1
|
Seto T, Nakane S, Ji L, Ueda Y, Sugano H, Takei N, Kobayashi M, Murayama A, Yamamoto N. Real-world safety and effectiveness of entrectinib in Japanese patients with ROS1 gene fusion-positive, unresectable, advanced/recurrent non-small cell lung cancer: Post-marketing surveillance. Lung Cancer 2025; 203:108478. [PMID: 40179540 DOI: 10.1016/j.lungcan.2025.108478] [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: 08/05/2024] [Revised: 02/13/2025] [Accepted: 03/01/2025] [Indexed: 04/05/2025]
Abstract
OBJECTIVES To evaluate the safety and effectiveness of entrectinib, an orally-administered potent multi-kinase inhibitor, for the treatment of proto-oncogene tyrosine-protein kinase-1 (ROS1) gene fusion-positive, unresectable, advanced/recurrent non-small cell lung cancer (NSCLC) in Japan. MATERIALS AND METHODS Patients with ROS1 gene fusion-positive, unresectable, advanced/recurrent NSCLC who initiated entrectinib therapy were enrolled in this all-case post marketing surveillance between February 21, 2020 and November 30, 2021. Outcomes were to identify the: (1) type and onset of initial cognitive disorder and ataxia during entrectinib therapy; (2) status of treatment and outcome of drug-related cognitive disorder and ataxia events; (3) incidence of other adverse drug reactions (ADRs) of safety concern: cognitive disorder and/or ataxia, cardiac disorder (excluding QT interval prolongation), QT interval prolongation, syncope, and interstitial lung disease; (4) incidence of serious adverse events (AEs) and ADRs; and (5) effectiveness. RESULTS Of the 276 patients who initiated entrectinib, 269 and 260 were included in the safety and effectiveness analysis sets, respectively. Cognitive disorder/ataxia was the most common ADR of safety concern, occurring in 72 patients (26.8 %). The median time to onset of initial cognitive disorder/ataxia symptoms was 2.0 days. Overall, entrectinib dose reduction, interruption, or discontinuation occurred in 9.7 %, 28.3 %, and 15.2 % of patients, respectively. Most ADRs of safety concern were manageable; 86.9 % of patients with ADRs were recovered/recovering. Serious AEs were reported in 42.8 % of patients. The overall response rate (ORR) was 38.8 % and median time to treatment failure was 6.4 months. ORR was 70.8 % versus 26.8 % to 34.7 % with entrectinib as first-line versus second- or later-line treatment, and 65.3 % versus 28.2 % in patients without versus with a history of tyrosine kinase inhibitor treatment. CONCLUSIONS Consistent with clinical trials, entrectinib is tolerable and effective in Japanese patients with ROS1 gene fusion-positive, unresectable, advanced/recurrent NSCLC. STUDY REGISTRATION UMIN Clinical Trials Registry (UMIN000046619).
Collapse
Affiliation(s)
| | - Sayuri Nakane
- Safety Science 2 Department, Chugai Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Lyu Ji
- Safety Science 2 Department, Chugai Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Yuya Ueda
- Safety Science 2 Department, Chugai Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Hiroshi Sugano
- Safety Science 2 Department, Chugai Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Nobuki Takei
- Safety Science 2 Department, Chugai Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Mizuki Kobayashi
- Safety Science 2 Department, Chugai Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Ayako Murayama
- Safety Science 2 Department, Chugai Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Noboru Yamamoto
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
2
|
Lin L, Lucassen MJJ, van der Noort V, Egberts TCG, Beijnen JH, Huitema ADR. The feasibility of using real world data as external control arms in oncology trials. Drug Discov Today 2025; 30:104324. [PMID: 40054765 DOI: 10.1016/j.drudis.2025.104324] [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: 12/04/2023] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 03/12/2025]
Abstract
Before real world data (RWD)-derived external control arms (ECAs) can be applied in clinical trials within the field of oncology, it must be determined whether these ECAs can function as appropriate and reliable control arms. This review provides an overview of studies in which RWD-derived ECAs were constructed and compared to a randomized clinical trial (RCT) arm. RWD-derived ECAs had similar survival outcomes as the RCT arm of comparison in six of the eight included studies. Therefore, when performing RCTs is not feasible, RWD-derived ECAs can act as control arms in single-arm clinical trials, provided that suitable RWD sources are used and suitable methodological decisions are made.
Collapse
Affiliation(s)
- Lishi Lin
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Merel J J Lucassen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Vincent van der Noort
- Department of Biometrics, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Toine C G Egberts
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands; Department of Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands; Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| |
Collapse
|
3
|
Daigl M, Abogunrin S, Castro F, McGough SF, Sturrup RH, Boersma C, Abrams KR. Advancing the role of real-world evidence in comparative effectiveness research. J Comp Eff Res 2024; 13:e240101. [PMID: 39392412 DOI: 10.57264/cer-2024-0101] [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] [Indexed: 10/12/2024] Open
Abstract
Aim: Comparative effectiveness research (CER) is essential for making informed decisions about drug access. It provides insights into the effectiveness and safety of new drugs compared with existing treatments, thereby guiding better healthcare decisions and ensuring that new therapies meet the real-world needs of patients and healthcare systems. Objective: To provide a tool that assists analysts and decision-makers in identifying the most suitable analytical approach for answering a CER question, given specific data availability contexts. Methods: A systematic literature review of the scientific literature was performed and existing regulatory and health technology assessment (HTA) guidance were evaluated to identify and compare recommendations and best practices. Based on this review a methods flowchart that synthesizes current practices and requirements was proposed. Results: The review did not find any papers that clearly identified the most appropriate analytical approach for answering CER questions under various conditions. Therefore, a methods flowchart was designed to inform analyst and decision makers choices starting from a well-defined scientific question. Conclusion: The proposed methods flowchart offers clear guidance on CER methodologies across a range of settings and research needs. It begins with a well-defined research question and considers multiple feasibility aspects related to CER. This tool aims to standardize methods, ensure rigorous and consistent research quality and promote a culture of evidence-based decision-making in healthcare.
Collapse
Affiliation(s)
- Monica Daigl
- Global Access F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Seye Abogunrin
- Global Access F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Felipe Castro
- Data Science, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Sarah F McGough
- Computational Sciences, Genentech Inc., South San Franscisco, CA 94080, USA
| | | | - Cornelis Boersma
- Health-Ecore, 3704 HE Zeist, The Netherlands
- Department of Health Sciences, University Medical Center Groningen, Groningen, 9700 AB, The Netherlands
- Department of Management Sciences, Open University, Heerlen, 6419 AT, The Netherlands
| | - Keith R Abrams
- Department of Statistics & Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
- Centre for Health Economics, University of York, York, YO10 5DD, UK
| |
Collapse
|
4
|
Nadal E, Rifi N, Kane S, Mbacke S, Starkman L, Suero B, Le H, Samjoo IA. Efficacy and safety of crizotinib in the treatment of advanced non-small cell lung cancer with ROS1 gene fusion: a systematic literature review and meta-analysis of real-world evidence. Lung Cancer 2024; 192:107816. [PMID: 38749072 DOI: 10.1016/j.lungcan.2024.107816] [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: 02/01/2024] [Revised: 04/22/2024] [Accepted: 05/06/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Crizotinib was approved to treat patients with advanced non-small cell lung cancer (aNSCLC) with ROS proto-oncogene 1 (ROS1) gene fusion in 2016. We conducted a systematic literature review to identify real-world evidence (RWE) studies and estimated the efficacy and safety of crizotinib using meta-analyses (MA) for objective response rate (ORR), real-world progression-free survival (PFS), and overall survival (OS). METHODS We searched MEDLINE®, Embase, and Cochrane CENTRAL from January 2016 to March 2023 using Ovid® for published single-arm or comparative RWE studies evaluating patients (N ≥ 20) receiving crizotinib monotherapy for aNSCLC with ROS1 gene fusion. Pooled estimates for ORR and grade 3/4 adverse events (AEs) were derived using the metafor package in R while pooled estimates for median real-world PFS (rwPFS) and OS were derived using reconstructed individual patient data from published Kaplan-Meier curves. The primary analysis included all studies regardless of crizotinib line of therapy; a subgroup analysis (SA) was conducted using studies evaluating patients receiving first-line crizotinib. RESULTS Fourteen studies met the eligibility criteria and were considered feasible for MA. For the primary analysis, the pooled ORR (N = 9 studies) was 70.6 % (95 % confidence interval [CI]: 57.0, 81.3), median rwPFS was 14.5 months (N = 11 studies), and OS was 40.2 months (N = 9 studies). In the SA, the pooled ORR (N = 4 studies) was 81.1 % (95 % CI: 76.1, 85.2) and the median rwPFS (N = 4 studies) and OS (N = 2 studies) were 18.1 and 60 months, respectively. All MAs were associated with significant heterogeneity (I2 > 25 %). Grade 3/4 AEs occurred in 18.7 % of patients (pooled estimate). CONCLUSION The results from this study are consistent with clinical trial data and, taken collectively, supports crizotinib as a safe and effective treatment across different lines of therapy in patients with ROS1 aNSCLC in the real-world setting.
Collapse
Affiliation(s)
- Ernest Nadal
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Avda Gran via, 199-203. L'Hospitalet, 08908, Barcelona, Spain; Preclinical and Experimental Research in Thoracic Tumors (PReTT) Group, OncoBell Program, IDIBELL, L'Hospitalet, Barcelona, Spain.
| | - Nada Rifi
- Pfizer, Inc., New York, New York, USA
| | | | | | | | | | - Hannah Le
- Pfizer, Inc., New York, New York, USA
| | | |
Collapse
|
5
|
Perri F, Fusco R, Sabbatino F, Fasano M, Ottaiano A, Cascella M, Marciano ML, Pontone M, Salzano G, Maiello ME, Montano M, Calogero E, D’Aniello R, Maiolino P, Ciardiello F, Zotta A, Alfieri S, Ionna F. Translational Insights in the Landscape of Salivary Gland Cancers: Ready for a New Era? Cancers (Basel) 2024; 16:970. [PMID: 38473330 PMCID: PMC10931369 DOI: 10.3390/cancers16050970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 02/15/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Salivary gland carcinomas (SGCs) are rare neoplasms, representing less than 10% of all head and neck tumors, but they are extremely heterogeneous from the histological point of view, their clinical behavior, and their genetics. The guidelines regarding their treatment include surgery in most cases, which can also play an important role in oligometastatic disease. Where surgery cannot be used, systemic therapy comes into play. Systemic therapy for many years has been represented by polychemotherapy, but recently, with the affirmation of translational research, it can also count on targeted therapy, at least in some subtypes of SGCs. Interestingly, in some SGC histotypes, predominant mutations have been identified, which in some cases behave as "driver mutations", namely mutations capable of governing the carcinogenesis process. Targeting these driver mutations may be an effective therapeutic strategy. Nonetheless, it is not always possible to have drugs suitable for targeting driver mutations-and targeting driver mutations is not always accompanied by a clinical benefit. In this review, we will analyze the main mutations predominant in the various histotypes of SGCs.
Collapse
Affiliation(s)
- Francesco Perri
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Napoli, Italy; (A.O.); (M.C.); (M.P.); (M.E.M.); (M.M.); (E.C.); (R.D.); (P.M.); (F.I.)
| | - Roberta Fusco
- Medical Oncology Devision, IGEA S.p.A., 80013 Naples, Italy;
| | - Francesco Sabbatino
- Medical Oncology Department, Università degli Studi di Salerno, Via Giovanni Paolo II, 132, 84084 Salerno, Italy;
| | - Morena Fasano
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, 80128 Naples, Italy; (M.F.); (F.C.); (A.Z.)
| | - Alessandro Ottaiano
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Napoli, Italy; (A.O.); (M.C.); (M.P.); (M.E.M.); (M.M.); (E.C.); (R.D.); (P.M.); (F.I.)
| | - Marco Cascella
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Napoli, Italy; (A.O.); (M.C.); (M.P.); (M.E.M.); (M.M.); (E.C.); (R.D.); (P.M.); (F.I.)
| | - Maria Luisa Marciano
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Napoli, Italy; (A.O.); (M.C.); (M.P.); (M.E.M.); (M.M.); (E.C.); (R.D.); (P.M.); (F.I.)
| | - Monica Pontone
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Napoli, Italy; (A.O.); (M.C.); (M.P.); (M.E.M.); (M.M.); (E.C.); (R.D.); (P.M.); (F.I.)
| | - Giovanni Salzano
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, 80131 Naples, Italy;
| | - Maria Elena Maiello
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Napoli, Italy; (A.O.); (M.C.); (M.P.); (M.E.M.); (M.M.); (E.C.); (R.D.); (P.M.); (F.I.)
| | - Massimo Montano
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Napoli, Italy; (A.O.); (M.C.); (M.P.); (M.E.M.); (M.M.); (E.C.); (R.D.); (P.M.); (F.I.)
| | - Ester Calogero
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Napoli, Italy; (A.O.); (M.C.); (M.P.); (M.E.M.); (M.M.); (E.C.); (R.D.); (P.M.); (F.I.)
| | - Roberta D’Aniello
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Napoli, Italy; (A.O.); (M.C.); (M.P.); (M.E.M.); (M.M.); (E.C.); (R.D.); (P.M.); (F.I.)
| | - Piera Maiolino
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Napoli, Italy; (A.O.); (M.C.); (M.P.); (M.E.M.); (M.M.); (E.C.); (R.D.); (P.M.); (F.I.)
| | - Fortunato Ciardiello
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, 80128 Naples, Italy; (M.F.); (F.C.); (A.Z.)
| | - Alessia Zotta
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, 80128 Naples, Italy; (M.F.); (F.C.); (A.Z.)
| | - Salvatore Alfieri
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Franco Ionna
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via Mariano Semmola, 80131 Napoli, Italy; (A.O.); (M.C.); (M.P.); (M.E.M.); (M.M.); (E.C.); (R.D.); (P.M.); (F.I.)
| |
Collapse
|
6
|
Jansen JP, Ragavan MV, Chen C, Douglas MP, Phillips KA. The Health Inequality Impact of Liquid Biopsy to Inform First-Line Treatment of Advanced Non-Small Cell Lung Cancer: A Distributional Cost-Effectiveness Analysis. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2023; 26:1697-1710. [PMID: 37741446 PMCID: PMC10859998 DOI: 10.1016/j.jval.2023.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 07/28/2023] [Accepted: 08/10/2023] [Indexed: 09/25/2023]
Abstract
OBJECTIVES To perform a distributional cost-effectiveness analysis of liquid biopsy (LB) followed by, if needed, tissue biopsy (TB) (LB-first strategy) relative to a TB-only strategy to inform first-line treatment of advanced non-small cell lung cancer (aNSCLC) from a US payer perspective by which we quantify the impact of LB-first on population health inequality according to race and ethnicity. METHODS With a health economic model, quality-adjusted life-years (QALYs) and costs per patient were estimated for each subgroup. Given the lifetime risk of aNSCLC, and assuming equally distributed opportunity costs, the incremental net health benefits of LB-first were calculated, which were used to estimate general population quality-adjusted life expectancy at birth (QALE) by race and ethnicity with and without LB-first. The degree of QALYs and QALE differences with the strategies was expressed with inequality indices. Their differences were defined as the inequality impact of LB-first. RESULTS LB-first resulted in an additional 0.21 (95% uncertainty interval: 0.07-0.39) QALYs among treated patients, with the greatest gain observed among Asian patients (0.31 QALYs [0.09-0.61]). LB-first resulted in an increase in relative inequality in QALYs among patients, but a minor decrease in relative inequality in QALE. CONCLUSIONS LB-first to inform first-line aNSCLC therapy can improve health outcomes. With current diagnostic performance, the benefit is the greatest among Asian patients, thereby potentially widening racial and ethnic differences in survival among patients with aNSCLC. Assuming equally distributed opportunity costs and access, LB-first does not worsen and, in fact, may reduce inequality in general population health according to race and ethnicity.
Collapse
Affiliation(s)
- Jeroen P Jansen
- Department of Clinical Pharmacy, UCSF Center for Translational and Policy Research on Precision Medicine (TRANSPERS), San Francisco, CA, USA; UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA; UCSF Philip R. Lee Institute for Health Policy, San Francisco, CA, USA.
| | - Meera V Ragavan
- Division of Hematology and Oncology, UCSF Department of Medicine, San Francisco, CA, USA
| | - Cheng Chen
- Department of Clinical Pharmacy, UCSF Center for Translational and Policy Research on Precision Medicine (TRANSPERS), San Francisco, CA, USA
| | - Michael P Douglas
- Department of Clinical Pharmacy, UCSF Center for Translational and Policy Research on Precision Medicine (TRANSPERS), San Francisco, CA, USA
| | - Kathryn A Phillips
- Department of Clinical Pharmacy, UCSF Center for Translational and Policy Research on Precision Medicine (TRANSPERS), San Francisco, CA, USA; UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA; UCSF Philip R. Lee Institute for Health Policy, San Francisco, CA, USA
| |
Collapse
|
7
|
Araghi M, Mannani R, Heidarnejad maleki A, Hamidi A, Rostami S, Safa SH, Faramarzi F, Khorasani S, Alimohammadi M, Tahmasebi S, Akhavan-Sigari R. Recent advances in non-small cell lung cancer targeted therapy; an update review. Cancer Cell Int 2023; 23:162. [PMID: 37568193 PMCID: PMC10416536 DOI: 10.1186/s12935-023-02990-y] [Citation(s) in RCA: 109] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 07/12/2023] [Indexed: 08/13/2023] Open
Abstract
Lung cancer continues to be the leading cause of cancer-related death worldwide. In the last decade, significant advancements in the diagnosis and treatment of lung cancer, particularly NSCLC, have been achieved with the help of molecular translational research. Among the hopeful breakthroughs in therapeutic approaches, advances in targeted therapy have brought the most successful outcomes in NSCLC treatment. In targeted therapy, antagonists target the specific genes, proteins, or the microenvironment of tumors supporting cancer growth and survival. Indeed, cancer can be managed by blocking the target genes related to tumor cell progression without causing noticeable damage to normal cells. Currently, efforts have been focused on improving the targeted therapy aspects regarding the encouraging outcomes in cancer treatment and the quality of life of patients. Treatment with targeted therapy for NSCLC is changing rapidly due to the pace of scientific research. Accordingly, this updated study aimed to discuss the tumor target antigens comprehensively and targeted therapy-related agents in NSCLC. The current study also summarized the available clinical trial studies for NSCLC patients.
Collapse
Affiliation(s)
- Mahmood Araghi
- Department of Pathology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Reza Mannani
- Vascular Surgeon, Department of Surgery, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | - Adel Hamidi
- Razi Vaccine and Serum Research Institute, Arak Branch, karaj, Iran
| | - Samaneh Rostami
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | - Fatemeh Faramarzi
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sahar Khorasani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Safa Tahmasebi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center, Tuebingen, Germany
- Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University Warsaw, Warsaw, Poland
| |
Collapse
|
8
|
Ten Berge DMHJ, Damhuis RAM, Aerts JGJV, Dingemans AMC. Real-world treatment patterns and survival of patients with ROS1 rearranged stage IV non-squamous NSCLC in the Netherlands. Lung Cancer 2023; 181:107253. [PMID: 37236088 DOI: 10.1016/j.lungcan.2023.107253] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/09/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023]
Abstract
INTRODUCTION Rearrangement of c-ros oncogene 1 (ROS1) is a rare gene alteration in patients with stage IV non-squamous non-small cell lung cancer (NSCLC). Molecular testing for ROS1 is recommended to enable primary treatment with tyrosine kinase inhibitors (TKI). Aim of this study was to describe real-world treatment patterns and survival for patients with ROS1 in the Netherlands. METHODS All non-squamous NSCLC stage IV patients, diagnosed 2015-2019, were identified from the population-based Netherlands Cancer Registry (N = 19,871). For patients with ROS1 rearrangements (ROS1+ ) who received first line TKI, additional information about progression and second-line treatment was retrieved by active follow-up. Overall survival (OS) and progression-free survival (PFS) were calculated using Kaplan-Meier estimators. RESULTS A total of 67 patients (0.43%) were diagnosed with a ROS1+ NSCLC. Systemic treatment was administered in 75% which was most often TKI (n = 34) followed by chemotherapy (n = 14). Two-year OS for patients receiving upfront TKI versus other systemic treatment was 53% (95% CI 35-68) and 50% (95% CI 25-71), respectively. For patients receiving TKI, median OS was 24.3 months. Survival was inferior in case of brain metastasis (BM) at diagnosis (5.2 months). One in five patients receiving TKI as a first line treatment had BM at diagnosis, of the remaining 22 another 9 developed BM during follow up. PFS was also inferior for patients with BM at diagnosis with a median PFS of 4.3 months versus 9.0 without BM. CONCLUSION In this real-world population of ROS1+ NSCLC patients, only half received primary treatment with TKI. Overall survival and PFS during TKI were disappointing, mainly related to brain metastasis. TKI treatment with agents that have intra-cranial activity may be beneficial in this patient population and our results confirm the importance of performing an MRI of the brain as part of the standard diagnostic work up in patients with ROS1+ NSCLC.
Collapse
Affiliation(s)
- Deirdre M H J Ten Berge
- Dept. of Radiology, ADRZ, 's-Gravenpolderseweg 114, 4462 RA Goes, the Netherlands; Dept. of Pulmonary Medicine, Erasmus MC Cancer Institute, Doctor Molewaterplein 40, 3015 GD Rotterdam, the Netherlands
| | - Ronald A M Damhuis
- Dept. of Research and Development, Netherlands Cancer Registry, Netherlands Comprehensive Cancer Organization (IKNL), Godebaldkwartier 419, 3511 DT Utrecht, the Netherlands
| | - Joachim G J V Aerts
- Dept. of Pulmonary Medicine, Erasmus MC Cancer Institute, Doctor Molewaterplein 40, 3015 GD Rotterdam, the Netherlands
| | - Anne-Marie C Dingemans
- Dept. of Pulmonary Medicine, Erasmus MC Cancer Institute, Doctor Molewaterplein 40, 3015 GD Rotterdam, the Netherlands.
| |
Collapse
|
9
|
Benedum CM, Sondhi A, Fidyk E, Cohen AB, Nemeth S, Adamson B, Estévez M, Bozkurt S. Replication of Real-World Evidence in Oncology Using Electronic Health Record Data Extracted by Machine Learning. Cancers (Basel) 2023; 15:1853. [PMID: 36980739 PMCID: PMC10046618 DOI: 10.3390/cancers15061853] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/07/2023] [Accepted: 03/14/2023] [Indexed: 03/22/2023] Open
Abstract
Meaningful real-world evidence (RWE) generation requires unstructured data found in electronic health records (EHRs) which are often missing from administrative claims; however, obtaining relevant data from unstructured EHR sources is resource-intensive. In response, researchers are using natural language processing (NLP) with machine learning (ML) techniques (i.e., ML extraction) to extract real-world data (RWD) at scale. This study assessed the quality and fitness-for-use of EHR-derived oncology data curated using NLP with ML as compared to the reference standard of expert abstraction. Using a sample of 186,313 patients with lung cancer from a nationwide EHR-derived de-identified database, we performed a series of replication analyses demonstrating some common analyses conducted in retrospective observational research with complex EHR-derived data to generate evidence. Eligible patients were selected into biomarker- and treatment-defined cohorts, first with expert-abstracted then with ML-extracted data. We utilized the biomarker- and treatment-defined cohorts to perform analyses related to biomarker-associated survival and treatment comparative effectiveness, respectively. Across all analyses, the results differed by less than 8% between the data curation methods, and similar conclusions were reached. These results highlight that high-performance ML-extracted variables trained on expert-abstracted data can achieve similar results as when using abstracted data, unlocking the ability to perform oncology research at scale.
Collapse
Affiliation(s)
- Corey M. Benedum
- Flatiron Health, Inc., 233 Spring Street, New York, NY 10003, USA; (C.M.B.); (A.S.); (E.F.); (A.B.C.); (S.N.); (B.A.); (S.B.)
| | - Arjun Sondhi
- Flatiron Health, Inc., 233 Spring Street, New York, NY 10003, USA; (C.M.B.); (A.S.); (E.F.); (A.B.C.); (S.N.); (B.A.); (S.B.)
| | - Erin Fidyk
- Flatiron Health, Inc., 233 Spring Street, New York, NY 10003, USA; (C.M.B.); (A.S.); (E.F.); (A.B.C.); (S.N.); (B.A.); (S.B.)
| | - Aaron B. Cohen
- Flatiron Health, Inc., 233 Spring Street, New York, NY 10003, USA; (C.M.B.); (A.S.); (E.F.); (A.B.C.); (S.N.); (B.A.); (S.B.)
- Department of Medicine, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Sheila Nemeth
- Flatiron Health, Inc., 233 Spring Street, New York, NY 10003, USA; (C.M.B.); (A.S.); (E.F.); (A.B.C.); (S.N.); (B.A.); (S.B.)
| | - Blythe Adamson
- Flatiron Health, Inc., 233 Spring Street, New York, NY 10003, USA; (C.M.B.); (A.S.); (E.F.); (A.B.C.); (S.N.); (B.A.); (S.B.)
- Comparative Health Outcomes, Policy and Economics (CHOICE) Institute, University of Washington, Seattle, WA 98195, USA
| | - Melissa Estévez
- Flatiron Health, Inc., 233 Spring Street, New York, NY 10003, USA; (C.M.B.); (A.S.); (E.F.); (A.B.C.); (S.N.); (B.A.); (S.B.)
| | - Selen Bozkurt
- Flatiron Health, Inc., 233 Spring Street, New York, NY 10003, USA; (C.M.B.); (A.S.); (E.F.); (A.B.C.); (S.N.); (B.A.); (S.B.)
| |
Collapse
|
10
|
Tyler LC, Le AT, Chen N, Nijmeh H, Bao L, Wilson TR, Chen D, Simmons B, Turner KM, Perusse D, Kasibhatla S, Christiansen J, Dudek AZ, Doebele RC. MET gene amplification is a mechanism of resistance to entrectinib in ROS1+ NSCLC. Thorac Cancer 2022; 13:3032-3041. [PMID: 36101520 PMCID: PMC9626307 DOI: 10.1111/1759-7714.14656] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND ROS1 tyrosine kinase inhibitors (TKIs) have demonstrated significant clinical benefit for ROS1+ NSCLC patients. However, TKI resistance inevitably develops through ROS1 kinase domain (KD) modification or another kinase driving bypass signaling. While multiple TKIs have been designed to target ROS1 KD mutations, less is known about bypass signaling in TKI-resistant ROS1+ lung cancers. METHODS Utilizing a primary, patient-derived TPM3-ROS1 cell line (CUTO28), we derived an entrectinib-resistant line (CUTO28-ER). We evaluated proliferation and signaling responses to TKIs, and utilized RNA sequencing, whole exome sequencing, and fluorescence in situ hybridization to detect transcriptional, mutational, and copy number alterations, respectively. We substantiated in vitro findings using a CD74-ROS1 NSCLC patient's tumor samples. Last, we analyzed circulating tumor DNA (ctDNA) from ROS1+ NSCLC patients in the STARTRK-2 entrectinib trial to determine the prevalence of MET amplification. RESULTS CUTO28-ER cells did not exhibit ROS1 KD mutations. MET TKIs inhibited proliferation and downstream signaling and MET transcription was elevated in CUTO28-ER cells. CUTO28-ER cells displayed extrachromosomal (ecDNA) MET amplification without MET activating mutations, exon 14 skipping, or fusions. The CD74-ROS1 patient samples illustrated MET amplification while receiving ROS1 TKI. Finally, two of 105 (1.9%) entrectinib-resistant ROS1+ NSCLC STARTRK-2 patients with ctDNA analysis at enrollment and disease progression displayed MET amplification. CONCLUSIONS Treatment with ROS1-selective inhibitors may lead to MET-mediated resistance. The discovery of ecDNA MET amplification is noteworthy, as ecDNA is associated with more aggressive cancers. Following progression on ROS1-selective inhibitors, MET gene testing and treatments targeting MET should be explored to overcome MET-driven resistance.
Collapse
Affiliation(s)
- Logan C. Tyler
- Department of Medicine—Division of Medical OncologyUniversity of Colorado—Anschutz Medical CampusAuroraColoradoUSA
| | - Anh T. Le
- Department of Medicine—Division of Medical OncologyUniversity of Colorado—Anschutz Medical CampusAuroraColoradoUSA
| | - Nan Chen
- Department of Medicine—Division of Medical OncologyUniversity of Colorado—Anschutz Medical CampusAuroraColoradoUSA
| | - Hala Nijmeh
- Department of PathologyUniversity of Colorado—Anschutz Medical CampusAuroraColoradoUSA
| | - Liming Bao
- Department of PathologyUniversity of Colorado—Anschutz Medical CampusAuroraColoradoUSA
| | | | - David Chen
- Genentech, Inc.South San FranciscoCaliforniaUSA
| | | | | | | | | | | | - Arkadiusz Z. Dudek
- HealthPartners Cancer Center at Regions HospitalSt. PaulMinnesotaUSA,Department of Medicine—Division of Hematology, Oncologyand Transplantation University of MinnesotaMinneapolisMinnesotaUSA
| | - Robert C. Doebele
- Department of Medicine—Division of Medical OncologyUniversity of Colorado—Anschutz Medical CampusAuroraColoradoUSA
| |
Collapse
|
11
|
O'Haire S, Degeling K, Franchini F, Tran B, Luen SJ, Gaff C, Smith K, Fox S, Desai J, IJzerman M. Comparing Survival Outcomes for Advanced Cancer Patients Who Received Complex Genomic Profiling Using a Synthetic Control Arm. Target Oncol 2022; 17:539-548. [PMID: 36063280 PMCID: PMC9512745 DOI: 10.1007/s11523-022-00910-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Complex genomic profiling (CGP) has transformed cancer treatment decision making, yet there is a lack of robust and quantifiable evidence for how utilisation of CGP improves patient outcomes. OBJECTIVE This study evaluated cohort level clinical effectiveness of CGP to improve overall survival (OS) in real-world advanced cancer patients using a registry-based matched control population. PATIENTS AND METHODS Two cohorts of advanced and refractory cancer patients were seen in consecutive series for early phase trial enrolment consideration. The first cohort (CGP group) accessed tumour profiling via a research study; while the second cohort that followed was not profiled. Overall survival between cohorts was compared using Kaplan-Meier curves and Cox proportional hazard models. Potential confounding was analysed and adjusted for using stabilised weights based on propensity scores. RESULTS Within the CGP group, 25 (17.6%) patients received treatment informed by CGP results and this subgroup had significantly improved survival compared with CGP patients in whom results did not impact their treatment (unadjusted HR = 0.44, (0.22-0.88), p = 0.02). However, when comparing the entire CGP cohort with the No CGP cohort, no significant survival benefit was evident with adjusted median OS for CGP of 13.5 months (9.2-17.0) compared with 11.0 (9.2-17.4) for No CGP (adjusted HR = 0.92, (0.65-1.30), p = 0.63). CONCLUSIONS This study utilised real-world data to simulate a control arm and quantify the clinical effectiveness of genomic testing. The magnitude of survival benefit for patients who had CGP result-led treatments was insufficient to drive an overall survival gain for the entire tested population. Translation of CGP into clinics requires strategies to ensure higher rates of tested patients obtain clinical benefit to deliver on the value proposition of CGP in an advanced cancer population.
Collapse
Affiliation(s)
- Sophie O'Haire
- Cancer Health Services Research, Centre for Cancer Research, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia. .,Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia. .,Peter MacCallum Cancer Centre, Melbourne, Australia.
| | - Koen Degeling
- Cancer Health Services Research, Centre for Cancer Research, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia.,Centre for Health Policy, School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| | - Fanny Franchini
- Cancer Health Services Research, Centre for Cancer Research, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia.,Centre for Health Policy, School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| | - Ben Tran
- Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia.,Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia.,Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Stephen J Luen
- Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia.,Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia.,Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Clara Gaff
- Melbourne Genomics Health Alliance, Melbourne, Australia.,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia.,Murdoch Children's Research Institute, Melbourne, Australia
| | - Kortnye Smith
- Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia.,Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Stephen Fox
- Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia.,Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Australia.,Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Jayesh Desai
- Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia.,Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia.,Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Maarten IJzerman
- Cancer Health Services Research, Centre for Cancer Research, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia.,Centre for Health Policy, School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
12
|
Tremblay G, Groff M, Iadeluca L, Daniele P, Wilner K, Wiltshire R, Bartolome L, Usari T, Cappelleri JC, Camidge DR. Effectiveness of crizotinib versus entrectinib in ROS1-positive non-small-cell lung cancer using clinical and real-world data. Future Oncol 2022; 18:2063-2074. [PMID: 35232230 DOI: 10.2217/fon-2021-1102] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aims: To compare clinical trial results for crizotinib and entrectinib in ROS1-positive non-small-cell lung cancer and compare clinical trial data and real-world outcomes for crizotinib. Patients & methods: We analyzed four phase I-II studies using a simulated treatment comparison (STC). A STC of clinical trial versus real-world evidence compared crizotinib clinical data to real-world outcomes. Results: Adjusted STC found nonsignificant trends favoring crizotinib over entrectinib: objective response rate, risk ratio = 1.04 (95% CI: 0.85-1.28); median duration of response, mean difference = 16.11 months (95% CI: -1.57- 33.69); median progression-free survival, mean difference = 3.99 months (95% CI: -6.27-14.25); 12-month overall survival, risk ratio = 1.01 (95% CI: 0.90-1.12). Nonsignificant differences were observed between the trial end point values and the real-world evidence for crizotinib. Conclusions: Crizotinib and entrectinib have comparable efficacy in ROS1-positive non-small-cell lung cancer.
Collapse
Affiliation(s)
- Gabriel Tremblay
- Cytel Inc. Health Economics & Outcomes Research (HEOR). 1050 Winter St no. 2700, Waltham, MA 02451, USA
| | - Michael Groff
- Cytel Inc. Health Economics & Outcomes Research (HEOR). 1050 Winter St no. 2700, Waltham, MA 02451, USA
| | - Laura Iadeluca
- Pfizer Inc. Health Economics & Outcomes Research (HEOR). 235 East 42nd Street NY, NY 10017, USA
| | - Patrick Daniele
- Cytel Inc. Health Economics & Outcomes Research (HEOR). 1050 Winter St no. 2700, Waltham, MA 02451, USA
| | - Keith Wilner
- Pfizer Inc. Health Economics & Outcomes Research (HEOR). 235 East 42nd Street NY, NY 10017, USA
| | - Robin Wiltshire
- Pfizer Inc. Health Economics & Outcomes Research (HEOR). 235 East 42nd Street NY, NY 10017, USA
| | - Lauren Bartolome
- Pfizer Inc. Health Economics & Outcomes Research (HEOR). 235 East 42nd Street NY, NY 10017, USA
| | - Tiziana Usari
- Pfizer Inc. Health Economics & Outcomes Research (HEOR). 235 East 42nd Street NY, NY 10017, USA
| | - Joseph C Cappelleri
- Pfizer Inc. Health Economics & Outcomes Research (HEOR). 235 East 42nd Street NY, NY 10017, USA
| | - D Ross Camidge
- University of Colorado Cancer Center. Thoracic Oncology Clinical and Clinical Research Programs. 1665 Aurora Court, Aurora, CO 80045, USA
| |
Collapse
|