1
|
Berlanga P, Aerts I, Corradini N, Ndounga-Diakou LA, Entz-Werle N, Ducassou S, André N, Sevrin F, Chastagner P, Puiseux C, Cleirec M, Plantaz D, De Carli E, Gambart M, Khanfar C, Thouvenin S, Petit A, Klein S, Briandet C, Millot F, Pluchart C, Reguerre Y, Schneider P, Serre J, Halfon-Domenech C, Carausu L, Piguet C, Saumet L, Benadiba J, Abbou S, Laghouati S, Geoerger B, Vassal G. Centralized Investigator Review of Radiological and Functional Imaging Reports in Real-World Oncology Studies: The SACHA-France Experience. Pediatr Blood Cancer 2025; 72:e31449. [PMID: 39558828 DOI: 10.1002/pbc.31449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/20/2024]
Abstract
SACHA-France (NCT04477681) is a prospective real-world study that collects clinical safety and efficacy data of novel anticancer therapies prescribed off-label or on compassionate use to patients <25 years. From March 2020 until February 2024, 640 patients with solid tumors or lymphomas were included, with 176 (28%) reported objective tumor responses. Centralized medical monitoring of local radiological/functional imaging reports by the SACHA coordinating investigator led to response modification in 45 out of 176 cases (26%), highlighting the relevance of the medical review of study data. We suggest this pragmatic approach for improving clinical trial data when centralized radiological review is not performed.
Collapse
Affiliation(s)
- Pablo Berlanga
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Isabelle Aerts
- SIREDO Oncology Center (Care, Innovation and Research for Children and AYA with Cancer); RTOP (Recherche Translationelle en Oncologie Pediatrique), U830 INSERM, Institut Curie, PSL Research University, Paris, France
| | - Nadège Corradini
- Department of Pediatric Oncology, Institute for Paediatric Haematology and Oncology, Léon Bérard Center, Lyon, France
| | - Lee Aymar Ndounga-Diakou
- Pharmacovigilance Unit, Clinical Research Direction, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Natacha Entz-Werle
- Pediatric Onco-Hematology Unit, University Hospital of Strasbourg, Strasbourg, France
| | - Stéphane Ducassou
- Paediatric Haematology-Oncology Department, Centre Hospitalier Universitaire (CHU), Bordeaux, France
| | - Nicolas André
- Department of Pediatric Hematology & Oncology, Hôpital de La Timone, AP-HM; REMAP4KIDS, UMR INSERM 1068, CNRS UMR 7258, Aix Marseille Université U105, Cancer Research Center (CRCM), Marseille, France
| | - François Sevrin
- Department of Pediatric Oncology, Oscar Lambret Cancer Center, Lille, France
| | - Pascal Chastagner
- Department of Pediatric Hematology-Oncology, Children's Hospital of Brabois, Vandoeuvre Les Nancy, France
| | - Chloe Puiseux
- Department of Pediatric Hemato-Oncology, University Hospital of Rennes, Rennes, France
| | - Morgane Cleirec
- Pediatric Immuno-Hemato-Oncology Unit, CHU Nantes, Nantes, France
| | - Dominique Plantaz
- Department of Pediatric Onco-Immuno-Hematology, Grenoble Alpes University Hospital, Grenoble, France
| | - Emilie De Carli
- Department of Pediatric Oncology, University Hospital, Angers, France
| | - Marion Gambart
- Department of Pediatric Oncology, Toulouse University Hospital, Toulouse, France
| | - Camille Khanfar
- Department of Pediatric Oncology, CHU Amiens Picardie, Amiens, France
| | - Sandrine Thouvenin
- Department of Pediatric Hematology-Oncology, University Hospital St Etienne, St Etienne, France
| | - Arnaud Petit
- Department of Pediatric Hematology and Oncology, Hôpital Armand Trousseau, Paris, France
| | - Sébastien Klein
- Pediatric Oncology and Hematology, CHU Jean-Minjoz, Besançon, France
| | | | - Frédéric Millot
- Department of Paediatric Haematology and Oncology, Centre Hospitalo-Universitaire de Poitiers, Poitiers, France
| | - Claire Pluchart
- Department of Paediatric Haematology and Oncology, Centre Hospitalo-Universitaire de Reims, Reims, France
| | - Yves Reguerre
- Pediatric Oncology and Hematology Unit, CHU Saint Denis de la Réunion, Bellepierre, France
| | - Pascale Schneider
- Department of Pediatric Hematology and Oncology, Centre Hospitalo-Universitaire de Rouen, Rouen, France
| | - Jill Serre
- Department of Pediatric Hematology-Oncology, CHRU de Tours, Tours, France
| | | | - Liana Carausu
- Department of Pediatric Hematology-Oncology, University Hospital of Brest, Brest, France
| | | | - Laure Saumet
- Department of Pediatric Hematology-Oncology, University Hospital of Montpellier, Montpellier, France
| | - Joy Benadiba
- Department of Hemato-Oncology Pediatric, Nice University Hospital, Nice, France
| | - Samuel Abbou
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Salim Laghouati
- Pharmacovigilance Unit, Clinical Research Direction, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Birgit Geoerger
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
- INSERM U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Gilles Vassal
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
2
|
Saal J, Eckstein M, Ritter M, Brossart P, Luetkens J, Ellinger J, Grünwald V, Hölzel M, Klümper N. Dissection of Progressive Disease Patterns for a Modified Classification for Immunotherapy. JAMA Oncol 2025; 11:154-161. [PMID: 39724246 PMCID: PMC11843377 DOI: 10.1001/jamaoncol.2024.5672] [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: 06/12/2024] [Accepted: 09/18/2024] [Indexed: 12/28/2024]
Abstract
Importance Progressive disease (PD) in patients treated with immune checkpoint inhibitors (ICIs) varies widely in outcomes according to the Response Evaluation Criteria in Solid Tumors (RECIST), version 1.1. Efforts to modify RECIST for ICI treatment have not resolved the heterogeneity in PD patterns, posing a clinical challenge. Objective To develop and validate a modified PD classification based on PD patterns and evaluate its association with postprogression survival (PPOS) in patients treated with the programmed cell death protein ligand 1 antibody atezolizumab across various solid tumors. Design, Setting, and Participants This study analyzed data from 5 phase 3 trials (IMmotion151, IMvigor211, OAK, Impower133, and IMspire150) involving patients treated with atezolizumab for renal cell carcinoma (RCC), urothelial carcinoma, small cell lung cancer, non-small cell lung cancer, and melanoma. This post hoc analysis was conducted from March to September 2024. Exposure Treatment with atezolizumab. Main Outcomes and Measures The primary outcome was the association of PD patterns with PPOS. Seven PD patterns were identified based on the enlargement of target and nontarget lesions or new lesions and their combinations. Results A total of 1377 patients were analyzed across the 5 trials. In RCC, 7 PD patterns significantly affected prognosis. The 6-month PPOS probability ranged from 26% for progression in target and nontarget lesions plus new lesions to 90% for progression in either target or nontarget lesions alone. A modified PD classification was developed that categorized PD into 3 risk levels: low risk (progression of existing lesions), intermediate risk (new lesions without progression of existing lesions), and high risk (progression of existing lesions plus new lesions). This score was associated with PPOS in ICI-treated RCC, with hazard ratios of 0.23 (95% CI, 0.13-0.41; P < .001) and 0.39 (95% CI, 0.23-0.66; P < .001) for low-risk and intermediate-risk PD compared with high-risk PD, respectively. Validation in additional trials confirmed the score's applicability across various tumors. Conclusions and Relevance In this study, a survival score was developed based on PD patterns. The risk classification was associated with PPOS across various solid tumors treated with immunotherapy and may therefore enhance prognostication and clinical decision-making, potentially providing a valuable tool for treating patients with PD who are receiving immunotherapy.
Collapse
Affiliation(s)
- Jonas Saal
- Medical Clinic III for Oncology, Hematology, Immune-Oncology and Rheumatology, University Hospital Bonn, Bonn, Germany
- Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf, Germany
| | - Markus Eckstein
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Bavarian Center for Cancer Research, Erlangen, Germany
| | - Manuel Ritter
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf, Germany
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
| | - Peter Brossart
- Medical Clinic III for Oncology, Hematology, Immune-Oncology and Rheumatology, University Hospital Bonn, Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf, Germany
| | - Julian Luetkens
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Bonn, Germany
- Quantitative Imaging Lab Bonn, Bonn, Germany
| | - Jörg Ellinger
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf, Germany
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
| | - Viktor Grünwald
- Clinic for Internal Medicine (Tumor Research) and Clinic for Urology, Interdisciplinary Genitourinary Oncology at the West German Cancer Center, Essen University Hospital, Essen, Germany
| | - Michael Hölzel
- Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf, Germany
| | - Niklas Klümper
- Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf, Germany
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
| |
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
|
Castellanos EH, Wittmershaus BK, Chandwani S. Raising the Bar for Real-World Data in Oncology: Approaches to Quality Across Multiple Dimensions. JCO Clin Cancer Inform 2024; 8:e2300046. [PMID: 38241599 PMCID: PMC10807898 DOI: 10.1200/cci.23.00046] [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: 03/24/2023] [Revised: 09/15/2023] [Accepted: 10/17/2023] [Indexed: 01/21/2024] Open
Abstract
PURPOSE Electronic health record (EHR)-based real-world data (RWD) are integral to oncology research, and understanding fitness for use is critical for data users. Complexity of data sources and curation methods necessitate transparency into how quality is approached. We describe the application of data quality dimensions in curating EHR-derived oncology RWD. METHODS A targeted review was conducted to summarize data quality dimensions in frameworks published by the European Medicines Agency, The National Institute for Healthcare and Excellence, US Food and Drug Administration, Duke-Margolis Center for Health Policy, and Patient-Centered Outcomes Research Institute. We then characterized quality processes applied to curation of Flatiron Health RWD, which originate from EHRs of a nationwide network of academic and community cancer clinics, across the summarized quality dimensions. RESULTS The primary quality dimensions across frameworks were relevance (including subdimensions of availability, sufficiency, and representativeness) and reliability (including subdimensions of accuracy, completeness, provenance, and timeliness). Flatiron Health RWD quality processes were aligned to each dimension. Relevancy to broad or specific use cases is optimized through data set size and variable breadth and depth. Accuracy is addressed using validation approaches, such as comparison with external or internal reference standards or indirect benchmarking, and verification checks for conformance, consistency, and plausibility, selected on the basis of feasibility and criticality of the variable to the intended use case. Completeness is assessed against expected source documentation; provenance by recording data transformation, management procedures, and auditable metadata; and timeliness by setting refresh frequency to minimize data lags. CONCLUSION Development of high-quality, scaled, EHR-based RWD requires integration of systematic processes across the data lifecycle. Approaches to quality are optimized through knowledge of data sources, curation processes, and use case needs. By addressing quality dimensions from published frameworks, Flatiron Health RWD enable transparency in determining fitness for real-world evidence generation.
Collapse
|
5
|
Lerro CC, Bradley MC, Forshee RA, Rivera DR. The Bar Is High: Evaluating Fit-for-Use Oncology Real-World Data for Regulatory Decision Making. JCO Clin Cancer Inform 2024; 8:e2300261. [PMID: 38241598 PMCID: PMC10807892 DOI: 10.1200/cci.23.00261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 12/15/2023] [Indexed: 01/21/2024] Open
Affiliation(s)
- Catherine C. Lerro
- Oncology Center of Excellence, Office of the Commissioner, US Food and Drug Administration, Silver Spring, MD
| | - Marie C. Bradley
- Office of Medical Policy, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Richard A. Forshee
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Donna R. Rivera
- Oncology Center of Excellence, Office of the Commissioner, US Food and Drug Administration, Silver Spring, MD
| |
Collapse
|
6
|
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
|
7
|
Error in Author Byline Credential. JAMA Netw Open 2022; 5:e2221224. [PMID: 35797054 PMCID: PMC9264038 DOI: 10.1001/jamanetworkopen.2022.21224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|