkers' prognostic or predictive utilities on those endpoints, whether in a trial or an observational study setting. In this educational review article, we briefly introduce some basic concepts of time-to-event endpoints and point out potential pitfalls in the context of imaging biomarker research in hope of improving radiologists' understanding of related subjects. Besides, we have included some review and discussions on the benefits of using time-to-event endpoints and considerations on selecting overall survival or progression-free survival for primary analysis. LEVEL OF EVIDENCE: 5 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Ruizhe Chen
- The Sidney Kimmel Comprehensive Cancer Center, Division of Quantitative Sciences, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hao Wang
- The Sidney Kimmel Comprehensive Cancer Center, Division of Quantitative Sciences, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
3
|
Siu DHW, Lin FPY, Cho D, Lord SJ, Heller GZ, Simes RJ, Lee CK. Framework for the Use of External Controls to Evaluate Treatment Outcomes in Precision Oncology Trials. JCO Precis Oncol 2024; 8:e2300317. [PMID: 38190581 DOI: 10.1200/po.23.00317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/03/2023] [Accepted: 10/13/2023] [Indexed: 01/10/2024] Open
Abstract
Advances in genomics have enabled anticancer therapies to be tailored to target specific genomic alterations. Single-arm trials (SATs), including those incorporated within umbrella, basket, and platform trials, are widely adopted when it is not feasible to conduct randomized controlled trials in rare biomarker-defined subpopulations. External controls (ECs), defined as control arm data derived outside the clinical trial, have gained renewed interest as a strategy to supplement evidence generated from SATs to allow comparative analysis. There are increasing examples demonstrating the application of EC in precision oncology trials. The prospective application of EC in conducting comparative studies is associated with distinct methodological challenges, the specific considerations for EC use in biomarker-defined subpopulations have not been adequately discussed, and a formal framework is yet to be established. In this review, we present a framework for conducting a prospective comparative analysis using EC. Key steps are (1) defining the purpose of using EC to address the study question, (2) determining if the external data are fit for purpose, (3) developing a transparent study protocol and a statistical analysis plan, and (iv) interpreting results and drawing conclusions on the basis of a prespecified hypothesis. We specify the considerations required for the biomarker-defined subpopulations, which include (1) specifying the comparator and biomarker status of the comparator group, (2) defining lines of treatment, (3) assessment of the biomarker testing panels used, and (4) assessment of cohort stratification in tumor-agnostic studies. We further discuss novel clinical trial designs and statistical techniques leveraging EC to propose future directions to advance evidence generation and facilitate drug development in precision oncology.
Collapse
Affiliation(s)
- Derrick H W Siu
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
- Department of Medical Oncology, Illawarra Cancer Care Centre, Wollongong, NSW, Australia
| | - Frank P Y Lin
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- School of Clinical Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Doah Cho
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Sarah J Lord
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
- School of Medicine, University of Notre Dame, Sydney, NSW, Australia
| | - Gillian Z Heller
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
- Mathematics and Statistics, Macquarie University, Macquarie Park, NSW, Australia
| | - R John Simes
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Chee Khoon Lee
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
- Cancer Care Centre, St George Hospital, Kogarah, NSW, Australia
| |
Collapse
|
4
|
Mhatre SK, Machado RJM, Ton TGN, Trinh H, Mazieres J, Rittmeyer A, Bretscher MT. Real-World Progression-Free Survival as an Endpoint in Lung Cancer: Replicating Atezolizumab and Docetaxel Arms of the OAK Trial Using Real-World Data. Clin Pharmacol Ther 2023; 114:1313-1322. [PMID: 37696652 DOI: 10.1002/cpt.3045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/23/2023] [Indexed: 09/13/2023]
Abstract
Evaluating cancer treatments in real-world data (RWD) requires informative endpoints. This study replicated the atezolizumab and docetaxel arms of the OAK trial using RWD and compared progression-free survival (PFS) outcomes derived from abstracted physician's notes in RWD (rwPFS) against PFS outcomes derived from the clinical trial PFS (ctPFS). Atezolizumab and docetaxel arms of the phase III OAK randomized controlled trial (RCT; NCT02008227) were replicated in a US nationwide real-world database using selected OAK inclusion/exclusion criteria and propensity score-based adjustment for baseline prognostic variables. Concordance of outcomes was assessed using Kaplan-Meier medians and hazard ratios (HRs). The RWD cohorts comprised 133 patients on atezolizumab and 479 patients on docetaxel. After adjustment, prognostic variables were balanced between RCT arms and corresponding RWD cohorts. The rwPFS and ctPFS outcomes showed better concordance for docetaxel (2.99 vs. 3.52 months; HR: 0.99, 95% confidence interval (CI): 0.85-1.15) than for atezolizumab (3.71 vs. 2.76 months; HR: 0.8, 95% CI: 0.61-1.02). Excluding events labeled "pseudo-progression" from both RWD and RCT improved concordance for atezolizumab (4.24 vs. 4.14 months; HR: 0.95, 95% CI: 0.70-1.25). These findings were robust across sensitivity analyses. Replicating RCTs using RWD and comparing outcomes can help characterize RWD endpoints. Similarity of results between rwPFS and ctPFS at the cohort level may depend on drug category, highlighting the need for further studies to verify and understand when the corresponding outcomes can be compared, including within the same patient.
Collapse
Affiliation(s)
| | | | - Thanh G N Ton
- Genentech, Inc., South San Francisco, California, USA
| | - Huong Trinh
- Genentech, Inc., South San Francisco, California, USA
| | - Julien Mazieres
- Centre Hospitalier Universitaire, Université Paul Sabatier, Toulouse, France
| | - Achim Rittmeyer
- Department of Thoracic Oncology, Lungenfachklinik Immenhausen, Immenhausen, Germany
| | | |
Collapse
|
5
|
Amorrortu R, Garcia M, Zhao Y, El Naqa I, Balagurunathan Y, Chen DT, Thieu T, Schabath MB, Rollison DE. Overview of approaches to estimate real-world disease progression in lung cancer. JNCI Cancer Spectr 2023; 7:pkad074. [PMID: 37738580 PMCID: PMC10637832 DOI: 10.1093/jncics/pkad074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/28/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023] Open
Abstract
BACKGROUND Randomized clinical trials of novel treatments for solid tumors normally measure disease progression using the Response Evaluation Criteria in Solid Tumors. However, novel, scalable approaches to estimate disease progression using real-world data are needed to advance cancer outcomes research. The purpose of this narrative review is to summarize examples from the existing literature on approaches to estimate real-world disease progression and their relative strengths and limitations, using lung cancer as a case study. METHODS A narrative literature review was conducted in PubMed to identify articles that used approaches to estimate real-world disease progression in lung cancer patients. Data abstracted included data source, approach used to estimate real-world progression, and comparison to a selected gold standard (if applicable). RESULTS A total of 40 articles were identified from 2008 to 2022. Five approaches to estimate real-world disease progression were identified including manual abstraction of medical records, natural language processing of clinical notes and/or radiology reports, treatment-based algorithms, changes in tumor volume, and delta radiomics-based approaches. The accuracy of these progression approaches were assessed using different methods, including correlations between real-world endpoints and overall survival for manual abstraction (Spearman rank ρ = 0.61-0.84) and area under the curve for natural language processing approaches (area under the curve = 0.86-0.96). CONCLUSIONS Real-world disease progression has been measured in several observational studies of lung cancer. However, comparing the accuracy of methods across studies is challenging, in part, because of the lack of a gold standard and the different methods used to evaluate accuracy. Concerted efforts are needed to define a gold standard and quality metrics for real-world data.
Collapse
Affiliation(s)
| | - Melany Garcia
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Yayi Zhao
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Issam El Naqa
- Department of Machine Learning, Moffitt Cancer Center, Tampa, FL, USA
| | | | - Dung-Tsa Chen
- Department of Biostatistics and Bionformatics, Moffitt Cancer Center, Tampa, FL, USA
| | - Thanh Thieu
- Department of Machine Learning, Moffitt Cancer Center, Tampa, FL, USA
| | - Matthew B Schabath
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Dana E Rollison
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
6
|
Ton TG, Pal N, Trinh H, Mahrus S, Bretscher MT, Machado RJ, Sadetsky N, Chaudhary N, Lu MW, Riely GJ. Replication of Overall Survival, Progression-Free Survival, and Overall Response in Chemotherapy Arms of Non-Small Cell Lung Cancer Trials Using Real-World Data. Clin Cancer Res 2022; 28:2844-2853. [PMID: 35511917 PMCID: PMC9355621 DOI: 10.1158/1078-0432.ccr-22-0471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/22/2022] [Accepted: 05/03/2022] [Indexed: 01/12/2023]
Abstract
PURPOSE The utility of real-world data (RWD) for use as external controls in drug development is informed by studies that replicate trial control arms for different endpoints. The purpose of this study was to replicate control arms from four non-small cell lung cancer (NSCLC) randomized controlled trials (RCT) to analyze overall survival (OS), progression-free survival (PFS), and overall response rate (ORR) using RWD. PATIENTS AND METHODS This study used RWD from a nationwide de-identified database and a clinico-genomic database to replicate OS, PFS, and ORR endpoints in the chemotherapy control arms of four first-line NSCLC RCTs evaluating atezolizumab [IMpower150-wild-type (WT), IMpower130-WT, IMpower131, and IMpower132]. Additional objectives were to develop a definition of real-world PFS (rwPFS) and to evaluate the real-world response rate (rwRR) endpoint. RESULTS Baseline demographic and clinical characteristics were balanced after application of propensity score weighting methods. For rwPFS and OS, RWD external controls were generally similar to their RCT control counterparts. Across all four trials, the hazard ratio (HR) point estimates comparing trial controls with external controls were closer to 1.0 for the PFS endpoint than for the OS endpoint. An exploratory assessment of rwRR in RWD revealed a slight but nonsignificant overestimation of RCT ORR, which was unconfounded by baseline characteristics. CONCLUSIONS RWD can be used to reasonably replicate the OS and PFS of chemotherapy control arms of first-line NSCLC RCTs. Additional studies can provide greater insight into the utility of RWD in drug development.
Collapse
Affiliation(s)
- Thanh G.N. Ton
- Genentech, Inc., South San Francisco, California
- Corresponding Author: Thanh G.N. Ton, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080. Phone: 206-375-9710; E-mail:
| | - Navdeep Pal
- Genentech, Inc., South San Francisco, California
| | - Huong Trinh
- Genentech, Inc., South San Francisco, California
| | - Sami Mahrus
- Genentech, Inc., South San Francisco, California
| | | | | | | | | | | | | |
Collapse
|
7
|
Bourla AB, Meropol NJ. Bridging the divide between clinical research and clinical care in oncology: An integrated real-world evidence generation platform. Digit Health 2021; 7:20552076211059975. [PMID: 34868623 PMCID: PMC8638071 DOI: 10.1177/20552076211059975] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 10/27/2021] [Indexed: 11/16/2022] Open
Abstract
Real world data (RWD) are data relating to patient health status and/or the delivery of health care routinely collected from a variety of sources; real-world evidence (RWE) generated by RWD analyses can become an important component of drug development programs and, potentially, regulatory decision-making. As a RWD source, electronic health records (EHRs) can now provide patient-level data at unparalleled depth and granularity. We propose a RWE generation framework that could maximize the synergy between RWD and prospective clinical trials by capitalizing on an emerging data curation infrastructure that may be applied to both retrospective and prospective research. In this platform, centralized data collection and monitoring could be enabled via routine EHR use, and seamlessly integrated with select intentional data capture during prospective study periods. By bridging the divide between routine care and clinical research, this integrated platform aggregates retrospective and prospective data, collected both routinely and intentionally. This approach makes clinical trial participation more available to patients, increasing the potential depth of data, representativeness and efficiency of clinical research.
Collapse
|