1
|
Matsubara J, Mukai K, Kondo T, Yoshioka M, Kage H, Oda K, Ikeda S, Ebi H, Muro K, Kajiura S, Hayashi R, Ashida R, Kitano M, Muto M. First-Line Genomic Profiling in Previously Untreated Advanced Solid Tumors: 1-Year Follow-Up of the FIRST-Dx Study. Cancer Sci 2025. [PMID: 40217582 DOI: 10.1111/cas.70077] [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: 12/01/2024] [Revised: 03/11/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
The FIRST-Dx study prospectively evaluated the clinical utility of the comprehensive genomic profiling (CGP) test (FoundationOne CDx) in the first-line setting for patients with chemotherapy-naïve advanced solid tumors (gastrointestinal, biliary, pancreatic, lung, breast, gynecologic, melanoma) in six hospitals in Japan. Here, we report the results of the 1-year interim analysis of the follow-up study about the clinical benefits provided by the upfront CGP test. The primary endpoint was overall survival (OS), and secondary endpoints were the proportion of patients who actually received molecular-based recommended therapy (MBRT) determined by the molecular tumor board, best overall response rate (ORR) in each line of therapy, and progression-free survival (PFS) ratio (PFS on MBRT/PFS on the first-line therapy). Data from 172 patients with a median follow-up of 15.1 months (range: 0.1-21.5 months) were available. The median OS was not reached. Thirty-nine patients (22.7%) received MBRT during this follow-up period. ORR in first-line therapy was 56.3% in the MBRT group (n = 16) vs. 42.3% in the non-MBRT group (n = 137), and in the second-line was 26.3% in the MBRT group (n = 19) vs. 17.1% in the non-MBRT group (n = 82). Regarding the PFS ratio of second-line MBRT (n = 12), the median PFS ratio was 1.1, and four patients (33.3%) had a ratio ≥ 1.3, indicating that MBRT might be effective in changing the clinical outcome. The findings of this study imply that CGP testing before the standard of care for patients with advanced solid tumors could prove to be a clinically beneficial strategy for guiding subsequent precision anticancer treatments. Trial Registration: Japan Registry of Clinical Trials (jRCT) ID: jRCT1050220041.
Collapse
Affiliation(s)
- Junichi Matsubara
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Kumi Mukai
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Tomohiro Kondo
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Masahiro Yoshioka
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Hidenori Kage
- Department of Clinical Genomics, The University of Tokyo Hospital, Tokyo, Japan
| | - Katsutoshi Oda
- Department of Clinical Genomics, The University of Tokyo Hospital, Tokyo, Japan
| | - Sadakatsu Ikeda
- Department of Precision Cancer Medicine, Tokyo Medical and Dental University Hospital, Tokyo, Japan
| | - Hiromichi Ebi
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center, Nagoya, Japan
| | - Shinya Kajiura
- Department of Clinical Oncology, Toyama University Hospital, Toyama, Japan
| | - Ryuji Hayashi
- Department of Clinical Oncology, Toyama University Hospital, Toyama, Japan
| | - Reiko Ashida
- Second Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan
| | - Masayuki Kitano
- Second Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan
| | - Manabu Muto
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| |
Collapse
|
2
|
Trédan O, Pouessel D, Penel N, Chabaud S, Gomez-Roca C, Delord JP, Pannier D, Brahmi M, Fabbro M, Garcia ME, Larrieu-Ciron D, Ray-Coquard I, Viala M, Italiano A, Tosi D, Cassier P, Dufresne A, Attignon V, Boyault S, Treilleux I, Viari A, Pérol D, Blay JY. Broad versus limited gene panels to guide treatment in patients with advanced solid tumors: a randomized controlled trial. Nat Med 2025:10.1038/s41591-025-03613-x. [PMID: 40195451 DOI: 10.1038/s41591-025-03613-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 02/26/2025] [Indexed: 04/09/2025]
Abstract
Large genomic programs have contributed to improving drug development in cancer. To assess the potential benefit of using larger gene panels to guide molecular-based treatments, we conducted a multicenter randomized trial in patients with advanced and/or metastatic solid cancer. Molecular alterations were determined using either a panel of 324 cancer-related genes (Foundation OneCDX (F1CDX)) or a limited panel of 87 single-nucleotide/indel genes and genome-wide copy number variations (CTL) and reviewed by a molecular tumor board to identify molecular-based recommended therapies (MBRTs). Using paired data from both panels for each patient, the primary endpoint was the proportion of patients with an MBRT identified. Main secondary endpoints included the number of patients with at least one actionable alteration leading to MBRT identification, the number of patients with and without MBRTs initiated, progression-free survival, best overall response, duration of response and safety. Among the 741 patients screened, 45.7% had quality-checked tumor samples. MBRTs were identified with F1CDX in 175 (51.6%) patients and with CTL in 125 (36.9%) patients, translating to a significant increase of 14.8 percentage points (P < 0.001) with the more comprehensive gene panel versus the more limited panel, meeting the primary endpoint. However, no differences in clinical outcomes were observed in these patients with advanced and/or metastatic cancer in need of treatment beyond standard genomic alterations. These findings illustrate the potential for larger gene panels to increase the number of molecularly matched therapies. Larger studies are needed to assess the clinical benefit of expanded MBRTs. ClinicalTrials.gov registration: NCT03163732 .
Collapse
Affiliation(s)
- Olivier Trédan
- Centre Léon Bérard, Lyon, France.
- Cancer Research Center of Lyon, Lyon, France.
| | | | - Nicolas Penel
- Centre Oscar Lambret, Lille and Université de Lille ULR 2694, Lille, France
| | | | | | | | - Diane Pannier
- Centre Oscar Lambret, Lille and Université de Lille ULR 2694, Lille, France
| | | | - Michel Fabbro
- Institut de Cancérologie de Montpellier, Montpellier, France
| | | | | | | | - Marie Viala
- Institut de Cancérologie de Montpellier, Montpellier, France
| | | | - Diego Tosi
- Institut de Cancérologie de Montpellier, Montpellier, France
| | | | | | | | - Sandrine Boyault
- Centre Léon Bérard, Lyon, France
- Cancer Research Center of Lyon, Lyon, France
| | | | - Alain Viari
- Centre Léon Bérard, Lyon, France
- INRIA, Grenoble, France
| | | | - Jean Yves Blay
- Centre Léon Bérard, Lyon, France
- Cancer Research Center of Lyon, Lyon, France
- Université Claude Bernard Lyon I, Lyon, France
| |
Collapse
|
3
|
Huang CY, Huang WK, Yeh KY, Chang JWC, Lin YC, Chou WC. Integrating comprehensive genomic profiling in the management of oncology patients: applications and challenges in Taiwan. Biomed J 2025:100851. [PMID: 40185203 DOI: 10.1016/j.bj.2025.100851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 03/25/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025] Open
Abstract
Comprehensive genomic profiling (CGP) refers to the detailed genomic analysis of cancers for oncology patients. With the rapid development of next-generation sequencing (NGS) technologies, CGP has been widely applied to clinical practice and managing oncology patients. CGP can be performed on the tumor DNA and RNA, as well as non-tumor tissues (e.g., blood, pleural effusion, and ascites). In this article, we review the current evidence supporting the use of CGP in the management of oncology patients, both in real-world practice and the bridging to clinical trials. We also discuss the role of the molecular tumor board on the application of CGP in oncology patients. We provide an overview of the current scheme of CGP reimbursement in Taiwan and the precision oncology branch of the National Biobank Consortium of Taiwan. Finally, we discuss about the potential barriers and challenges of applying CGP in managing oncology patients and the future perspectives of CGP in precision oncology.
Collapse
Affiliation(s)
- Chen-Yang Huang
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan
| | - Wen-Kuan Huang
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kun-Yun Yeh
- College of Medicine, Chang Gung University, Taoyuan, Taiwan; Division of Hematology-Oncology, Department of Internal Medicine, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
| | - John Wen-Cheng Chang
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan
| | - Yung-Chang Lin
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan
| | - Wen-Chi Chou
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan.
| |
Collapse
|
4
|
Soliman N, Maqsood A, Connor AA. Role of genomics in liver transplantation for cholangiocarcinoma. Curr Opin Organ Transplant 2025; 30:158-170. [PMID: 39917813 DOI: 10.1097/mot.0000000000001209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarize the current knowledge of cholangiocarcinoma molecular biology and to suggest a framework for implementation of next-generation sequencing in all stages of liver transplantation. This is timely as recent guidelines recommend increased use of these technologies with promising results. RECENT FINDINGS The main themes covered here address germline and somatic genetic alterations recently discovered in cholangiocarcinoma, particularly those associated with prognosis and treatment responses, and nascent efforts to translate these into contemporary practice in the peri-liver transplantation period. SUMMARY Early efforts to translate molecular profiling to cholangiocarcinoma care demonstrate a growing number of potentially actionable alterations. Still lacking is a consensus on what biomarkers and technologies to adopt, at what scale and cost, and how to integrate them most effectively into care with the ambition of increasing the number of patients eligible for liver transplantation and improving their long-term outcomes.
Collapse
Affiliation(s)
- Nadine Soliman
- Department of Surgery
- J. C. Walter Jr. Transplant Center, Houston Methodist Hospital
- Houston Methodist Academic Institute
| | - Anaum Maqsood
- Department of Medicine
- Neill Cancer Center, Houston Methodist Hospital, Houston, Texas
| | - Ashton A Connor
- Department of Surgery
- J. C. Walter Jr. Transplant Center, Houston Methodist Hospital
- Houston Methodist Academic Institute
- Neill Cancer Center, Houston Methodist Hospital, Houston, Texas
- Department of Surgery, Weill Cornell Medicine, Cornell University, New York, New York, USA
| |
Collapse
|
5
|
Rizzo M, Pezzicoli G, Porta C, Povero M, Pradelli L, Sicari E, Barbiero VS, Porta C. The genomic landscape of metastatic clear-cell renal cell carcinoma and its prognostic value: a comprehensive analysis of a large real-world clinico-genomic database. ESMO Open 2025; 10:104294. [PMID: 39965361 PMCID: PMC11876921 DOI: 10.1016/j.esmoop.2025.104294] [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: 09/21/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Translating findings on the genomic landscape of metastatic clear-cell renal cell carcinoma (mccRCC) into clinical practice remains challenging. A better understanding of the molecular features of mccRCC could identify a prognostic and/or predictive role for ccRCC genomic alterations. PATIENTS AND METHODS In this real-world observational study based on the nationwide (US-based) de-identified Flatiron Health-Foundation Medicine, Inc. clinico-genomic database (FH-FMI-CGDB), we investigate the frequency and co-occurrence of genomic alterations in mccRCC patients and assess their prognostic role. Patients (n = 858) were adults diagnosed with mccRCC, with FH electronic health records between 2011 and 2022. RESULTS The top 10 mutated genes were VHL (73.9%), PBRM1 (42.4%), SETD2 (25.3%), CDKN2A (20.0%), BAP1 (16.4%), CDKN2B (16.0%), KDM5C (14.5%), TP53 (12.9%), PTEN (11.7%), and TERT (9.2%). Eight genes showed prognostic value: CDKN2A, CDKN2B, TP53, PTEN, NF2, PIK3CA, and MTAP were linked to worse prognosis, whereas PBRM1 was associated with better overall survival (OS). Two of the three identified gene clusters had prognostic value: cluster 1 (VHL, SETD2, PBRM1, KDM5C, NFE2L2) correlated with better OS [adjusted hazard ratio (aHR) 0.63, P < 0.001], whereas cluster 3 (CDKN2A, CDKN2B, BAP1, NF2, MTAP) correlated with shorter OS (aHR 1.36, P = 0.023). CONCLUSION We identified eight genes and two gene clusters with prognostic significance for mccRCC. Future research will explore the predictive value of gene clusters in various treatments.
Collapse
Affiliation(s)
- M Rizzo
- Division of Medical Oncology, AOU Consorziale Policlinico di Bari, Bari, Italy.
| | - G Pezzicoli
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | | | | | | | | | | | - C Porta
- Division of Medical Oncology, AOU Consorziale Policlinico di Bari, Bari, Italy; Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| |
Collapse
|
6
|
Sasaki T, Hiraki H, Yashima‐Abo A, Nagashima H, Endo F, Yaegashi M, Miura S, Obata K, Yanagawa N, Itamochi H, Shirota H, Iwaya T, Nishizuka SS. Comprehensive Genome Profiling-Initiated Tumor-Informed Circulating Tumor DNA Monitoring for Patients With Advanced Cancer. Cancer Sci 2025; 116:764-774. [PMID: 39757125 PMCID: PMC11875764 DOI: 10.1111/cas.16446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 12/21/2024] [Indexed: 01/07/2025] Open
Abstract
In Japan, comprehensive genome profiling (CGP) as a companion diagnostic (CDx) has been covered by public insurance since June 2019, but the proportion of patients with cancer who actually received drug therapy based on CGP data is low. In the present study, we attempted to use CGP as a starting point for tumor-informed circulating tumor DNA (ctDNA) monitoring. We retrospectively validated 219 patients with malignant tumors who underwent CGP at Iwate Medical University Hospital between October 2019 and April 2023 in terms of patient demographics, genetic analysis, drug recommendations, and drug administration rate. The 219 cancer cases analyzed by CGP for 27 target organs, including prostate (n = 27, 12.3%), colorectal (n = 25, 11.4%), lung (n = 19, 8.7%), and other neoplasms (n = 148, 67.6%). Among the cohort, only 14 cases (6.4%) subsequently were able to undertake the recommended action by Molecular Tumor Board. Of patients who underwent ctDNA monitoring based on somatic mutations identified by CGP (n = 11), clinical validity was confirmed in terms of early relapse prediction (n = 5, 45.5%), treatment response evaluation (n = 10, 90.9%), and no relapse/regrowth corroboration (n = 2, 18.2%) whereas 90.9% (n = 10) of patients obtained information with at least one source of the clinical validity. Although the current rate of CGP contributing to a drug recommendation is low, CGP results can be an alternate resource for tumor-informed longitudinal ctDNA monitoring to provide information concerning early relapse prediction, treatment response evaluation, and no relapse/regrowth corroboration.
Collapse
Affiliation(s)
- Taiga Sasaki
- Division of Biomedical Research and DevelopmentIwate Medical University Institute for Biomedical SciencesYahabaJapan
- Department of Respiratory MedicineIwate Medical University School of MedicineYahabaJapan
| | - Hayato Hiraki
- Division of Biomedical Research and DevelopmentIwate Medical University Institute for Biomedical SciencesYahabaJapan
| | - Akiko Yashima‐Abo
- Division of Biomedical Research and DevelopmentIwate Medical University Institute for Biomedical SciencesYahabaJapan
| | - Hiromi Nagashima
- Department of Respiratory MedicineIwate Medical University School of MedicineYahabaJapan
| | - Fumitaka Endo
- Department of Clinical OncologyIwate Medical University School of MedicineYahabaJapan
| | - Mizunori Yaegashi
- Department of SurgeryIwate Medical University School of MedicineYahabaJapan
| | - Shimpei Miura
- Department of DermatologyIwate Medical University School of MedicineYahabaJapan
| | - Keiko Obata
- Department of Clinical GeneticsIwate Medical University School of MedicineYahabaJapan
| | - Naoki Yanagawa
- Department of Diagnostic PathologyIwate Medical University School of MedicineYahabaJapan
| | - Hiroaki Itamochi
- Department of Clinical OncologyIwate Medical University School of MedicineYahabaJapan
| | - Hidekazu Shirota
- Department of Medical OncologyTohoku University HospitalSendaiJapan
| | - Takeshi Iwaya
- Department of Clinical OncologyIwate Medical University School of MedicineYahabaJapan
| | - Satoshi S. Nishizuka
- Division of Biomedical Research and DevelopmentIwate Medical University Institute for Biomedical SciencesYahabaJapan
| |
Collapse
|
7
|
Gueguen L, Olgiati L, Brutti-Mairesse C, Sans A, Le Texier V, Verlingue L. A prospective pragmatic evaluation of automatic trial matching tools in a molecular tumor board. NPJ Precis Oncol 2025; 9:28. [PMID: 39870746 PMCID: PMC11772588 DOI: 10.1038/s41698-025-00806-y] [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: 10/22/2024] [Accepted: 01/06/2025] [Indexed: 01/29/2025] Open
Abstract
Publicly available trial matching tools can improve the access to therapeutic innovations, but errors may expose to over-solicitation and disappointment. We performed a pragmatic non-interventional prospective evaluation on sequential patients at the Molecular Tumor Board of Centre Leon Berard. During 10 weeks in 2024, we analysed 157 patients with four clinical trial matching tools from the 19 screened: Klineo, ScreenAct, Trialing and DigitalECMT. Each patient had 2.19 trials proposed on average, and 38% had no trials suggested. The mean performances were precision = 0.33, recall = 0.32, AP@3 = 0.45, and NDCG@3 = 0.34. Using all the tools can increase to 26% the clinical trial options. The most frequent error concerned the type of gene variants required by the selection criteria. We showed that using a Large Language Model on the patients' molecular reports could improve the performance by up to 5%. We recommend that experts supervise the results and we advocate for improved technologies.
Collapse
Affiliation(s)
| | | | | | | | | | - Loic Verlingue
- CRCL, Centre Léon Bérard, Lyon, France.
- Phase 1 Clinical Unit, Centre Léon Bérard, Lyon, France.
| |
Collapse
|
8
|
Nikanjam M, Kato S, Allen T, Sicklick JK, Kurzrock R. Novel clinical trial designs emerging from the molecular reclassification of cancer. CA Cancer J Clin 2025. [PMID: 39841128 DOI: 10.3322/caac.21880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/28/2024] [Accepted: 12/09/2024] [Indexed: 01/23/2025] Open
Abstract
Next-generation sequencing has revealed the disruptive reality that advanced/metastatic cancers have complex and individually distinct genomic landscapes, necessitating a rethinking of treatment strategies and clinical trial designs. Indeed, the molecular reclassification of cancer suggests that it is the molecular underpinnings of the disease, rather than the tissue of origin, that mostly drives outcomes. Consequently, oncology clinical trials have evolved from standard phase 1, 2, and 3 tissue-specific studies; to tissue-specific, biomarker-driven trials; to tissue-agnostic trials untethered from histology (all drug-centered designs); and, ultimately, to patient-centered, N-of-1 precision medicine studies in which each patient receives a personalized, biomarker-matched therapy/combination of drugs. Innovative technologies beyond genomics, including those that address transcriptomics, immunomics, proteomics, functional impact, epigenetic changes, and metabolomics, are enabling further refinement and customization of therapy. Decentralized studies have the potential to improve access to trials and precision medicine approaches for underserved minorities. Evaluation of real-world data, assessment of patient-reported outcomes, use of registry protocols, interrogation of exceptional responders, and exploitation of synthetic arms have all contributed to personalized therapeutic approaches. With greater than 1 × 1012 potential patterns of genomic alterations and greater than 4.5 million possible three-drug combinations, the deployment of artificial intelligence/machine learning may be necessary for the optimization of individual therapy and, in the near future, also may permit the discovery of new treatments in real time.
Collapse
Affiliation(s)
- Mina Nikanjam
- Division of Hematology-Oncology, University of California San Diego, La Jolla, California, USA
- Moores Cancer Center, University of California San Diego Health, La Jolla, California, USA
| | - Shumei Kato
- Division of Hematology-Oncology, University of California San Diego, La Jolla, California, USA
- Moores Cancer Center, University of California San Diego Health, La Jolla, California, USA
| | | | - Jason K Sicklick
- Moores Cancer Center, University of California San Diego Health, La Jolla, California, USA
- Division of Surgical Oncology, Department of Surgery, University of California San Diego, San Diego, California, USA
- Department of Pharmacology, University of California San Diego, San Diego, California, USA
| | - Razelle Kurzrock
- Medical College of Wisconsin Cancer Center, Milwaukee, Wisconsin, USA
- Worldwide Innovative Networking in Personalized Cancer Medicine Consortium, Paris, France
| |
Collapse
|
9
|
Schettini F, Sirico M, Loddo M, Williams GH, Hardisty KM, Scorer P, Thatcher R, Rivera P, Milani M, Strina C, Ferrero G, Ungari M, Bottin C, Zanconati F, de Manzini N, Aguggini S, Tancredi R, Fiorio E, Fioravanti A, Scaltriti M, Generali D. Next-generation sequencing-based evaluation of the actionable landscape of genomic alterations in solid tumors: the "MOZART" prospective observational study. Oncologist 2025; 30:oyae206. [PMID: 39177668 PMCID: PMC11783315 DOI: 10.1093/oncolo/oyae206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 07/10/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND The identification of the most appropriate targeted therapies for advanced cancers is challenging. We performed a molecular profiling of metastatic solid tumors utilizing a comprehensive next-generation sequencing (NGS) assay to determine genomic alterations' type, frequency, actionability, and potential correlations with PD-L1 expression. METHODS A total of 304 adult patients with heavily pretreated metastatic cancers treated between January 2019 and March 2021 were recruited. The CLIA-/UKAS-accredit Oncofocus assay targeting 505 genes was used on newly obtained or archived biopsies. Chi-square, Kruskal-Wallis, and Wilcoxon rank-sum tests were used where appropriate. Results were significant for P < .05. RESULTS A total of 237 tumors (78%) harbored potentially actionable genomic alterations. Tumors were positive for PD-L1 in 68.9% of cases. The median number of mutant genes/tumor was 2.0 (IQR: 1.0-3.0). Only 34.5% were actionable ESCAT Tier I-II with different prevalence according to cancer type. The DNA damage repair (14%), the PI3K/AKT/mTOR (14%), and the RAS/RAF/MAPK (12%) pathways were the most frequently altered. No association was found among PD-L1, ESCAT, age, sex, and tumor mutational status. Overall, 62 patients underwent targeted treatment, with 37.1% obtaining objective responses. The same molecular-driven treatment for different cancer types could be associated with opposite clinical outcomes. CONCLUSIONS We highlight the clinical value of molecular profiling in metastatic solid tumors using comprehensive NGS-based panels to improve treatment algorithms in situations of uncertainty and facilitate clinical trial recruitment. However, interpreting genomic alterations in a tumor type-specific manner is critical.
Collapse
Affiliation(s)
- Francesco Schettini
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Medical Oncology Department, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
- Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
| | - Marianna Sirico
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori,”47014, Meldola, Italy
| | - Marco Loddo
- Oncologica UK Ltd, Cambridge CB10 1XL, United Kingdom
| | | | | | - Paul Scorer
- Oncologica UK Ltd, Cambridge CB10 1XL, United Kingdom
| | | | - Pablo Rivera
- Medical Oncology Department, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
| | - Manuela Milani
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34147, Trieste, Italy
| | - Carla Strina
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34147, Trieste, Italy
| | - Giuseppina Ferrero
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, 26100, Cremona, Italy
| | - Marco Ungari
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, 26100, Cremona, Italy
| | - Cristina Bottin
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34147, Trieste, Italy
| | - Fabrizio Zanconati
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34147, Trieste, Italy
| | - Nicolò de Manzini
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34147, Trieste, Italy
| | - Sergio Aguggini
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, 26100, Cremona, Italy
| | - Richard Tancredi
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, 26100, Cremona, Italy
| | - Elena Fiorio
- Section of Oncology, Department of Medicine, University of Verona School of Medicine and Verona University Hospital Trust, 37134, Verona, Italy
| | | | - Maurizio Scaltriti
- Neurosurgery Unit, ASST Cremona, 26100, Cremona, Italy
- AstraZeneca, Gaithersburg, MD 20876, United States
| | - Daniele Generali
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34147, Trieste, Italy
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, 26100, Cremona, Italy
| |
Collapse
|
10
|
Roazzi L, Patelli G, Bencardino KB, Amatu A, Bonazzina E, Tosi F, Amoruso B, Bombelli A, Mariano S, Stabile S, Porta C, Siena S, Sartore-Bianchi A. Ongoing Clinical Trials and Future Research Scenarios of Circulating Tumor DNA for the Treatment of Metastatic Colorectal Cancer. Clin Colorectal Cancer 2024; 23:295-308. [PMID: 38519391 DOI: 10.1016/j.clcc.2024.02.001] [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: 09/14/2023] [Revised: 01/04/2024] [Accepted: 02/11/2024] [Indexed: 03/24/2024]
Abstract
Liquid biopsy using circulating tumor DNA (ctDNA) has emerged as a minimally invasive, timely approach to provide molecular diagnosis and monitor tumor evolution in patients with cancer. Since the molecular landscape of metastatic colorectal cancer (mCRC) is substantially heterogeneous and dynamic over space and time, ctDNA holds significant advantages as a biomarker for this disease. Numerous studies have demonstrated that ctDNA broadly recapitulates the molecular profile of the primary tumor and metastases, and have mainly focused on the genotyping of RAS and BRAF, that is propaedeutic for anti-EGFR treatment selection. However, ctDNA soon broadened its scope towards the assessment of early tumor response, as well as the identification of drug resistance biomarkers to drive potential molecular actionability. In this review article, we provide an overview of the current state-of-the-art of this methodology and its applications, focusing on ongoing clinical trials that employ ctDNA to prospectively guide treatment in patients with mCRC.
Collapse
Affiliation(s)
- Laura Roazzi
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy; Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Giorgio Patelli
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy; Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy; IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Katia Bruna Bencardino
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Alessio Amatu
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Erica Bonazzina
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Federica Tosi
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Brunella Amoruso
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy; Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Anna Bombelli
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Sara Mariano
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Stefano Stabile
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Camillo Porta
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy; Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Salvatore Siena
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy; Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy.
| | - Andrea Sartore-Bianchi
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy; Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Division of Clinical Research and Innovation, Grande Ospedale Metropolitano Niguarda, Milan, Italy.
| |
Collapse
|
11
|
Ehman M, Punian J, Weymann D, Regier DA. Next-generation sequencing in oncology: challenges in economic evaluations. Expert Rev Pharmacoecon Outcomes Res 2024; 24:1115-1132. [PMID: 39096135 DOI: 10.1080/14737167.2024.2388814] [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: 06/20/2024] [Revised: 07/19/2024] [Accepted: 08/01/2024] [Indexed: 08/04/2024]
Abstract
INTRODUCTION Next-generation sequencing (NGS) identifies genetic variants to inform personalized treatment plans. Insufficient evidence of cost-effectiveness impedes the integration of NGS into routine cancer care. The complexity of personalized treatment challenges conventional economic evaluation. Clearly delineating challenges informs future cost-effectiveness analyses to better value and contextualize health, preference-, and equity-based outcomes. AREAS COVERED We conducted a scoping review to characterize the applied methods and outcomes of economic evaluations of NGS in oncology and identify existing challenges. We included 27 articles published since 2016 from a search of PubMed, Embase, and Web of Science. Identified challenges included defining the evaluative scope, managing evidentiary limitations including lack of causal evidence, incorporating preference-based utility, and assessing distributional and equity-based impacts. These challenges reflect the difficulty of generating high-quality clinical effectiveness and real-world evidence (RWE) for NGS-guided interventions. EXPERT OPINION Adapting methodological approaches and developing life-cycle health technology assessment (HTA) guidance using RWE is crucial for implementing NGS in oncology. Healthcare systems, decision-makers, and HTA organizations are facing a pivotal opportunity to adapt to an evolving clinical paradigm and create innovative regulatory and reimbursement processes that will enable more sustainable, equitable, and patient-oriented healthcare.
Collapse
Affiliation(s)
- Morgan Ehman
- Cancer Control Research, BC Cancer, Vancouver, BC, Canada
| | - Jesman Punian
- Cancer Control Research, BC Cancer, Vancouver, BC, Canada
| | - Deirdre Weymann
- Cancer Control Research, BC Cancer, Vancouver, BC, Canada
- Faculty of Health Sciences, Simon Fraser University, Vancouver, BC, Canada
| | - Dean A Regier
- Cancer Control Research, BC Cancer, Vancouver, BC, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
12
|
Navarro P, Beato C, Rodriguez-Moreno JF, Ruiz-Llorente S, Mielgo X, Pineda E, Navarro M, Bruixola G, Grazioso TP, Viudez A, Fuster J, Nogueron E, Mediano MD, Balaña C, Mendez C, Rodriguez RM, Del Barco Berron S, Gongora B, Carmona-Bayonas A, Garcia-Donas J. Prospective study of the real impact of fusion centered genomic assays in patient management in a national collaborative group: the GETHI-XX-16 study. Clin Transl Oncol 2024:10.1007/s12094-024-03745-5. [PMID: 39485597 DOI: 10.1007/s12094-024-03745-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/21/2024] [Indexed: 11/03/2024]
Abstract
PURPOSE Precision medicine represents a paradigm shift in oncology. Access to genetic testing and targeted therapies is frequently limited. Assays based on DNA sequencing can miss druggable alterations. We aimed to determine the impact of a free access program to RNA tests in patient management. METHODS We designed a multicenter prospective observational study within the Spanish National Group for Translational Oncology and Rare and Orphan Tumors (GETTHI). Eligible patients were adults with solid cancers that had progressed on standard therapies. Tumor samples were analyzed using two RNA sequencing assays (Trailblaze PharosTM and Archer FusionPlex Solid TumorTM). A central committee evaluated the actionability of genetic alterations and reported the findings to attending physicians, who made the final clinical management decisions. RESULTS Between November 2016 and April 2019, 395 patients with 41 different tumors across 30 hospitals were included. Molecular analysis revealed actionable genetic alterations in 57 individuals (14.4%). Targeted therapies were advised for 23 and seven received a matched targeted therapy: two lung cancers (EML4-ALK and CD74-ROS1 fusion), three glioblastomas (EGFR point mutations), one oligodendroglioma (FGFR3-TACC3 fusion) and a prostate cancer (SND1-BRAF fusion). The outcomes included two tumor responses, one disease stabilization, one early withdrawal due to toxicity, one progression, and one unknown. CONCLUSION Despite the growing knowledge of cancer biology and its translation to drug development, the overall impact of personalized treatments remains low. Access to comprehensive molecular tests covering properly all known actionable alterations and programs for a wide access to targeted therapies seem to be critical steps.
Collapse
Affiliation(s)
- Paloma Navarro
- Laboratory of Innovation in Oncology; Gynecological, Genitourinary and Skin Tumor Unit, HM CIOCC (Clara Campal Comprehensive Cancer Centre), Sanchinarro HM Universitary Hospital, HM Hospitales, Madrid, Spain, Madrid, Spain
- HM Faculty of Health Sciences, Camilo José Cela University, Madrid, Spain
- HM Hospitals Health Research Institute, Madrid, Spain
- Institute of Applied Molecular Medicine (IMMA), School of Medicine, Universidad San Pablo CEU, CEU Universities, Madrid, Spain
| | - Carmen Beato
- Medical Oncology Department, Hospital Virgen de La Macarena, Seville, Spain
| | - Juan Francisco Rodriguez-Moreno
- Laboratory of Innovation in Oncology; Gynecological, Genitourinary and Skin Tumor Unit, HM CIOCC (Clara Campal Comprehensive Cancer Centre), Sanchinarro HM Universitary Hospital, HM Hospitales, Madrid, Spain, Madrid, Spain
- HM Faculty of Health Sciences, Camilo José Cela University, Madrid, Spain
- HM Hospitals Health Research Institute, Madrid, Spain
- Institute of Applied Molecular Medicine (IMMA), School of Medicine, Universidad San Pablo CEU, CEU Universities, Madrid, Spain
| | - Sergio Ruiz-Llorente
- Laboratory of Innovation in Oncology; Gynecological, Genitourinary and Skin Tumor Unit, HM CIOCC (Clara Campal Comprehensive Cancer Centre), Sanchinarro HM Universitary Hospital, HM Hospitales, Madrid, Spain, Madrid, Spain
- Institute of Applied Molecular Medicine (IMMA), School of Medicine, Universidad San Pablo CEU, CEU Universities, Madrid, Spain
- Departamento de Biomedicina y Biotecnología, Área de Genética, Universidad de Alcalá, Madrid, Spain
| | - Xabier Mielgo
- Medical Oncology Department , Hospital Universitario Fundacion Alcorcon, Madrid, Spain
| | - Estela Pineda
- Medical Oncology Department, Hospital Clinic Barcelona, Barcelona, Spain
| | - Miguel Navarro
- Medical Oncology Department, Complejo Asistencial Universitario de Salamanca, Salamanca, Spain
| | - Gema Bruixola
- Medical Oncology Department, Hospital Clinico Universtario - INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Tatiana P Grazioso
- Laboratory of Innovation in Oncology; Gynecological, Genitourinary and Skin Tumor Unit, HM CIOCC (Clara Campal Comprehensive Cancer Centre), Sanchinarro HM Universitary Hospital, HM Hospitales, Madrid, Spain, Madrid, Spain
- HM Faculty of Health Sciences, Camilo José Cela University, Madrid, Spain
- HM Hospitals Health Research Institute, Madrid, Spain
- Institute of Applied Molecular Medicine (IMMA), School of Medicine, Universidad San Pablo CEU, CEU Universities, Madrid, Spain
| | - Antonio Viudez
- Medical Oncology Department, Hospital de Navarra, Pamplona, Spain
| | - Jose Fuster
- Medical Oncology Department, Hospital Universitario Son Espases, Palma, Spain
| | - Esther Nogueron
- Medical Oncology Department, Complejo Hospitalario Universitario de Albacete, Albacete, Spain
| | | | - Carmen Balaña
- Instituto Catalán de Oncología-Badalona, Barcelona, Spain
| | | | | | | | | | | | - Jesus Garcia-Donas
- Laboratory of Innovation in Oncology; Gynecological, Genitourinary and Skin Tumor Unit, HM CIOCC (Clara Campal Comprehensive Cancer Centre), Sanchinarro HM Universitary Hospital, HM Hospitales, Madrid, Spain, Madrid, Spain.
- HM Faculty of Health Sciences, Camilo José Cela University, Madrid, Spain.
- HM Hospitals Health Research Institute, Madrid, Spain.
- Institute of Applied Molecular Medicine (IMMA), School of Medicine, Universidad San Pablo CEU, CEU Universities, Madrid, Spain.
| |
Collapse
|
13
|
Boileve A, Smolenschi C, Lambert A, Boige V, Tarabay A, Valery M, Fuerea A, Pudlarz T, Conroy T, Hollebecque A, Ducreux M. Role of molecular biology in the management of pancreatic cancer. World J Gastrointest Oncol 2024; 16:2902-2914. [PMID: 39072173 PMCID: PMC11271790 DOI: 10.4251/wjgo.v16.i7.2902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/04/2024] [Accepted: 05/21/2024] [Indexed: 07/12/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) presents significant challenges in patient management due to a dismal prognosis, increasing incidence, and limited treatment options. In this regard, precision medicine, which personalizes treatments based on tumour molecular characteristics, has gained great interest. However, its widespread implementation is not fully endorsed in current recommendations. This review explores key molecular alterations in PDAC, while emphasizing differences between KRAS-mutated and KRAS-wild-type tumours. It assesses the practical application of precision medicine in clinical settings and outlines potential future directions with respect to PDAC. Actionable molecular targets are examined with the aim of enhancing our understanding of PDAC molecular biology. Insights from this analysis may contribute to a more refined and personalized approach to pancreatic cancer treatment, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Alice Boileve
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| | | | - Aurélien Lambert
- Department of Medical Oncology, Institut de Cancérologie de Lorraine, Nancy 54519, France
| | - Valérie Boige
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| | - Anthony Tarabay
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| | - Marine Valery
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| | - Alina Fuerea
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| | - Thomas Pudlarz
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| | - Thierry Conroy
- Department of Medical Oncology, Institut de Cancérologie de Lorraine, Nancy 54519, France
| | | | - Michel Ducreux
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| |
Collapse
|
14
|
Metzger P, Boerries M. [The collaborative project "Personalized medicine for oncology" (PM4Onco) as part of the Medical Informatics Initiative (MII)]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2024; 67:668-675. [PMID: 38739266 PMCID: PMC11166753 DOI: 10.1007/s00103-024-03886-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2024] [Indexed: 05/14/2024]
Abstract
The collaborative project Personalized Medicine for Oncology (PM4Onco) was launched in 2023 as part of the National Decade against Cancer (NKD) and is executed within the Medical Informatics Initiative (MII). Its aim is to establish a sustainable infrastructure for the integration and use of data from clinical and biomedical research and therefore combines the experience and preliminary work of all four consortia of the MII and the leading oncology centers in Germany. The data provided by PM4Onco will be prepared in a suitable form to support decision making in molecular tumor boards. This concept and infrastructure will be extended to 23 participating partner sites and thus improve access to targeted therapies based on clinical information and analysis of molecular genetic alterations in tumors at different stages of the disease. This will help to improve the treatment and prognosis of tumor diseases.Clinical cancer registries are involved in the project to improve data quality through standardized documentation routines. Clinical experts advise on the expansion of the core datasets for personalized medicine (PM). Information on quality of life and treatment outcomes reported by patients in questionnaires, which is rarely collected outside of clinical trials, will make a significant contribution. Patient representatives are involved from the onset to ensure that the important perspective of patients is taken into account in the decision-making process. PM4Onco thus creates an alliance between the MII, oncological centers of excellence, clinical cancer registries, young scientists, patients, and citizens to strengthen and advance PM in cancer therapy.
Collapse
Affiliation(s)
- Patrick Metzger
- Institut für Medizinische Bioinformatik und Systemmedizin (IBSM), Universitätsklinikum Freiburg, Medizinische Fakultät, Universität Freiburg, Breisacher Straße 153, 79110, Freiburg, Deutschland
| | - Melanie Boerries
- Institut für Medizinische Bioinformatik und Systemmedizin (IBSM), Universitätsklinikum Freiburg, Medizinische Fakultät, Universität Freiburg, Breisacher Straße 153, 79110, Freiburg, Deutschland.
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Standort Freiburg, Kooperation zwischen DKFZ und Universitätsklinikum Freiburg, Universität Freiburg, Freiburg, Deutschland.
- Comprehensive Cancer Center Freiburg (CCCF), Universitätsklinikum Freiburg, Universität Freiburg, Freiburg, Deutschland.
| |
Collapse
|
15
|
Bogdan L, Saleh RR, Avery L, Del Rossi S, Yu C, Bedard PL. Clinical Utility of Tumor Next-Generation Sequencing Panel Testing to Inform Treatment Decisions for Patients With Advanced Solid Tumors in a Tertiary Care Center. JCO Precis Oncol 2024; 8:e2400092. [PMID: 38935894 PMCID: PMC11371095 DOI: 10.1200/po.24.00092] [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: 02/06/2024] [Revised: 03/28/2024] [Accepted: 05/02/2024] [Indexed: 06/29/2024] Open
Abstract
PURPOSE There is limited information about the clinical utility of targeted next-generation sequencing (NGS) panel testing to inform decision making for patients with advanced solid tumors. The Ontario-wide Cancer Targeted Nucleic Acid Evaluation (OCTANE) is a prospective study that enrolled more than 4,500 patients with solid tumor for NGS panel testing. We performed a retrospective survey of medical oncologists to evaluate the impact of NGS testing on treatment decisions. METHODS Patients and treating oncologists were identified at the Princess Margaret Cancer Center between 2016 and 2021. Tumor-only sequencing was performed using a gene panel of either 555 or 161 cancer genes. Oncologists were asked to review testing results and complete a survey indicating whether NGS testing affected treatment decisions. The primary outcome of this study was rate of treatment change on the basis of mutation results. Patient, test, and physician factors were evaluated for association with treatment changes using univariate analyses and a mixed-effects model. RESULTS Of the 582 surveys sent, 394 (67.7%) were completed. We found that 188 (47.7%) patients had testing results classified as actionable by the oncologist and 62 (15.7%) patients were matched to treatment, of whom 37 (60%) were enrolled in a clinical trial, 13 (21%) received an approved drug, four (6%) were prescribed off-label therapy, and eight (13%) avoided ineffective treatment. Patient, test, and physician characteristics were not significantly associated with treatment change. There was no difference in overall survival between patients who received matched treatment versus those who did not (P = .55, median survival not reached). CONCLUSION OCTANE testing led to a change in drug treatment in 15.7% of patients, supporting the clinical utility of NGS panel testing for patients with advanced solid tumors.
Collapse
Affiliation(s)
- Lucia Bogdan
- Division of Medical Oncology, Department of Medicine, University of Toronto, Toronto, Canada
| | - Ramy R. Saleh
- Department of Medical Oncology, McGill University Health Centre, Montreal, Canada
| | - Lisa Avery
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - Samanta Del Rossi
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - Celeste Yu
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - Philippe L. Bedard
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| |
Collapse
|
16
|
Verlingue L, Desevre M, Polito M, Garin G, Rodriguez C, Qing W, Tredan O, Perol D, Ray-Coquard I, Chabaud S, Blay JY. The French multicentric molecular analysis platforms and personalized medicine trials MOST, MOST Plus and MEGAMOST. Acta Oncol 2024; 63:411-417. [PMID: 38807312 PMCID: PMC11332485 DOI: 10.2340/1651-226x.2024.32745] [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: 11/30/2023] [Accepted: 02/29/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND AND PURPOSE In this manuscript we describe the academic French multicentric molecular analysis platforms including PROFILER, promoted by Centre Léon Berard, and the multicentric personalized medicine trials MOST, MOST Plus and MEGAMOST. PATIENTS/MATERIAL AND METHODS MOST, MOST Plus and MEGAMOST comprise 14 cohorts with different targeted agents and immunotherapies. RESULTS AND INTERPRETATION PROFILER has recruited 5,991 patients in 10 years, MOST and MOST Plus 875 patients since 2014 and MEGAMOST 172 patients since 2020, and are still ongoing. We provide a description of the local, national and international implications of these initiatives, and we review the results of the sorafenib and olaparib cohorts.
Collapse
Affiliation(s)
- Loic Verlingue
- Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France.
| | - Marine Desevre
- Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Marie Polito
- Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Gwenaelle Garin
- Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Christine Rodriguez
- Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Wang Qing
- Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Olivier Tredan
- Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - David Perol
- Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | | | - Sylvie Chabaud
- Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Jean Yves Blay
- Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| |
Collapse
|
17
|
Kim SY, Kim JH, Kim TY, Park SR, Yoon S, Lee S, Lee SH, Kim TM, Han SW, Kim HR, Yun H, Lee S, Kim J, Choi YL, Choi KS, Chae H, Ryu H, Lee GW, Zang DY, Ahn JB. Pragmatic nationwide master observational trial based on genomic alterations in advanced solid tumors: KOrean Precision Medicine Networking Group Study of MOlecular profiling guided therapy based on genomic alterations in advanced Solid tumors (KOSMOS)-II study protocol KCSG AL-22-09. BMC Cancer 2024; 24:574. [PMID: 38724991 PMCID: PMC11080169 DOI: 10.1186/s12885-024-12338-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 05/03/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND Next-generation sequencing (NGS) has been introduced to many Korean institutions to support molecular diagnostics in cancer since 2017, when it became eligible for reimbursement by the National Health Insurance Service. However, the uptake of molecularly guided treatment (MGT) based on NGS results has been limited because of stringent regulations regarding prescriptions outside of approved indications, a lack of clinical trial opportunities, and limited access to molecular tumor boards (MTB) at most institutions. The KOSMOS-II study was designed to demonstrate the feasibility and effectiveness of MGT, informed by MTBs, using a nationwide precision medicine platform. METHODS The KOSMOS-II trial is a large-scale nationwide master observational study. It involves a framework for screening patients with metastatic solid tumors for actionable genetic alterations based on local NGS testing. It recommends MGT through a remote and centralized MTB meeting held biweekly. MGT can include one of the following options: Tier 1, the therapeutic use of investigational drugs targeting genetic alterations such as ALK, EGFR, ERBB2, BRAF, FH, ROS1, and RET, or those with high tumor mutational burden; Tier 2, comprising drugs with approved indications or those permitted for treatment outside of the indications approved by the Health Insurance Review and Assessment Service of Korea; Tier 3, involving clinical trials matching the genetic alterations recommended by the MTB. Given the anticipated proportion of patients receiving MGT in the range of 50% ± 3.25%, this study aims to enroll 1,000 patients. Patients must have progressed to one or more lines of therapy and undergone NGS before enrollment. DISCUSSION This pragmatic master protocol provides a mass-screening platform for rare genetic alterations and high-quality real-world data. Collateral clinical trials, translational studies, and clinico-genomic databases will contribute to generating evidence for drug repositioning and the development of new biomarkers. TRIAL REGISTRATION NCT05525858.
Collapse
Grants
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- Roche, Basel, Switzerland
- Lunit, Seoul, Republic of Korea
Collapse
Affiliation(s)
- Sun Young Kim
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Jee Hyun Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea.
| | - Tae-Yong Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Sook Ryun Park
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Shinkyo Yoon
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Soohyeon Lee
- Department of Internal Medicine, Korea University College of Medicine, Korea University Anam Hospital, Seoul, South Korea
| | - Se-Hoon Lee
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Tae Min Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Sae-Won Han
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Hye Ryun Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Yonsei Cancer Center, Seoul, South Korea
| | - Hongseok Yun
- Center for Genomic Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Sejoon Lee
- Center for Precision Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Jihun Kim
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Yoon-La Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kui Son Choi
- Department of Cancer Control and Population Health, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, South Korea
| | - Heejung Chae
- Department of Internal Medicine, National Cancer Center, Goyang, South Korea
| | - Hyewon Ryu
- Division of Hematology and Oncology, Department of Internal Medicine, Chungnam National University Hospital, Chungnam National University College of Medicine, Daejeon, South Korea
| | - Gyeong-Won Lee
- Division of Hematology-Oncology, Department of Internal Medicine, Institute of Health Science, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju, South Korea
| | - Dae Young Zang
- Department of Internal Medicine, Hallym University College of Medicine, Hallym University Sacred Heart Hospital, Anyang, South Korea
| | - Joong Bae Ahn
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Yonsei Cancer Center, Seoul, South Korea
| |
Collapse
|
18
|
Lörsch AM, Jung J, Lange S, Pfarr N, Mogler C, Illert AL. [Personalized medicine in oncology]. PATHOLOGIE (HEIDELBERG, GERMANY) 2024; 45:180-189. [PMID: 38568256 DOI: 10.1007/s00292-024-01315-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/16/2024] [Indexed: 04/26/2024]
Abstract
Due to the considerable technological progress in molecular and genetic diagnostics as well as increasing insights into the molecular pathogenesis of diseases, there has been a fundamental paradigm shift in the past two decades from a "one-size-fits-all approach" to personalized, molecularly informed treatment strategies. Personalized medicine or precision medicine focuses on the genetic, physiological, molecular, and biochemical differences between individuals and considers their effects on the development, prevention, and treatment of diseases. As a pioneer of personalized medicine, the field of oncology is particularly noteworthy, where personalized diagnostics and treatment have led to lasting change in the treatment of cancer patients in recent years. In this article, the significant change towards personalized treatment concepts, especially in the field of personalized oncology, will be discussed and examined in more detail.
Collapse
Affiliation(s)
- Alisa Martina Lörsch
- Zentrum für Personalisierte Medizin (ZPM), Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, München, Deutschland
- Klinik und Poliklinik für Innere Medizin III, Hämatologie und Onkologie, Klinikum rechts der Isar, Technische Universität München, München, Deutschland
- Bayerisches Zentrum für Krebsforschung (BZKF), Standort Technische Universität München, München, Deutschland
| | - Johannes Jung
- Zentrum für Personalisierte Medizin (ZPM), Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, München, Deutschland
- Klinik und Poliklinik für Innere Medizin III, Hämatologie und Onkologie, Klinikum rechts der Isar, Technische Universität München, München, Deutschland
- Bayerisches Zentrum für Krebsforschung (BZKF), Standort Technische Universität München, München, Deutschland
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Standort München, München, Deutschland
| | - Sebastian Lange
- Zentrum für Personalisierte Medizin (ZPM), Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, München, Deutschland
- Bayerisches Zentrum für Krebsforschung (BZKF), Standort Technische Universität München, München, Deutschland
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technische Universität München, München, Deutschland
- Comprehensive Cancer Center München, Klinikum rechts der Isar, Technische Universität München, München, Deutschland
| | - Nicole Pfarr
- Zentrum für Personalisierte Medizin (ZPM), Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, München, Deutschland
- Bayerisches Zentrum für Krebsforschung (BZKF), Standort Technische Universität München, München, Deutschland
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Standort München, München, Deutschland
- Institut für Allgemeine Pathologie und Pathologische Anatomie, Technische Universität München, München, Deutschland
| | - Carolin Mogler
- Zentrum für Personalisierte Medizin (ZPM), Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, München, Deutschland
- Bayerisches Zentrum für Krebsforschung (BZKF), Standort Technische Universität München, München, Deutschland
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Standort München, München, Deutschland
- Comprehensive Cancer Center München, Klinikum rechts der Isar, Technische Universität München, München, Deutschland
- Institut für Allgemeine Pathologie und Pathologische Anatomie, Technische Universität München, München, Deutschland
| | - Anna Lena Illert
- Zentrum für Personalisierte Medizin (ZPM), Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, München, Deutschland.
- Klinik und Poliklinik für Innere Medizin III, Hämatologie und Onkologie, Klinikum rechts der Isar, Technische Universität München, München, Deutschland.
- Bayerisches Zentrum für Krebsforschung (BZKF), Standort Technische Universität München, München, Deutschland.
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Standort München, München, Deutschland.
- Comprehensive Cancer Center München, Klinikum rechts der Isar, Technische Universität München, München, Deutschland.
- Klinik für Innere Medizin I, Abteilung für Hämatologie, Onkologie und Stammzelltransplantation, Universitätsklinikum Freiburg, Freiburg, Deutschland.
| |
Collapse
|
19
|
Aleksakhina SN, Ivantsov AO, Imyanitov EN. Agnostic Administration of Targeted Anticancer Drugs: Looking for a Balance between Hype and Caution. Int J Mol Sci 2024; 25:4094. [PMID: 38612902 PMCID: PMC11012409 DOI: 10.3390/ijms25074094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
Many tumors have well-defined vulnerabilities, thus potentially allowing highly specific and effective treatment. There is a spectrum of actionable genetic alterations which are shared across various tumor types and, therefore, can be targeted by a given drug irrespective of tumor histology. Several agnostic drug-target matches have already been approved for clinical use, e.g., immune therapy for tumors with microsatellite instability (MSI) and/or high tumor mutation burden (TMB), NTRK1-3 and RET inhibitors for cancers carrying rearrangements in these kinases, and dabrafenib plus trametinib for BRAF V600E mutated malignancies. Multiple lines of evidence suggest that this histology-independent approach is also reasonable for tumors carrying ALK and ROS1 translocations, biallelic BRCA1/2 inactivation and/or homologous recombination deficiency (HRD), strong HER2 amplification/overexpression coupled with the absence of other MAPK pathway-activating mutations, etc. On the other hand, some well-known targets are not agnostic: for example, PD-L1 expression is predictive for the efficacy of PD-L1/PD1 inhibitors only in some but not all cancer types. Unfortunately, the individual probability of finding a druggable target in a given tumor is relatively low, even with the use of comprehensive next-generation sequencing (NGS) assays. Nevertheless, the rapidly growing utilization of NGS will significantly increase the number of patients with highly unusual or exceptionally rare tumor-target combinations. Clinical trials may provide only a framework for treatment attitudes, while the decisions for individual patients usually require case-by-case consideration of the probability of deriving benefit from agnostic versus standard therapy, drug availability, associated costs, and other circumstances. The existing format of data dissemination may not be optimal for agnostic cancer medicine, as conventional scientific journals are understandably biased towards the publication of positive findings and usually discourage the submission of case reports. Despite all the limitations and concerns, histology-independent drug-target matching is certainly feasible and, therefore, will be increasingly utilized in the future.
Collapse
Affiliation(s)
- Svetlana N. Aleksakhina
- Department of Tumor Growth Biology, N. N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia
| | - Alexander O. Ivantsov
- Department of Tumor Growth Biology, N. N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia
- Department of Medical Genetics, St. Petersburg Pediatric Medical University, 194100 St. Petersburg, Russia
| | - Evgeny N. Imyanitov
- Department of Tumor Growth Biology, N. N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia
- Department of Medical Genetics, St. Petersburg Pediatric Medical University, 194100 St. Petersburg, Russia
| |
Collapse
|
20
|
Giacomini P, Ciliberto G. Molecular Tumor Boards: On the evolution of species. Eur J Cancer 2024; 201:113910. [PMID: 38382152 DOI: 10.1016/j.ejca.2024.113910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/23/2024]
Affiliation(s)
- Patrizio Giacomini
- Clinical Trial Center, Biostatistics and Bioinformatics, IRCSS Regina Elena National Cancer Institute, Rome, Italy.
| | | |
Collapse
|
21
|
Kubo T, Sunami K, Koyama T, Kitami M, Fujiwara Y, Kondo S, Yonemori K, Noguchi E, Morizane C, Goto Y, Maejima A, Iwasa S, Hamaguchi T, Kawai A, Namikawa K, Arakawa A, Sugiyama M, Ohno M, Yoshida T, Hiraoka N, Yoshida A, Yoshida M, Nishino T, Furukawa E, Narushima D, Nagai M, Kato M, Ichikawa H, Fujiwara Y, Kohno T, Yamamoto N. The impact of rare cancer and early-line treatments on the benefit of comprehensive genome profiling-based precision oncology. ESMO Open 2024; 9:102981. [PMID: 38613908 PMCID: PMC11033064 DOI: 10.1016/j.esmoop.2024.102981] [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: 07/11/2023] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 04/15/2024] Open
Abstract
BACKGROUND Comprehensive genome profiling (CGP) serves as a guide for suitable genomically matched therapies for patients with cancer. However, little is known about the impact of the timing and types of cancer on the therapeutic benefit of CGP. MATERIALS AND METHODS A single hospital-based pan-cancer prospective study (TOP-GEAR; UMIN000011141) was conducted to examine the benefit of CGP with respect to the timing and types of cancer. Patients with advanced solid tumors (>30 types) who either progressed with or without standard treatments were genotyped using a single CGP test. The subjects were followed up for a median duration of 590 days to examine therapeutic response, using progression-free survival (PFS), PFS ratio, and factors associated with therapeutic response. RESULTS Among the 507 patients, 62 (12.2%) received matched therapies with an overall response rate (ORR) of 32.3%. The PFS ratios (≥1.3) were observed in 46.3% (19/41) of the evaluated patients. The proportion of subjects receiving such therapies in the rare cancer cohort was lower than that in the non-rare cancer cohort (9.6% and 17.4%, respectively; P = 0.010). However, ORR of the rare cancer patients was higher than that in the non-rare cancer cohort (43.8% and 20.0%, respectively; P = 0.046). Moreover, ORR of matched therapies in the first or second line after receiving the CGP test was higher than that in the third or later lines (62.5% and 21.7%, respectively; P = 0.003). Rare cancer and early-line treatment were significantly and independently associated with ORR of matched therapies in multivariable analysis (P = 0.017 and 0.004, respectively). CONCLUSION Patients with rare cancer preferentially benefited from tumor mutation profiling by increasing the chances of therapeutic response to matched therapies. Early-line treatments after profiling increase the therapeutic benefit, irrespective of tumor types.
Collapse
Affiliation(s)
- T Kubo
- Department of Laboratory Medicine, National Cancer Center Hospital, Tokyo; Department of Clinical Genomics, National Cancer Center Research Institute, Tokyo
| | - K Sunami
- Department of Laboratory Medicine, National Cancer Center Hospital, Tokyo; Division of Genome Biology, National Cancer Center Research Institute, Tokyo
| | - T Koyama
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo
| | - M Kitami
- Department of Laboratory Medicine, National Cancer Center Hospital, Tokyo
| | - Y Fujiwara
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo; Department of Thoracic Oncology, Aichi Cancer Center Hospital, Aichi
| | - S Kondo
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo; Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo
| | - K Yonemori
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo; Department of Medical Oncology, National Cancer Center Hospital, Tokyo
| | - E Noguchi
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo
| | - C Morizane
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo
| | - Y Goto
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo
| | - A Maejima
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo; Department of Urology, National Cancer Center Hospital, Tokyo
| | - S Iwasa
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo; Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo
| | - T Hamaguchi
- Department of Medical Oncology, Saitama Medical University International Medical Center, Saitama
| | - A Kawai
- Department of Musculoskeletal Oncology and Rehabilitation, National Cancer Center Hospital, Tokyo
| | - K Namikawa
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo
| | - A Arakawa
- Department of Pediatric Oncology, National Cancer Center Hospital, Tokyo
| | - M Sugiyama
- Department of Pediatric Oncology, National Cancer Center Hospital, Tokyo
| | - M Ohno
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo
| | - T Yoshida
- Department of Genetic Services and Medicine, National Cancer Center Hospital, Tokyo
| | - N Hiraoka
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo
| | - A Yoshida
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo
| | - M Yoshida
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo
| | - T Nishino
- Department of Laboratory Medicine, National Cancer Center Hospital, Tokyo
| | - E Furukawa
- Division of Bioinformatics, National Cancer Center Research Institute, Tokyo
| | - D Narushima
- Division of Bioinformatics, National Cancer Center Research Institute, Tokyo
| | - M Nagai
- Division of Bioinformatics, National Cancer Center Research Institute, Tokyo
| | - M Kato
- Division of Bioinformatics, National Cancer Center Research Institute, Tokyo
| | - H Ichikawa
- Department of Clinical Genomics, National Cancer Center Research Institute, Tokyo; Division of Translational Genomics, National Cancer Center Exploratory Oncology Research & Clinical Trial Center, Tokyo, Japan
| | - Y Fujiwara
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo
| | - T Kohno
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo; Division of Translational Genomics, National Cancer Center Exploratory Oncology Research & Clinical Trial Center, Tokyo, Japan
| | - N Yamamoto
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo.
| |
Collapse
|
22
|
Corbaux P, Bayle A, Besle S, Vinceneux A, Vanacker H, Ouali K, Hanvic B, Baldini C, Cassier PA, Terret C, Verlingue L. Patients' selection and trial matching in early-phase oncology clinical trials. Crit Rev Oncol Hematol 2024; 196:104307. [PMID: 38401694 DOI: 10.1016/j.critrevonc.2024.104307] [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: 11/15/2023] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Early-phase clinical trials (EPCT) represent an important part of innovations in medical oncology and a valuable therapeutic option for patients with metastatic cancers, particularly in the era of precision medicine. Nevertheless, adult patients' participation in oncology clinical trials is low, ranging from 2% to 8% worldwide, with unequal access, and up to 40% risk of early discontinuation in EPCT, mostly due to cancer-related complications. DESIGN We review the tools and initiatives to increase patients' orientation and access to early phase cancer clinical trials, and to limit early discontinuation. RESULTS New approaches to optimize the early-phase clinical trial referring process in oncology include automatic trial matching, tools to facilitate the estimation of patients' prognostic and/or to better predict patients' eligibility to clinical trials. Classical and innovative approaches should be associated to double patient recruitment, improve clinical trial enrollment experience and reduce early discontinuation rates. CONCLUSIONS Whereas EPCT are essential for patients to access the latest medical innovations in oncology, offering the appropriate trial when it is relevant for patients should increase by organizational and technological innovations. The oncologic community will need to closely monitor their performance, portability and simplicity for implementation in daily clinical practice.
Collapse
Affiliation(s)
- P Corbaux
- Medical Oncology Department, Centre Léon Bérard, Lyon, France; Medical Oncology, Institut de Cancérologie et d'Hématologie Universitaire de Saint-Étienne (ICHUSE), Centre Hospitalier Universitaire de Saint-Etienne, France
| | - A Bayle
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif F-94805, France
| | - S Besle
- Centre de Recherche en Cancérologie de Lyon (CRCL), France
| | - A Vinceneux
- Medical Oncology Department, Centre Léon Bérard, Lyon, France
| | - H Vanacker
- Medical Oncology Department, Centre Léon Bérard, Lyon, France; Centre de Recherche en Cancérologie de Lyon (CRCL), France
| | - K Ouali
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif F-94805, France
| | - B Hanvic
- Medical Oncology Department, Centre Léon Bérard, Lyon, France
| | - C Baldini
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif F-94805, France
| | - P A Cassier
- Medical Oncology Department, Centre Léon Bérard, Lyon, France; Centre de Recherche en Cancérologie de Lyon (CRCL), France
| | - C Terret
- Medical Oncology Department, Centre Léon Bérard, Lyon, France
| | - L Verlingue
- Medical Oncology Department, Centre Léon Bérard, Lyon, France; Centre de Recherche en Cancérologie de Lyon (CRCL), France.
| |
Collapse
|
23
|
Dufresne A, Attignon V, Ferrari A, Tonon L, Boyault S, Tabone‐Eglinger S, Cassier P, Trédan O, Corradini N, Vinceneux A, Swalduz A, Viari A, Chabaud S, Pérol D, Blay JY, Saintigny P. Added value of whole-exome and RNA sequencing in advanced and refractory cancer patients with no molecular-based treatment recommendation based on a 90-gene panel. Cancer Med 2024; 13:e7115. [PMID: 38553950 PMCID: PMC10980928 DOI: 10.1002/cam4.7115] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 02/21/2024] [Accepted: 03/04/2024] [Indexed: 04/02/2024] Open
Abstract
INTRODUCTION The objective was to determine the added value of comprehensive molecular profile by whole-exome and RNA sequencing (WES/RNA-Seq) in advanced and refractory cancer patients who had no molecular-based treatment recommendation (MBTR) based on a more limited targeted gene panel (TGP) plus array-based comparative genomic hybridization (aCGH). MATERIALS AND METHODS In this retrospective analysis, we selected 50 patients previously included in the PROFILER trial (NCT01774409) for which no MBT could be recommended based on a targeted 90-gene panel and aCGH. For each patient, the frozen tumor sample mirroring the FFPE sample used for TGP/aCGH analysis were processed for WES and RNA-Seq. Data from TGP/aCGH were reanalyzed, and together with WES/RNA-Seq, findings were simultaneously discussed at a new molecular tumor board (MTB). RESULTS After exclusion of variants of unknown significance, a total of 167 somatic molecular alterations were identified in 50 patients (median: 3 [1-10]). Out of these 167 relevant molecular alterations, 51 (31%) were common to both TGP/aCGH and WES/RNA-Seq, 19 (11%) were identified by the TGP/aCGH only and 97 (58%) were identified by WES/RNA-Seq only, including two fusion transcripts in two patients. A MBTR was provided in 4/50 (8%) patients using the information from TGP/aCGH versus 9/50 (18%) patients using WES/RNA-Seq findings. Three patients had similar recommendations based on TGP/aCGH and WES/RNA-Seq. CONCLUSIONS In advanced and refractory cancer patients in whom no MBTR was recommended from TGP/aCGH, WES/RNA-Seq allowed to identify more alterations which may in turn, in a limited fraction of patients, lead to new MBTR.
Collapse
Affiliation(s)
| | | | - Anthony Ferrari
- Platform of Bioinformatics Gilles‐ThomasCentre Léon BérardLyonFrance
| | - Laurie Tonon
- Platform of Bioinformatics Gilles‐ThomasCentre Léon BérardLyonFrance
| | | | | | | | - Olivier Trédan
- Department of Medical OncologyCentre Léon BérardLyonFrance
| | - Nadège Corradini
- Department of Pediatric Oncology, Institute of Pediatric Hematology and OncologyCentre Leon BérardLyonFrance
| | | | | | - Alain Viari
- Platform of Bioinformatics Gilles‐ThomasCentre Léon BérardLyonFrance
| | - Sylvie Chabaud
- Department of Clinical ResearchCentre Léon BérardLyonFrance
| | - David Pérol
- Department of Clinical ResearchCentre Léon BérardLyonFrance
| | - Jean Yves Blay
- Department of Medical OncologyCentre Léon BérardLyonFrance
- Univ Lyon, Claude Bernard Lyon 1 University, INSERM 1052, CNRS 5286, Centre Léon BérardCancer Research Center of LyonLyonFrance
| | - Pierre Saintigny
- Department of Medical OncologyCentre Léon BérardLyonFrance
- Univ Lyon, Claude Bernard Lyon 1 University, INSERM 1052, CNRS 5286, Centre Léon BérardCancer Research Center of LyonLyonFrance
| |
Collapse
|
24
|
Bottosso M, Mosele F, Michiels S, Cournède PH, Dogan S, Labaki C, André F. Moving toward precision medicine to predict drug sensitivity in patients with metastatic breast cancer. ESMO Open 2024; 9:102247. [PMID: 38401248 PMCID: PMC10982863 DOI: 10.1016/j.esmoop.2024.102247] [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: 07/29/2023] [Revised: 01/03/2024] [Accepted: 01/10/2024] [Indexed: 02/26/2024] Open
Abstract
Tumor heterogeneity represents a major challenge in breast cancer, being associated with disease progression and treatment resistance. Precision medicine has been extensively applied to dissect tumor heterogeneity and, through a deeper molecular understanding of the disease, to personalize therapeutic strategies. In the last years, technological advances have widely improved the understanding of breast cancer biology and several trials have been developed to translate these new insights into clinical practice, with the ultimate aim of improving patients' outcomes. In the era of molecular oncology, genomics analyses and other methodologies are shaping a new treatment algorithm in breast cancer care. In this manuscript, we review the main steps of precision medicine to predict drug sensitivity in breast cancer from a translational point of view. Genomic developments and their clinical implications are discussed, along with technological advancements that could broaden precision medicine applications. Current achievements are put into perspective to provide an overview of the state-of-art of breast cancer precision oncology as well as to identify future research directions.
Collapse
Affiliation(s)
- M Bottosso
- INSERM Unit U981, Gustave Roussy Cancer Campus, Villejuif, France; Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - F Mosele
- INSERM Unit U981, Gustave Roussy Cancer Campus, Villejuif, France; Department of Medical Oncology, Gustave Roussy, Villejuif
| | - S Michiels
- Gustave Roussy, Department of Biostatistics and Epidemiology, Villejuif; Oncostat U1018, Inserm, Université Paris-Saclay, Ligue Contre le Cancer, Villejuif
| | - P-H Cournède
- Université Paris-Saclay, Centrale Supélec, Laboratory of Mathematics and Computer Science (MICS), Gif-Sur-Yvette, France
| | - S Dogan
- INSERM Unit U981, Gustave Roussy Cancer Campus, Villejuif, France
| | - C Labaki
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, USA
| | - F André
- INSERM Unit U981, Gustave Roussy Cancer Campus, Villejuif, France; Department of Medical Oncology, Gustave Roussy, Villejuif; PRISM, INSERM, Gustave Roussy, Villejuif; Paris Saclay University, Gif Sur-Yvette, France.
| |
Collapse
|
25
|
Ishimaru S, Shimoi T, Sunami K, Nakajima M, Ando Y, Okita N, Nakamura K, Shibata T, Fujiwara Y, Yamamoto N. Platform trial for off-label oncology drugs using comprehensive genomic profiling under the universal public healthcare system: the BELIEVE trial. Int J Clin Oncol 2024; 29:89-95. [PMID: 38112833 PMCID: PMC10808137 DOI: 10.1007/s10147-023-02439-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 11/08/2023] [Indexed: 12/21/2023]
Abstract
BACKGROUND Precision medicine has transformed cancer treatment by focusing on personalized approaches based on genomic abnormalities. However, comprehensive genomic profiling (CGP) and access to targeted therapies are limited in Japan. This study investigates the BELIEVE trial, which aims to improve drug accessibility for patients with actionable genetic abnormalities through off-label drug administration. METHODS The BELIEVE trial is a platform trial with a single master protocol, conducted under the Clinical Trials Act and the patient-proposed health services (PPHS) scheme. Eligible patients with solid tumors exhibiting actionable alterations were enrolled, and CGP tests covered by national health insurance were employed. Treatment selection, study drugs from collaborating pharmaceutical companies, and treatment schedules adhered to predefined protocols. Primary and secondary endpoints were evaluated, and statistical analysis was conducted based on patient response rates. RESULTS The BELIEVE trial offered treatment opportunities for patients with relapse/refractory disease who lacked standard therapies or clinical trial options. This study addresses unmet medical needs and contributes to the establishment of precision medicine systems. Similar trials like NCI-MATCH and TAPUR are being conducted globally. The BELIEVE trial provides a platform for off-label drug administration, collects essential clinical data, and contributes to drug approval applications. CONCLUSION The BELIEVE trial provides hope for patients with actionable genetic abnormalities by facilitating access to targeted therapies through off-label drug administration. It establishes a regulatory framework and promotes collaboration between industry and academia by expanding organ-specific and cross-organ biomarker-based treatments.
Collapse
Affiliation(s)
- Sae Ishimaru
- Research Management Division, Clinical Research Support Office, National Cancer Center Hospital, Tokyo, Japan
- Department of Pediatric Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Tatsunori Shimoi
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kuniko Sunami
- Department of Laboratory Medicine, National Cancer Center Hospital, Tokyo, Japan
| | - Miho Nakajima
- Department of Pediatric Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yayoi Ando
- Research Management Division, Clinical Research Support Office, National Cancer Center Hospital, Tokyo, Japan
| | - Natsuko Okita
- Research Management Division, Clinical Research Support Office, National Cancer Center Hospital, Tokyo, Japan
| | - Kenichi Nakamura
- Department of International Clinical Development/Clinical Research Support Office, National Cancer Center Hospital, Tokyo, Japan
| | - Taro Shibata
- Biostatistics Division, Center for Research Administration and Support, National Cancer Center, Tokyo, Japan
| | - Yasuhiro Fujiwara
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Noboru Yamamoto
- Department of Experimental Therapeutics, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| |
Collapse
|
26
|
Dakilah I, Harb A, Abu-Gharbieh E, El-Huneidi W, Taneera J, Hamoudi R, Semreen MH, Bustanji Y. Potential of CDC25 phosphatases in cancer research and treatment: key to precision medicine. Front Pharmacol 2024; 15:1324001. [PMID: 38313315 PMCID: PMC10834672 DOI: 10.3389/fphar.2024.1324001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/04/2024] [Indexed: 02/06/2024] Open
Abstract
The global burden of cancer continues to rise, underscoring the urgency of developing more effective and precisely targeted therapies. This comprehensive review explores the confluence of precision medicine and CDC25 phosphatases in the context of cancer research. Precision medicine, alternatively referred to as customized medicine, aims to customize medical interventions by taking into account the genetic, genomic, and epigenetic characteristics of individual patients. The identification of particular genetic and molecular drivers driving cancer helps both diagnostic accuracy and treatment selection. Precision medicine utilizes sophisticated technology such as genome sequencing and bioinformatics to elucidate genetic differences that underlie the proliferation of cancer cells, hence facilitating the development of customized therapeutic interventions. CDC25 phosphatases, which play a crucial role in governing the progression of the cell cycle, have garnered significant attention as potential targets for cancer treatment. The dysregulation of CDC25 is a characteristic feature observed in various types of malignancies, hence classifying them as proto-oncogenes. The proteins in question, which operate as phosphatases, play a role in the activation of Cyclin-dependent kinases (CDKs), so promoting the advancement of the cell cycle. CDC25 inhibitors demonstrate potential as therapeutic drugs for cancer treatment by specifically blocking the activity of CDKs and modulating the cell cycle in malignant cells. In brief, precision medicine presents a potentially fruitful option for augmenting cancer research, diagnosis, and treatment, with an emphasis on individualized care predicated upon patients' genetic and molecular profiles. The review highlights the significance of CDC25 phosphatases in the advancement of cancer and identifies them as promising candidates for therapeutic intervention. This statement underscores the significance of doing thorough molecular profiling in order to uncover the complex molecular characteristics of cancer cells.
Collapse
Affiliation(s)
- Ibraheem Dakilah
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Amani Harb
- Department of Basic Sciences, Faculty of Arts and Sciences, Al-Ahliyya Amman University, Amman, Jordan
| | - Eman Abu-Gharbieh
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Waseem El-Huneidi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Jalal Taneera
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Rifat Hamoudi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
| | - Mohammed H Semreen
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Yasser Bustanji
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- School of Pharmacy, The University of Jordan, Amman, Jordan
| |
Collapse
|
27
|
Guven DC, Aykan MB, Muglu H, Bayram E, Helvaci K, Dursun B, Celayir M, Chelebiyev E, Nayir E, Erman M, Sezer A, Urun Y, Demirci U, Er O, Disel U, Bilici A, Arslan C, Karadurmus N, Kilickap S. The efficacy of immunotherapy and chemoimmunotherapy in patients with advanced rare tumors: A Turkish oncology group (TOG) study. Cancer Med 2024; 13:e6869. [PMID: 38140782 PMCID: PMC10809296 DOI: 10.1002/cam4.6869] [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: 07/06/2023] [Revised: 12/06/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
INTRODUCTION The advances in immune checkpoint inhibitors (ICIs) were relatively slow in rare tumors. Therefore, we conducted a multi-center study evaluating the efficacy of ICI monotherapy and the combination of ICIs with chemotherapy (CT) in patients with advanced rare tumors. METHODS In this retrospective cohort study, we included 93 patients treated with ICIs for NCI-defined rare tumors from the 12 cancer centers in Turkey. The primary endpoints were the overall response (ORR) and disease control rate (DCR). RESULTS The cohort's median age was 56, and 53.8% of the patients were male. The most frequent diagnosis was sarcoma (29%), and 81.7% of the patients were previously treated with at least one line of systemic therapy in the advanced stage. The ORR and DCR were 36.8% and 63.2%, respectively. The germ cell tumors had the lowest ORR (0%), while the Merkel cell carcinoma had the highest ORR to ICIs (57.1%). Patients treated with ICI + ICI or ICI plus chemotherapy combinations had higher ORR (55.2% vs. 27.6%, p = 0.012) and DCR (82.8% vs. 53.4%, p = 0.008). The median OS was 13.47 (95% CI: 7.79-19.15) months, and the six and 12-month survival rates were 71% and 52%. The median duration of response was 16.59 months, and the 12-month progression-free survival rate was 66% in responders. The median time-to-treatment failure was 5.06 months (95% CI: 3.42-6.71). Three patients had high-grade irAEs with ICIs (grade 3 colitis, grade 3 gastritis, and grade 3 encephalitis in one patient each). CONCLUSION We observed over 30% ORR and a 13-month median OS in patients with rare cancers treated with ICI monotherapy or ICI plus CT combinations. The response rates to ICIs or ICIs plus CT significantly varied across different tumor types. Responding patients had over 2 years of survival, highlighting a need for further trials with ICIs for patients with rare tumors.
Collapse
Affiliation(s)
- Deniz Can Guven
- Department of Medical OncologyHacettepe University Cancer InstituteAnkaraTurkey
| | - Musa Baris Aykan
- Department of Medical OncologyGulhane School of Medicine, University of Health SciencesAnkaraTurkey
| | - Harun Muglu
- Istanbul Medipol University Faculty of MedicineIstanbulTurkey
| | - Ertugrul Bayram
- Department of Medical OncologyCukurova UniversityAdanaTurkey
| | | | - Bengü Dursun
- Department of Medical OncologyAnkara UniversityAnkaraTurkey
| | - Melisa Celayir
- Department of Medical OncologyMAA Acıbadem UniversityİstanbulTurkey
| | - Elvin Chelebiyev
- Department of Medical OncologyHacettepe University Cancer InstituteAnkaraTurkey
| | - Erdinc Nayir
- Department of Medical OncologyMersin Medical Park HospitalMersinTurkey
| | - Mustafa Erman
- Department of Medical OncologyHacettepe University Cancer InstituteAnkaraTurkey
| | - Ahmet Sezer
- Baskent University Adana HospitalAdanaTurkey
| | - Yuksel Urun
- Department of Medical OncologyAnkara UniversityAnkaraTurkey
| | | | - Ozlem Er
- Department of Medical OncologyMAA Acıbadem UniversityİstanbulTurkey
| | - Umut Disel
- Department of Medical OncologyAcibadem Adana HospitalAdanaTurkey
| | - Ahmet Bilici
- Istanbul Medipol University Faculty of MedicineIstanbulTurkey
| | - Cagatay Arslan
- Department of Medical OncologySchool of Medicine, Medical Park Hospital, Izmir Economy UniversityIzmirTurkey
| | - Nuri Karadurmus
- Department of Medical OncologyGulhane School of Medicine, University of Health SciencesAnkaraTurkey
| | - Saadettin Kilickap
- Department of Medical OncologyIstinye University Faculty of MedicineIstanbulTurkey
| |
Collapse
|
28
|
Sunami K, Naito Y, Saigusa Y, Amano T, Ennishi D, Imai M, Kage H, Kanai M, Kenmotsu H, Komine K, Koyama T, Maeda T, Morita S, Sakai D, Hirata M, Ito M, Kozuki T, Sakashita H, Horinouchi H, Okuma Y, Takashima A, Kubo T, Hironaka S, Segawa Y, Yakushijin Y, Bando H, Makiyama A, Suzuki T, Kinoshita I, Kohsaka S, Ohe Y, Ishioka C, Yamamoto K, Tsuchihara K, Yoshino T. A Learning Program for Treatment Recommendations by Molecular Tumor Boards and Artificial Intelligence. JAMA Oncol 2024; 10:95-102. [PMID: 38032680 PMCID: PMC10690580 DOI: 10.1001/jamaoncol.2023.5120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 08/17/2023] [Indexed: 12/01/2023]
Abstract
Importance Substantial heterogeneity exists in treatment recommendations across molecular tumor boards (MTBs), especially for biomarkers with low evidence levels; therefore, the learning program is essential. Objective To determine whether a learning program sharing treatment recommendations for biomarkers with low evidence levels contributes to the standardization of MTBs and to investigate the efficacy of an artificial intelligence (AI)-based annotation system. Design, Setting, and Participants This prospective quality improvement study used 50 simulated cases to assess concordance of treatment recommendations between a central committee and participants. Forty-seven participants applied from April 7 to May 13, 2021. Fifty simulated cases were randomly divided into prelearning and postlearning evaluation groups to assess similar concordance based on previous investigations. Participants included MTBs at hub hospitals, treating physicians at core hospitals, and AI systems. Each participant made treatment recommendations for each prelearning case from registration to June 30, 2021; participated in the learning program on July 18, 2021; and made treatment recommendations for each postlearning case from August 3 to September 30, 2021. Data were analyzed from September 2 to December 10, 2021. Exposures The learning program shared the methodology of making appropriate treatment recommendations, especially for biomarkers with low evidence levels. Main Outcomes and Measures The primary end point was the proportion of MTBs that met prespecified accreditation criteria for postlearning evaluations (approximately 90% concordance with high evidence levels and approximately 40% with low evidence levels). Key secondary end points were chronological enhancements in the concordance of treatment recommendations on postlearning evaluations from prelearning evaluations. Concordance of treatment recommendations by an AI system was an exploratory end point. Results Of the 47 participants who applied, 42 were eligible. The accreditation rate of the MTBs was 55.6% (95% CI, 35.3%-74.5%; P < .001). Concordance in MTBs increased from 58.7% (95% CI, 52.8%-64.4%) to 67.9% (95% CI, 61.0%-74.1%) (odds ratio, 1.40 [95% CI, 1.06-1.86]; P = .02). In postlearning evaluations, the concordance of treatment recommendations by the AI system was significantly higher than that of MTBs (88.0% [95% CI, 68.7%-96.1%]; P = .03). Conclusions and Relevance The findings of this quality improvement study suggest that use of a learning program improved the concordance of treatment recommendations provided by MTBs to central ones. Treatment recommendations made by an AI system showed higher concordance than that for MTBs, indicating the potential clinical utility of the AI system.
Collapse
Affiliation(s)
- Kuniko Sunami
- Department of Laboratory Medicine, National Cancer Center Hospital, Tokyo, Japan
| | - Yoichi Naito
- Departments of General Internal Medicine, Experimental Therapeutics, and Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yusuke Saigusa
- Department of Biostatistics, Yokohama City University, Yokohama, Japan
| | - Toraji Amano
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Japan
| | - Daisuke Ennishi
- Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama, Japan
| | - Mitsuho Imai
- Translational Research Support Section, National Cancer Center Hospital East, Kashiwa, Japan
- Genomics Unit, Keio University School of Medicine, Tokyo, Japan
| | - Hidenori Kage
- Next-Generation Precision Medicine Development Laboratory, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masashi Kanai
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Keigo Komine
- Department of Medical Oncology, Tohoku University Hospital, Sendai, Japan
| | - Takafumi Koyama
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Takahiro Maeda
- Division of Precision Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Sachi Morita
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Japan
| | - Daisuke Sakai
- Department of Medical Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Makoto Hirata
- Department of Genetic Medicine and Services, National Cancer Center Hospital, Tokyo, Japan
| | - Mamoru Ito
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Toshiyuki Kozuki
- Department of Thoracic Oncology and Medicine, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | | | - Hidehito Horinouchi
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yusuke Okuma
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Atsuo Takashima
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Toshio Kubo
- Center for Clinical Oncology, Okayama University Hopital, Okayama, Japan
| | - Shuichi Hironaka
- Department of Medical Oncology, Kyorin University Hospital, Mitaka, Japan
| | - Yoshihiko Segawa
- Department of Medical Oncology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Yoshihiro Yakushijin
- Department of Clinical Oncology, Ehime University Graduate School of Medicine, Toon, Japan
| | - Hideaki Bando
- Translational Research Support Section, National Cancer Center Hospital East, Kashiwa, Japan
| | | | - Tatsuya Suzuki
- Department of Hematology, National Cancer Center Hospital, Tokyo, Japan
| | - Ichiro Kinoshita
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Japan
| | - Shinji Kohsaka
- Section for Knowledge Integration, Center for Cancer Genomics and Advanced Therapeutics, National Cancer Center, Tokyo, Japan
| | - Yuichiro Ohe
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Chikashi Ishioka
- Department of Medical Oncology, Tohoku University Hospital, Sendai, Japan
| | - Kouji Yamamoto
- Department of Biostatistics, Yokohama City University, Yokohama, Japan
| | - Katsuya Tsuchihara
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center Japan, Kashiwa, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
29
|
Heudel P, Crochet H, Durand T, Zrounba P, Blay JY. From data strategy to implementation to advance cancer research and cancer care: A French comprehensive cancer center experience. PLOS DIGITAL HEALTH 2023; 2:e0000415. [PMID: 38113207 PMCID: PMC10729983 DOI: 10.1371/journal.pdig.0000415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023]
Abstract
In a comprehensive cancer center, effective data strategies are essential to evaluate practices, and outcome, understanding the disease and prognostic factors, identifying disparities in cancer care, and overall developing better treatments. To achieve these goals, the Center Léon Bérard (CLB) considers various data collection strategies, including electronic medical records (EMRs), clinical trial data, and research projects. Advanced data analysis techniques like natural language processing (NLP) can be used to extract and categorize information from these sources to provide a more complete description of patient data. Data sharing is also crucial for collaboration across comprehensive cancer centers, but it must be done securely and in compliance with regulations like GDPR. To ensure data is shared appropriately, CLB should develop clear data sharing policies and share data in a controlled, standardized format like OSIRIS RWD, OMOP and FHIR. The UNICANCER initiative has launched the CONSORE project to support the development of a structured and standardized repository of patient data to improve cancer research and patient outcomes. Real-world data (RWD) studies are vital in cancer research as they provide a comprehensive and accurate picture of patient outcomes and treatment patterns. By incorporating RWD into data collection, analysis, and sharing strategies, comprehensive cancer centers can take a more comprehensive and patient-centered approach to cancer research. In conclusion, comprehensive cancer centers must take an integrated approach to data collection, analysis, and sharing to enhance their understanding of cancer and improve patient outcomes. Leveraging advanced data analytics techniques and developing effective data sharing policies can help cancer centers effectively harness the power of data to drive progress in cancer research.
Collapse
Affiliation(s)
- Pierre Heudel
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Hugo Crochet
- Data and Artificial Intelligence Team, Centre Léon Bérard, Lyon, France
| | - Thierry Durand
- Data protection officer, Centre Léon Bérard, Lyon, France
| | - Philippe Zrounba
- Department of Surgical Oncology, Centre Léon Bérard, Lyon, France
| | - Jean-Yves Blay
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
- General Director, Centre Léon Bérard, Lyon, France
| |
Collapse
|
30
|
Tsimberidou AM, Kahle M, Vo HH, Baysal MA, Johnson A, Meric-Bernstam F. Molecular tumour boards - current and future considerations for precision oncology. Nat Rev Clin Oncol 2023; 20:843-863. [PMID: 37845306 DOI: 10.1038/s41571-023-00824-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2023] [Indexed: 10/18/2023]
Abstract
Over the past 15 years, rapid progress has been made in developmental therapeutics, especially regarding the use of matched targeted therapies against specific oncogenic molecular alterations across cancer types. Molecular tumour boards (MTBs) are panels of expert physicians, scientists, health-care providers and patient advocates who review and interpret molecular-profiling results for individual patients with cancer and match each patient to available therapies, which can include investigational drugs. Interpretation of the molecular alterations found in each patient is a complicated task that requires an understanding of their contextual functional effects and their correlations with sensitivity or resistance to specific treatments. The criteria for determining the actionability of molecular alterations and selecting matched treatments are constantly evolving. Therefore, MTBs have an increasingly necessary role in optimizing the allocation of biomarker-directed therapies and the implementation of precision oncology. Ultimately, increased MTB availability, accessibility and performance are likely to improve patient care. The challenges faced by MTBs are increasing, owing to the plethora of identifiable molecular alterations and immune markers in tumours of individual patients and their evolving clinical significance as more and more data on patient outcomes and results from clinical trials become available. Beyond next-generation sequencing, broader biomarker analyses can provide useful information. However, greater funding, resources and expertise are needed to ensure the sustainability of MTBs and expand their outreach to underserved populations. Harmonization between practice and policy will be required to optimally implement precision oncology. Herein, we discuss the evolving role of MTBs and current and future considerations for their use in precision oncology.
Collapse
Affiliation(s)
- Apostolia M Tsimberidou
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Michael Kahle
- Khalifa Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Henry Hiep Vo
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mehmet A Baysal
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amber Johnson
- Khalifa Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
31
|
Weiss L, Dorman K, Boukovala M, Schwinghammer F, Jordan P, Fey T, Hasselmann K, Subklewe M, Bücklein V, Bargou R, Goebeler M, Sayehli C, Spoerl S, Lüke F, Heudobler D, Claus R, von Luettichau I, Lorenzen S, Lange S, Westphalen CB, von Bergwelt-Baildon M, Heinemann V, Gießen-Jung C. Early clinical trial unit tumor board: a real-world experience in a national cancer network. J Cancer Res Clin Oncol 2023; 149:13383-13390. [PMID: 37490102 PMCID: PMC10587227 DOI: 10.1007/s00432-023-05196-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 07/17/2023] [Indexed: 07/26/2023]
Abstract
PURPOSE Early clinical trials are the first step into clinical therapies for new drugs. Within the six Bavarian university-based hospitals (Augsburg, Erlangen, Regensburg, Munich (LMU and TU), Würzburg) we have enrolled a virtual network platform for patient discussion. METHODS The virtual Early Clinical Trial Unit Tumor Board (ECTU Tumor Board) is a secured web-based meeting to evaluate early clinical trial options for patients, where representatives from local ECTUs participate. We retrospectively analyzed patient cases discussed between November 2021 and November 2022. RESULTS From November 2021 to November 2022, a total of 43 patients were discussed in the ECTU Tumor Board. Median age at diagnosis was 44.6 years (range 10-76 years). The median number of previous lines of therapies was 3.7 (range 1-9 therapies) including systemic treatment, surgery, and radiation therapy. A total of 27 different tumor entities were presented and 83.7% (36/43) patients received at least one trial recommendation. In total, 21 different active or shortly recruiting clinical trials were recommended: ten antibody trials, four BiTE (bispecific T cell engager) trials, six CAR (chimeric antigen receptor) T-cell trials, and one chemotherapy trial. Only six trials (28.6%) were recommended on the basis of the previously performed comprehensive genetic profiling (CGP). CONCLUSION The ECTU Tumor Board is a feasible and successful network, highlighting the force of virtual patient discussions for improving patient care as well as trial recruitment in advanced diseases. It can provide further treatment options after local MTB presentation, aiming to close the gap to access clinical trials.
Collapse
Affiliation(s)
- L Weiss
- Department Medicine III (Hematology and Oncology), LMU University Hospital Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - K Dorman
- Department Medicine III (Hematology and Oncology), LMU University Hospital Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - M Boukovala
- Department Medicine III (Hematology and Oncology), LMU University Hospital Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - F Schwinghammer
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - P Jordan
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - T Fey
- Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany
| | - K Hasselmann
- Department Medicine III (Hematology and Oncology), LMU University Hospital Munich, Munich, Germany
- Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany
| | - M Subklewe
- Department Medicine III (Hematology and Oncology), LMU University Hospital Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - V Bücklein
- Department Medicine III (Hematology and Oncology), LMU University Hospital Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - R Bargou
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Würzburg, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - M Goebeler
- Early Clinical Trials Unit, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - C Sayehli
- Early Clinical Trials Unit, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - S Spoerl
- Department of Internal Medicine 5 (Hematology and Clinical Oncology), Friedrich-Alexander-Universität Erlangen-Nürnberg, University Hospital Erlangen, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - F Lüke
- Department of Internal Medicine III (Hematology and Oncology), University Hospital Regensburg, Regensburg, Germany
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - D Heudobler
- Department of Internal Medicine III (Hematology and Oncology), University Hospital Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - R Claus
- Department of Hematology and Clinical Oncology, University Medical Center Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - I von Luettichau
- Department of Pediatrics and Children's Cancer Research Center, TUM School of Medicine, Kinderklinik München Schwabing, Technical University of Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - S Lorenzen
- Department of Medicine II (Gastroenterology), Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - S Lange
- Department of Medicine II (Gastroenterology), Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - C B Westphalen
- Department Medicine III (Hematology and Oncology), LMU University Hospital Munich, Munich, Germany
- Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - M von Bergwelt-Baildon
- Department Medicine III (Hematology and Oncology), LMU University Hospital Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - V Heinemann
- Department Medicine III (Hematology and Oncology), LMU University Hospital Munich, Munich, Germany
- Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - C Gießen-Jung
- Department Medicine III (Hematology and Oncology), LMU University Hospital Munich, Munich, Germany.
- Bavarian Cancer Research Center (BZKF), Munich, Germany.
| |
Collapse
|
32
|
Aldea M, Friboulet L, Apcher S, Jaulin F, Mosele F, Sourisseau T, Soria JC, Nikolaev S, André F. Precision medicine in the era of multi-omics: can the data tsunami guide rational treatment decision? ESMO Open 2023; 8:101642. [PMID: 37769400 PMCID: PMC10539962 DOI: 10.1016/j.esmoop.2023.101642] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 09/30/2023] Open
Abstract
Precision medicine for cancer is rapidly moving to an approach that integrates multiple dimensions of the biology in order to model mechanisms of cancer progression in each patient. The discovery of multiple drivers per tumor challenges medical decision that faces several treatment options. Drug sensitivity depends on the actionability of the target, its clonal or subclonal origin and coexisting genomic alterations. Sequencing has revealed a large diversity of drivers emerging at treatment failure, which are potential targets for clinical trials or drug repurposing. To effectively prioritize therapies, it is essential to rank genomic alterations based on their proven actionability. Moving beyond primary drivers, the future of precision medicine necessitates acknowledging the intricate spatial and temporal heterogeneity inherent in cancer. The advent of abundant complex biological data will make artificial intelligence algorithms indispensable for thorough analysis. Here, we will discuss the advancements brought by the use of high-throughput genomics, the advantages and limitations of precision medicine studies and future perspectives in this field.
Collapse
Affiliation(s)
- M Aldea
- Department of Medical Oncology, Gustave Roussy, Villejuif; PRISM, INSERM, Gustave Roussy, Villejuif.
| | | | - S Apcher
- PRISM, INSERM, Gustave Roussy, Villejuif
| | - F Jaulin
- PRISM, INSERM, Gustave Roussy, Villejuif
| | - F Mosele
- Department of Medical Oncology, Gustave Roussy, Villejuif; PRISM, INSERM, Gustave Roussy, Villejuif
| | | | - J-C Soria
- Paris Saclay University, Orsay; Drug Development Department, Gustave Roussy, Villejuif, France
| | - S Nikolaev
- PRISM, INSERM, Gustave Roussy, Villejuif
| | - F André
- Department of Medical Oncology, Gustave Roussy, Villejuif; PRISM, INSERM, Gustave Roussy, Villejuif; Paris Saclay University, Orsay
| |
Collapse
|
33
|
Pham H, Dixon E. Integration of Next-Generation Sequencing in the Surgical Management of Colorectal Liver Metastasis. Ann Surg Oncol 2023; 30:6815-6823. [PMID: 37316745 DOI: 10.1245/s10434-023-13750-7] [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: 05/27/2022] [Accepted: 06/04/2023] [Indexed: 06/16/2023]
Abstract
Hepatic resection remains the treatment of choice for colorectal liver metastases. The advancement of surgical technique and use of perioperative systemic therapy has expanded the number and complexity of patients eligible for surgical resection. In recent years, investigation into gene mutations, such as RAS/RAF pathway, have led to targeted therapies that have significantly improved outcomes. Next-generation sequencing allows analysis of large number of genes that may have potential prognostic relevance in the clinical setting. This review summarizes the current applications of next-generation sequencing technology in metastatic colorectal cancer, focusing on its prognostic implications on patient management.
Collapse
Affiliation(s)
- Helen Pham
- Department of Surgery, Faculty of Medicine, University of Calgary, Foothills Medical Centre, Calgary, AB, Canada.
| | - Elijah Dixon
- Department of Surgery, Faculty of Medicine, University of Calgary, Foothills Medical Centre, Calgary, AB, Canada
| |
Collapse
|
34
|
Tarabay A, Boileve A, Smolenschi C, Antoun L, Valery M, Fuerea A, Perret A, Burtin P, Cosconea S, Belkhodja H, Malka D, Boige V, Hollebecque A, Ducreux M. Precision Medicine in Pancreatic Ductal Adenocarcinoma: The Impact of Targeted Therapies on Survival of Patients Harboring Actionable Mutations. Biomedicines 2023; 11:2569. [PMID: 37761010 PMCID: PMC10526242 DOI: 10.3390/biomedicines11092569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of death by cancer worldwide. Mostly diagnosed with locally advanced or metastatic disease, patients lack treatment options. Gene alterations (GAs) are frequently observed in PDAC, some of which are considered for molecular targeted therapies (MTTs), with potential clinical benefits and improved outcomes. However, the applicability of molecular profiling (MP) for precision medicine in PDAC remains to be demonstrated. METHODS We conducted a retrospective analysis of all patients, aged ≥18 years with histologically confirmed PDAC, who underwent tumor MP between 2010 and 2020 in our institution as part of personalized medicine trials. The primary study endpoint was overall survival (OS), and (minimal follow-up was 6 months after MP). RESULTS Of 115 eligible patients, MP was successful in 102 patients (89%). KRAS mutations were the most frequent GAs, mostly G12D. Based on ESCAT classification, actionable GAs were found in 29 patients (28%), involving mainly BRCA1 or BRCA2 (5 (18%)), HER2 (5 (18%)), MTAP (5 (18%)), and FGFR (3 (11%)). Only 12 of these 29 patients (41%, or 10% of the whole population) received MTTs, with a median progression-free survival of 1.6 months. Median OS was 19 months in patients with actionable GAs treated with MTTs (n = 12 (11.8%)), 14 months in patients with actionable GAs treated with standard therapies (n = 17 (16.7%)), and 17 months in patients without actionable GAs treated with standard therapies (n = 73 (71.5%); p = 0.26). The absence of liver metastases was associated with better OS (HR = 0.471, p = 0.01). The highest OS following MTT was observed in patients with BRCA mutations treated with olaparib. INTERPRETATION Actionable GAs were found in more than a quarter of patients with advanced PDAC. Overall, targeting actionable GAs with MTTs was not associated with improved OS in this retrospective study with limited patient numbers. However, selected GA/MTT combinations (e.g., BRCA mutations/olaparib) were associated with a better outcome.
Collapse
Affiliation(s)
- Anthony Tarabay
- Gustave Roussy, Département de Médecine, 94805 Villejuif, France; (A.B.); (C.S.); (L.A.); (M.V.); (A.F.); (A.P.); (P.B.); (S.C.); (H.B.); (D.M.); (V.B.); (M.D.)
| | - Alice Boileve
- Gustave Roussy, Département de Médecine, 94805 Villejuif, France; (A.B.); (C.S.); (L.A.); (M.V.); (A.F.); (A.P.); (P.B.); (S.C.); (H.B.); (D.M.); (V.B.); (M.D.)
| | - Cristina Smolenschi
- Gustave Roussy, Département de Médecine, 94805 Villejuif, France; (A.B.); (C.S.); (L.A.); (M.V.); (A.F.); (A.P.); (P.B.); (S.C.); (H.B.); (D.M.); (V.B.); (M.D.)
| | - Leony Antoun
- Gustave Roussy, Département de Médecine, 94805 Villejuif, France; (A.B.); (C.S.); (L.A.); (M.V.); (A.F.); (A.P.); (P.B.); (S.C.); (H.B.); (D.M.); (V.B.); (M.D.)
| | - Marine Valery
- Gustave Roussy, Département de Médecine, 94805 Villejuif, France; (A.B.); (C.S.); (L.A.); (M.V.); (A.F.); (A.P.); (P.B.); (S.C.); (H.B.); (D.M.); (V.B.); (M.D.)
| | - Alina Fuerea
- Gustave Roussy, Département de Médecine, 94805 Villejuif, France; (A.B.); (C.S.); (L.A.); (M.V.); (A.F.); (A.P.); (P.B.); (S.C.); (H.B.); (D.M.); (V.B.); (M.D.)
| | - Audrey Perret
- Gustave Roussy, Département de Médecine, 94805 Villejuif, France; (A.B.); (C.S.); (L.A.); (M.V.); (A.F.); (A.P.); (P.B.); (S.C.); (H.B.); (D.M.); (V.B.); (M.D.)
| | - Pascal Burtin
- Gustave Roussy, Département de Médecine, 94805 Villejuif, France; (A.B.); (C.S.); (L.A.); (M.V.); (A.F.); (A.P.); (P.B.); (S.C.); (H.B.); (D.M.); (V.B.); (M.D.)
| | - Simona Cosconea
- Gustave Roussy, Département de Médecine, 94805 Villejuif, France; (A.B.); (C.S.); (L.A.); (M.V.); (A.F.); (A.P.); (P.B.); (S.C.); (H.B.); (D.M.); (V.B.); (M.D.)
| | - Hichem Belkhodja
- Gustave Roussy, Département de Médecine, 94805 Villejuif, France; (A.B.); (C.S.); (L.A.); (M.V.); (A.F.); (A.P.); (P.B.); (S.C.); (H.B.); (D.M.); (V.B.); (M.D.)
| | - David Malka
- Gustave Roussy, Département de Médecine, 94805 Villejuif, France; (A.B.); (C.S.); (L.A.); (M.V.); (A.F.); (A.P.); (P.B.); (S.C.); (H.B.); (D.M.); (V.B.); (M.D.)
| | - Valérie Boige
- Gustave Roussy, Département de Médecine, 94805 Villejuif, France; (A.B.); (C.S.); (L.A.); (M.V.); (A.F.); (A.P.); (P.B.); (S.C.); (H.B.); (D.M.); (V.B.); (M.D.)
| | - Antoine Hollebecque
- Gustave Roussy, Département d’Innovation Thérapeutique et d’Essais Précoces, 94805 Villejuif, France;
| | - Michel Ducreux
- Gustave Roussy, Département de Médecine, 94805 Villejuif, France; (A.B.); (C.S.); (L.A.); (M.V.); (A.F.); (A.P.); (P.B.); (S.C.); (H.B.); (D.M.); (V.B.); (M.D.)
- Faculty of Medicine, Université Paris Saclay, 91400 Orsay, France
| |
Collapse
|
35
|
Rinaldi L, Guerini Rocco E, Spitaleri G, Raimondi S, Attili I, Ranghiero A, Cammarata G, Minotti M, Lo Presti G, De Piano F, Bellerba F, Funicelli G, Volpe S, Mora S, Fodor C, Rampinelli C, Barberis M, De Marinis F, Jereczek-Fossa BA, Orecchia R, Rizzo S, Botta F. Association between Contrast-Enhanced Computed Tomography Radiomic Features, Genomic Alterations and Prognosis in Advanced Lung Adenocarcinoma Patients. Cancers (Basel) 2023; 15:4553. [PMID: 37760521 PMCID: PMC10527057 DOI: 10.3390/cancers15184553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Non-invasive methods to assess mutational status, as well as novel prognostic biomarkers, are warranted to foster therapy personalization of patients with advanced non-small cell lung cancer (NSCLC). This study investigated the association of contrast-enhanced Computed Tomography (CT) radiomic features of lung adenocarcinoma lesions, alone or integrated with clinical parameters, with tumor mutational status (EGFR, KRAS, ALK alterations) and Overall Survival (OS). In total, 261 retrospective and 48 prospective patients were enrolled. A Radiomic Score (RS) was created with LASSO-Logistic regression models to predict mutational status. Radiomic, clinical and clinical-radiomic models were trained on retrospective data and tested (Area Under the Curve, AUC) on prospective data. OS prediction models were trained and tested on retrospective data with internal cross-validation (C-index). RS significantly predicted each alteration at training (radiomic and clinical-radiomic AUC 0.95-0.98); validation performance was good for EGFR (AUC 0.86), moderate for KRAS and ALK (AUC 0.61-0.65). RS was also associated with OS at univariate and multivariable analysis, in the latter with stage and type of treatment. The validation C-index was 0.63, 0.79, and 0.80 for clinical, radiomic, and clinical-radiomic models. The study supports the potential role of CT radiomics for non-invasive identification of gene alterations and prognosis prediction in patients with advanced lung adenocarcinoma, to be confirmed with independent studies.
Collapse
Affiliation(s)
- Lisa Rinaldi
- Radiation Research Unit, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy;
| | - Elena Guerini Rocco
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (E.G.R.); (A.R.); (M.B.)
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy; (S.V.)
| | - Gianluca Spitaleri
- Division of Thoracic Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (G.S.); (I.A.); (F.D.M.)
| | - Sara Raimondi
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy (F.B.)
| | - Ilaria Attili
- Division of Thoracic Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (G.S.); (I.A.); (F.D.M.)
| | - Alberto Ranghiero
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (E.G.R.); (A.R.); (M.B.)
| | - Giulio Cammarata
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy (F.B.)
| | - Marta Minotti
- Division of Radiology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.M.); (C.R.); (R.O.)
| | - Giuliana Lo Presti
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy (F.B.)
| | - Francesca De Piano
- Division of Radiology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.M.); (C.R.); (R.O.)
| | - Federica Bellerba
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy (F.B.)
| | - Gianluigi Funicelli
- Division of Radiology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.M.); (C.R.); (R.O.)
| | - Stefania Volpe
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy; (S.V.)
- Department of Radiation Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Serena Mora
- Data Management Unit, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (S.M.); (C.F.)
| | - Cristiana Fodor
- Data Management Unit, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (S.M.); (C.F.)
| | - Cristiano Rampinelli
- Division of Radiology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.M.); (C.R.); (R.O.)
| | - Massimo Barberis
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (E.G.R.); (A.R.); (M.B.)
| | - Filippo De Marinis
- Division of Thoracic Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (G.S.); (I.A.); (F.D.M.)
| | - Barbara Alicja Jereczek-Fossa
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy; (S.V.)
- Department of Radiation Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Roberto Orecchia
- Division of Radiology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.M.); (C.R.); (R.O.)
- Scientific Direction, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Stefania Rizzo
- Clinica di Radiologia EOC, Istituto Imaging della Svizzera Italiana (IIMSI), Via Tesserete 46, 6900 Lugano, Switzerland;
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Via G. Buffi 13, 6900 Lugano, Switzerland
| | - Francesca Botta
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy;
| |
Collapse
|
36
|
Leroy K, Audigier Valette C, Alexandre J, Boussemart L, Chiesa J, Deldycke C, Gomez-Rocca C, Hollebecque A, Lehmann-Che J, Lemoine A, Mansard S, Medioni J, Monnet I, Mourah S, Pierret T, Spaëth D, Civet A, Galoin S, Italiano A. Retrospective analysis of real-world data to evaluate actionability of a comprehensive molecular profiling panel in solid tumor tissue samples (REALM study). PLoS One 2023; 18:e0291495. [PMID: 37708140 PMCID: PMC10501576 DOI: 10.1371/journal.pone.0291495] [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: 01/10/2023] [Accepted: 08/23/2023] [Indexed: 09/16/2023] Open
Abstract
INTRODUCTION Considering the growing interest in matched cancer treatment, our aim was to evaluate the ability of a comprehensive genomic profiling (CGP) assay to propose at least one targeted therapy given an identified genomic alteration or signature (actionability), and to collect the treatment modifications based on the CGP test results in clinical practise for solid tumors. METHODS This retrospective, multicentre French study was conducted among 25 centres that participated in a free of charge program between 2017 and 2019 for a tissue CGP test. Data were collected on the patient, disease, tumor genomic profile, treatment suggested in the report (related to the genomic profile results) and subsequent therapeutic decisions according to the physician's declaration. RESULTS Among the 416 patients, most had lung cancer (35.6%), followed by biliary tract cancer (11.5%) or rare cancers (11.1%); 75% had a metastatic disease. The actionability was 75.0% (95% CI [70.6%-78.9%]) for all patients, 85.1% and 78.4%, respectively in lung cancer and metastatic patients. After exclusion of clinical trial suggestions, the actionability decreased to 62.3% (95% CI [57.5%-66.8%]). Treatment modification based on the test results was observed in 17.3% of the patients and was more frequent in metastatic disease (OR = 2.73, 95% CI [1.31-5.71], p = 0.007). The main reasons for no treatment modification were poor general condition (33.2%) and stable disease or remission (30.2%). The genomic-directed treatment changes were performed mostly during the first six months after the CGP test, and interestingly a substantial part was observed from six to 24 months after the genomic profiling. CONCLUSION This French study provides information on the real-life actionability of a CGP test based on tissue samples, and trends to confirm its utility in clinical practice across the course of the disease, in particularly for patients with lung cancer and/or advanced disease.
Collapse
Affiliation(s)
- Karen Leroy
- Université Paris Cité, Sorbonne Université, Inserm, Centre de Recherche des Cordeliers, Paris, France
- Département de Médecine Génomique des Tumeurs et Cancers, Service de Biochimie, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | | | - Jérôme Alexandre
- Université Paris Cité, Sorbonne Université, Inserm, Centre de Recherche des Cordeliers, Paris, France
- Service d’Oncologie, AP-HP, Hôpital Cochin, Paris, France
| | - Lise Boussemart
- Service de Dermatologie, CHU de Nantes—Hôtel Dieu, Nantes, France
| | - Jean Chiesa
- UF de Cytogénétique et Génétique Médicale, Hôpital Universitaire Carémeau, Nîmes, France
| | | | | | | | - Jacqueline Lehmann-Che
- Université Paris Cité, INSERM U976, Immunologie Humaine, Pathophysiologie, Immunothérapie (HIPI), Paris, France
- UF Oncologie Moléculaire, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Antoinette Lemoine
- Biochimie et Oncogénétique–Inserm UMRS 1193, Hôpital Paul Brousse, AP-HP, Paris, France
| | | | - Jacques Medioni
- Centre d’Essais Précoces en Cancérologie, Hôpital Européen Georges Pompidou, Paris, France
| | - Isabelle Monnet
- Service de Pneumologie, Hôpital Intercommunal de Créteil, Créteil, France
| | - Samia Mourah
- Université Paris Cité, INSERM U976, Immunologie Humaine, Pathophysiologie, Immunothérapie (HIPI), Paris, France
- Service de Génomique des Tumeurs et Pharmacologie, Hôpital Saint-Louis, AP-HP, Paris, France
| | | | - Dominique Spaëth
- Centre d’Oncologie de Gentilly, Institut Interrégional de Cancérologie, Nancy, France
| | - Alexandre Civet
- Centre de Données Médicales, Roche S.A.S, Boulogne-Billancourt, France
| | - Sandrine Galoin
- Affaires Médicales, Roche S.A.S, Boulogne-Billancourt, France
| | - Antoine Italiano
- Unité d’études de Phases Précoces, Institut Bergonié, Bordeaux, France
| |
Collapse
|
37
|
Kerle I, Heining C. [Histology-agnostic tumor treatment - a farewell to tumor entities?]. Dtsch Med Wochenschr 2023; 148:1174-1181. [PMID: 37657455 DOI: 10.1055/a-1933-8141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/03/2023]
Abstract
Considerable efforts concerning the molecular characterization and targeted treatment of cancer have significantly improved treatment options and prognosis of tumor patients. Nevertheless, in tumor entities without recurrent genetic alterations the application of molecular testing for potentially targetable lesions remains heterogeneous and, in most cases, the approval of targeted therapies is still restricted to defined tumor entities harboring corresponding predictive biomarkers.The broad genomic analysis of different tumor entities including rare cancers within several genome sequencing initiatives and precision oncology programs has revealed the occurrence of addressable molecular alterations across many tumor entities, although their incidence may differ significantly in the context of the underlying cancer type. The treatment of molecularly defined patient cohorts demonstrated an impressive tumor-agnostic efficacy of certain therapeutics such as NTRK inhibitors, while the outcome of other targeted therapies, such as ERBB or BRAF inhibitors, varied in the context of the underlying disease.In the meantime, a handful targeted therapeutics addressing NRTK and RET fusions, the BRAF V600E mutation or different features of defective DNA mismatch repair and high tumor mutational burden has been approved for histology-agnostic treatment of tumors harboring these target lesions. Ongoing molecularly stratified basket trials will further investigate the tumor-agnostic efficacy of different targeted treatment approaches.
Collapse
|
38
|
Matsubara J, Mukai K, Kondo T, Yoshioka M, Kage H, Oda K, Kudo R, Ikeda S, Ebi H, Muro K, Hayashi R, Tokudome N, Yamamoto N, Muto M. First-Line Genomic Profiling in Previously Untreated Advanced Solid Tumors for Identification of Targeted Therapy Opportunities. JAMA Netw Open 2023; 6:e2323336. [PMID: 37459099 DOI: 10.1001/jamanetworkopen.2023.23336] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/20/2023] Open
Abstract
IMPORTANCE Precision oncology using comprehensive genomic profiling (CGP) by next-generation sequencing is aimed at companion diagnosis and genomic profiling. The clinical utility of CGP before the standard of care (SOC) is still not resolved, and more evidence is needed. OBJECTIVE To investigate the clinical utility of next-generation CGP (FoundationOne CDx [F1CDx]) in patients with previously untreated metastatic or recurrent solid tumors. DESIGN, Setting, and Participants This multicenter, prospective, observational cohort study enrolled patients with previously untreated advanced solid tumors between May 18, 2021, and February 16, 2022, with follow-up through August 16, 2022. The study was conducted at 6 hospitals in Japan. Eligible patients were aged 20 years or older and had Eastern Cooperative Oncology Group performance status of 0 to 1 with previously untreated metastatic or recurrent cancers in the gastrointestinal or biliary tract; pancreas, lung, breast, uterus, or ovary; and malignant melanoma. EXPOSURE Comprehensive genomic profiling testing before SOC for advanced solid tumors. MAIN OUTCOMES AND MEASURES Proportion of patients with actionable or druggable genomic alterations and molecular-based recommended therapy (MBRT). RESULTS A total of 183 patients met the inclusion criteria and 180 patients (92 men [51.1%]) with a median age of 64 years (range, 23-88 years) subsequently underwent CGP (lung [n = 28], colon/small intestine [n = 27], pancreas [n = 27], breast [n = 25], biliary tract [n = 20], gastric [n = 19], uterus [n = 12], esophagus [n = 10], ovary [n = 6], and skin melanoma [n = 6]). Data from 172 patients were available for end point analyses. Actionable alterations were found in 172 patients (100.0%; 95% CI, 97.9%-100.0%) and druggable alternations were identified in 109 patients (63.4%; 95% CI, 55.7%-70.6%). The molecular tumor board identified MBRT for 105 patients (61.0%; 95% CI, 53.3%-68.4%). Genomic alterations included in the companion diagnostics list of the CGP test were found in 49 patients (28.5%; 95% CI, 21.9%-35.9%) in a tumor-agnostic setting. After a median follow-up of 7.9 months (range, 0.5-13.2 months), 34 patients (19.8%; 95% CI, 14.1%-26.5%) received MBRT. CONCLUSIONS AND RELEVANCE The findings of this study suggest that CGP testing before SOC for patients with advanced solid tumors may be clinically beneficial to guide the subsequent anticancer therapies, including molecularly matched treatments.
Collapse
Affiliation(s)
- Junichi Matsubara
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Kumi Mukai
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Tomohiro Kondo
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Masahiro Yoshioka
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Hidenori Kage
- Department of Clinical Genomics, The University of Tokyo Hospital, Tokyo, Japan
| | - Katsutoshi Oda
- Department of Clinical Genomics, The University of Tokyo Hospital, Tokyo, Japan
| | - Ryo Kudo
- Department of Precision Cancer Medicine, Tokyo Medical and Dental University Hospital, Tokyo, Japan
| | - Sadakatsu Ikeda
- Department of Precision Cancer Medicine, Tokyo Medical and Dental University Hospital, Tokyo, Japan
| | - Hiromichi Ebi
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center, Nagoya, Japan
| | - Ryuji Hayashi
- Department of Clinical Oncology, Toyama University Hospital, Toyama, Japan
| | - Nahomi Tokudome
- Internal Medicine III, Wakayama Medical University, Wakayama, Japan
| | | | - Manabu Muto
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| |
Collapse
|
39
|
Vingiani A, Agnelli L, Duca M, Lorenzini D, Damian S, Proto C, Niger M, Nichetti F, Tamborini E, Perrone F, Piccolo A, Manoukian S, Azzollini J, Brambilla M, Colombo E, Lopez S, Vernieri C, Marra F, Conca E, Busico A, Capone I, Bozzi F, Angelini M, Devecchi A, Salvatori R, De Micheli V, Baggi A, Pasini S, Jommi C, Ladisa V, Apolone G, De Braud F, Pruneri G. Molecular Tumor Board as a Clinical Tool for Converting Molecular Data Into Real-World Patient Care. JCO Precis Oncol 2023; 7:e2300067. [PMID: 37487147 PMCID: PMC10581623 DOI: 10.1200/po.23.00067] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/28/2023] [Accepted: 06/12/2023] [Indexed: 07/26/2023] Open
Abstract
PURPOSE The investigation of multiple molecular targets with next-generation sequencing (NGS) has entered clinical practice in oncology, yielding to a paradigm shift from the histology-centric approach to the mutational model for personalized treatment. Accordingly, most of the drugs recently approved in oncology are coupled to specific biomarkers. One potential tool for implementing the mutational model of precision oncology in daily practice is represented by the Molecular Tumor Board (MTB), a multidisciplinary team whereby molecular pathologists, biologists, bioinformaticians, geneticists, medical oncologists, and pharmacists cooperate to generate, interpret, and match molecular data with personalized treatments. PATIENTS AND METHODS Since May 2020, the institutional MTB set at Fondazione IRCCS Istituto Nazionale Tumori of Milan met weekly via teleconference to discuss molecular data and potential therapeutic options for patients with advanced/metastatic solid tumors. RESULTS Up to October 2021, among 1,996 patients evaluated, we identified >10,000 variants, 43.2% of which were functionally relevant (pathogenic or likely pathogenic). On the basis of functionally relevant variants, 711 patients (35.6%) were potentially eligible to targeted therapy according to European Society of Medical Oncology Scale for Clinical Actionability of Molecular Targets tiers, and 9.4% received a personalized treatment. Overall, larger NGS panels (containing >50 genes) significantly outperformed small panels (up to 50 genes) in detecting actionable gene targets across different tumor types. CONCLUSION Our real-world data provide evidence that MTB is a valuable tool for matching NGS data with targeted treatments, eventually implementing precision oncology in clinical practice.
Collapse
Affiliation(s)
- Andrea Vingiani
- Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Luca Agnelli
- Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Matteo Duca
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Daniele Lorenzini
- Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvia Damian
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Claudia Proto
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Monica Niger
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Federico Nichetti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
- Computational Oncology Group, Molecular Precision Oncology Program, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elena Tamborini
- Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Federica Perrone
- Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alberta Piccolo
- Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Siranoush Manoukian
- Unit of Medical Genetics, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Jacopo Azzollini
- Unit of Medical Genetics, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Marta Brambilla
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Elena Colombo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Salvatore Lopez
- Gynecologic Oncology, Department of Surgery, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Claudio Vernieri
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Francesca Marra
- Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elena Conca
- Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Adele Busico
- Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Iolanda Capone
- Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Fabio Bozzi
- Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marta Angelini
- Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Andrea Devecchi
- Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Rebecca Salvatori
- Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Precision Cancer Therapeutics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | | | - Anna Baggi
- Business Integration Partners S.p.A., Milan, Italy
| | | | - Claudio Jommi
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Vito Ladisa
- Hospital Pharmacy, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Giovanni Apolone
- Scientific Directorate, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Filippo De Braud
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Giancarlo Pruneri
- Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
40
|
Normanno N, De Luca A, Abate RE, Morabito A, Milella M, Tabbò F, Curigliano G, Masini C, Marchetti P, Pruneri G, Guarneri V, Frassineti GL, Fasola G, Adamo V, Daniele B, Berardi R, Feroce F, Maiello E, Pinto C. Current practice of genomic profiling of patients with advanced solid tumours in Italy: the Italian Register of Actionable Mutations (RATIONAL) study. Eur J Cancer 2023; 187:174-184. [PMID: 37167765 DOI: 10.1016/j.ejca.2023.03.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/23/2023] [Accepted: 03/23/2023] [Indexed: 04/03/2023]
Abstract
BACKGROUND The Italian Register of Actionable Mutations (RATIONAL) is a multicentric, observational study collecting next-generation sequencing (NGS)-based tumour profiling data of patients with advanced solid tumours. METHODS The study enrols patients who had available an NGS-based tumour profiling (Pathway-A) or undergo comprehensive genomic profiling (CGP) with FoundationOne CDx assays within the trial (Pathway-B). The primary endpoint was the rate of actionable mutations identified. RESULTS Sequencing data were available for 738 patients in Pathway-A (218) and -B (520). In Pathway-A, 154/218 (70.6%) tests were performed using NGS panels ≤52 genes, and genomic alterations (GAs) were found in 164/218 (75.2%) patients. In Pathway-B, CGP revealed GAs in 512/520 (98.5%) patients. Levels I/II/III actionable GAs according to the European Society of Medical Oncology Scale for Clinical Actionability of molecular Targets (ESCAT) were identified in 254/554 (45.8%) patients with non-small-cell lung cancer, cholangiocarcinoma, colorectal, gastric, pancreatic and breast cancer. The rate of patients with level I GAs was similar in Pathways A and B (69 versus 102). CGP in Pathway-B revealed a higher number of patients with level II/III GAs (99 versus 20) and potentially germline pathogenic/likely pathogenic variants (58 versus 15) as compared with standard testing in Pathway-A. In patients with cancer of unknown primary, CGP detected OncoKB levels 3B/4 GAs in 31/58 (53.4%) cases. Overall, 67/573 (11.7%) of patients received targeted therapy based on genomic testing. CONCLUSION The Italian Register of Actionable Mutations represents the first overview of genomic profiling in Italian current clinical practice and highlights the utility of CGP for identifying therapeutic targets in selected cancer patients.
Collapse
Affiliation(s)
- Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy.
| | - Antonella De Luca
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Riziero Esposito Abate
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Alessandro Morabito
- Thoracic Department, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Michele Milella
- U.O.C. Oncology, Azienda Ospedaliera Universitaria Integrata, University and Hospital Trust of Verona, Verona, Italy
| | - Fabrizio Tabbò
- Department of Oncology, Università degli Studi di Torino, AOU San Luigi, Orbassano, Italy
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milano, Milano, Italy; Division of Early Drug Development, European Institute of Oncology IRCCS, Milano, Italy
| | - Cristina Masini
- Medical Oncology, Comprehensive Cancer Centre IRCCS - AUSL Reggio Emilia, Reggio Emilia, Italy
| | - Paolo Marchetti
- Istituto Dermopatico dell'Immacolata (IDI-IRCCS), Roma, Italy
| | - Giancarlo Pruneri
- Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Oncology 2, Istituto Oncologico Veneto (IOV) IRCCS, Padova, Italy
| | - Giovanni L Frassineti
- Department of Medical Oncology-IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Gianpiero Fasola
- Department of Medical Oncology, University Hospital of Udine, Udine, Italy
| | - Vincenzo Adamo
- Scientific Direction Oncology Department, Papardo Hospital, Messina, Italy
| | | | | | - Florinda Feroce
- Surgical Pathology Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Evaristo Maiello
- Oncology Unit, Fondazione Casa Sollievo della Sofferenza IRCCS, San Giovanni Rotondo, Italy
| | - Carmine Pinto
- Medical Oncology, Comprehensive Cancer Centre IRCCS - AUSL Reggio Emilia, Reggio Emilia, Italy
| |
Collapse
|
41
|
Teuwen LA, Roets E, D’Hoore P, Pauwels P, Prenen H. Comprehensive Genomic Profiling and Therapeutic Implications for Patients with Advanced Cancers: The Experience of an Academic Hospital. Diagnostics (Basel) 2023; 13:1619. [PMID: 37175010 PMCID: PMC10177779 DOI: 10.3390/diagnostics13091619] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/30/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
Next-generation sequencing (NGS) can be used to detect tumor-specific genomic alterations. This retrospective single-center study aims to assess the application of an extensive NGS panel to identify actionable alterations and initiate matched targeted treatment for patients with advanced cancer. We analyzed genomic alterations in solid tumor biopsies from 464 patients with advanced cancer with the Foundation Medicine assay (FoundationOne®CDx). Therapeutic implications were determined using the Memorial Sloan Kettering Precision Oncology Knowledge Base (OncoKB) classification. The FoundationOne®CDx was successfully applied in 464/521 patients (89%). The most common altered genes were TP53 (61%), KRAS (20%), CDKN2A (20%), TERT (16%), and APC (16%). Among the 419 patients with successfully analyzed tumor mutational burden (TMB), 43 patients presented with a high TMB (≥10 mutations/megabase). Out of the 126 patients with an actionable target, 40 patients received matched treatment (32%) of which 17 were within a clinical trial. This study shows that the application of NGS is feasible in an academic center and increases the detection of actionable alterations and identification of patients eligible for targeted treatment or immunotherapy regardless of tumor histology. Strategies such as early referral for NGS, inclusion in clinical (basket) trials, and the development of new targeted drugs are necessary to improve the matched treatment rate.
Collapse
Affiliation(s)
- Laure-Anne Teuwen
- Department of Oncology, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium; (L.-A.T.); (E.R.); (P.D.)
| | - Evelyne Roets
- Department of Oncology, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium; (L.-A.T.); (E.R.); (P.D.)
| | - Pieter D’Hoore
- Department of Oncology, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium; (L.-A.T.); (E.R.); (P.D.)
| | - Patrick Pauwels
- Department of Pathology, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium;
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Hans Prenen
- Department of Oncology, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium; (L.-A.T.); (E.R.); (P.D.)
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| |
Collapse
|
42
|
Scheiter A, Hierl F, Lüke F, Keil F, Heudobler D, Einhell S, Klier-Richter M, Konstandin NP, Weber F, Scheiter A, Kandulski A, Schlosser S, Cosma LS, Tews H, Weiss ARR, Grube M, Bumes E, Hau P, Proescholdt M, Steger F, Troeger A, Haferkamp S, Reibenspies LE, Schnabel MJ, Schulz C, Drexler K, Hatzipanagiotou ME, Seitz S, Klinkhammer-Schalke M, Unberath P, Calvisi DF, Pukrop T, Dietmaier W, Evert M, Utpatel K. Critical evaluation of molecular tumour board outcomes following 2 years of clinical practice in a Comprehensive Cancer Centre. Br J Cancer 2023; 128:1134-1147. [PMID: 36572733 PMCID: PMC10006213 DOI: 10.1038/s41416-022-02120-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Recently, molecular tumour boards (MTBs) have been integrated into the clinical routine. Since their benefit remains debated, we assessed MTB outcomes in the Comprehensive Cancer Center Ostbayern (CCCO) from 2019 to 2021. METHODS AND RESULTS In total, 251 patients were included. Targeted sequencing was performed with PCR MSI-evaluation and immunohistochemistry for PD-L1, Her2, and mismatch repair enzymes. 125 treatment recommendations were given (49.8%). High-recommendation rates were achieved for intrahepatic cholangiocarcinoma (20/30, 66.7%) and gastric adenocarcinoma (10/16, 62.5%) as opposed to colorectal cancer (9/36, 25.0%) and pancreatic cancer (3/18, 16.7%). MTB therapies were administered in 47 (18.7%) patients, while 53 (21.1%) received alternative treatment regimens. Thus 37.6% of recommended MTB therapies were implemented (47/125 recommendations). The clinical benefit rate (complete + partial + mixed response + stable disease) was 50.0% for MTB and 63.8% for alternative treatments. PFS2/1 ratios were 34.6% and 16.1%, respectively. Significantly improved PFS could be achieved for m1A-tier-evidence-based MTB therapies (median 6.30 months) compared to alternative treatments (median 2.83 months; P = 0.0278). CONCLUSION The CCCO MTB yielded a considerable recommendation rate, particularly in cholangiocarcinoma patients. The discrepancy between the low-recommendation rates in colorectal and pancreatic cancer suggests the necessity of a weighted prioritisation of entities. High-tier recommendations should be implemented predominantly.
Collapse
Affiliation(s)
- Alexander Scheiter
- Institute of Pathology, University of Regensburg, 93053, Regensburg, Germany. .,Bavarian Center for Cancer Research / BZKF, Regensburg, Bavaria, Germany.
| | - Frederik Hierl
- Institute of Pathology, University of Regensburg, 93053, Regensburg, Germany
| | - Florian Lüke
- Bavarian Center for Cancer Research / BZKF, Regensburg, Bavaria, Germany.,Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, 93053, Regensburg, Germany.,Fraunhofer-Institut für Toxikologie und Experimentelle Medizin ITEM-R, Abteilung für personalisierte Onkologie, 93053, Regensburg, Germany
| | - Felix Keil
- Institute of Pathology, University of Regensburg, 93053, Regensburg, Germany
| | - Daniel Heudobler
- Bavarian Center for Cancer Research / BZKF, Regensburg, Bavaria, Germany.,Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Sabine Einhell
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, 93053, Regensburg, Germany
| | | | - Nikola P Konstandin
- Institute of Pathology, University of Regensburg, 93053, Regensburg, Germany.,Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Florian Weber
- Institute of Pathology, University of Regensburg, 93053, Regensburg, Germany
| | - Andrea Scheiter
- School of Engineering and Design, Chair of Ergonomics, Technical University of Munich, 85748, Garching, Germany
| | - Arne Kandulski
- Department of Internal Medicine I, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Sophie Schlosser
- Department of Internal Medicine I, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Lidia-Sabina Cosma
- Department of Internal Medicine I, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Hauke Tews
- Department of Internal Medicine I, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Andreas R R Weiss
- Department of Surgery, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Matthias Grube
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Elisabeth Bumes
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, 93053 Regensburg University Hospital, 93053, Regensburg, Germany
| | - Peter Hau
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, 93053 Regensburg University Hospital, 93053, Regensburg, Germany
| | - Martin Proescholdt
- Department of Neurosurgery, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Felix Steger
- Department of Radiotherapy, Regensburg University Medical Center, 93053, Regensburg, Germany
| | - Anja Troeger
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital of Regensburg, 93053, Regensburg, Germany
| | - Sebastian Haferkamp
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Lucas E Reibenspies
- Institute of Pathology, University of Regensburg, 93053, Regensburg, Germany
| | - Marco J Schnabel
- Department of Urology, Caritas St. Josef Medical Center, University of Regensburg, 93053, Regensburg, Germany
| | - Christian Schulz
- Department of Pneumology, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Konstantin Drexler
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Maria E Hatzipanagiotou
- University Medical Centre Regensburg, Department of Gynecology and Obstetrics, 93053, Regensburg, Germany
| | - Stephan Seitz
- University Medical Centre Regensburg, Department of Gynecology and Obstetrics, 93053, Regensburg, Germany
| | - Monika Klinkhammer-Schalke
- Tumour Center-Institute for Quality Management and Health Services Research, University of Regensburg, 93053, Regensburg, Germany
| | - Philipp Unberath
- Friedrich-Alexander University Erlangen-Nuremberg, Chair of Medical Informatics, 91054, Erlangen, Germany
| | - Diego F Calvisi
- Institute of Pathology, University of Regensburg, 93053, Regensburg, Germany
| | - Tobias Pukrop
- Bavarian Center for Cancer Research / BZKF, Regensburg, Bavaria, Germany.,Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, 93053, Regensburg, Germany.,Fraunhofer-Institut für Toxikologie und Experimentelle Medizin ITEM-R, Abteilung für personalisierte Onkologie, 93053, Regensburg, Germany
| | - Wolfgang Dietmaier
- Institute of Pathology, University of Regensburg, 93053, Regensburg, Germany
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, 93053, Regensburg, Germany
| | - Kirsten Utpatel
- Institute of Pathology, University of Regensburg, 93053, Regensburg, Germany
| |
Collapse
|
43
|
Boilève A, Faron M, Fodil-Cherif S, Bayle A, Lamartina L, Planchard D, Tselikas L, Kanaan C, Scoazec JY, Ducreux M, Italiano A, Baudin E, Hadoux J. Molecular profiling and target actionability for precision medicine in neuroendocrine neoplasms: real-world data. Eur J Cancer 2023; 186:122-132. [PMID: 37062210 DOI: 10.1016/j.ejca.2023.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 04/08/2023]
Abstract
BACKGROUND Key molecular alterations (MA) of neuroendocrine neoplasm (NEN) of various grade/primaries have been described but the applicability of molecular profiling (MP) for precision medicine in NEN remains to be demonstrated. METHODS We conducted a retrospective study of all patients with metastatic NEN who had MP on tumour tissue at Gustave Roussy. The primary objective was to assess the clinical applicability of MP by evaluating the growth modulator index (GMI) as the primary end-point. RESULTS MPs were obtained in 114 out of 156 eligible patients, including 12% NET-G1, 42% NET-G2, 13% NET-G3 and 35% neuroendocrine carcinoma (NEC). Primary sites were lung/thymus (40%), pancreas (19%), gastro-intestinal (16%), head&neck (10%), unknown (10%) and others (10%) with synchronous metastases in 61% of the patients. Most frequent MA were: MEN1 (25%), PTEN (13%), TP53 (11%) and TSC2 (9%), in neuroendocrine tumour (NET), and TP53 (50%) and RB1 (18%) in NEC. ESMO Scale for Clinical Actionability of Molecular Targets (ESCAT) classification of these MA were: I(5%), III(20%), IV(23%), X(27%); a putative actionable MA was identified in 48% patients. Median TMB was 5.7 Mut/Mb, with 3 TMB > 10 and 1 MSI NET. No MA was found in 26% patients. Molecularly matched treatment was administered to 19 patients (4 NEC, 15 NET): immunotherapy (n = 3), tipifarnib (n = 1), NOTCHi (n = 1), EGFRi (n = 2), HER2i (n = 1) and everolimus (n = 11). Overall, 67% of patients had a clinical benefit defined as a GMI over 1.3 with a 78% disease control rate. CONCLUSION We report 48% of NEN with a putative actionable MA of which 35% received molecularly matched treatment, with a clinical benefit in 67% of the cases.
Collapse
|
44
|
Simons MJHG, Uyl-de Groot CA, Retèl VP, Mankor JM, Ramaekers BLT, Joore MA, van Harten WH. Cost-Effectiveness and Budget Impact of Future Developments With Whole-Genome Sequencing for Patients With Lung Cancer. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2023; 26:71-80. [PMID: 35973926 DOI: 10.1016/j.jval.2022.07.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 06/19/2022] [Accepted: 07/08/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVES This study aimed to investigate the cost-effectiveness, budget impact (BI), and impact of uncertainty of future developments concerning whole-genome sequencing (WGS) as a clinical diagnostic test compared with standard of care (SoC) in patients with locally advanced and metastatic non-small cell lung cancer. METHODS A total of 3 likely scenarios to take place within 5 years (according to experts) were simulated using a previously developed, peer reviewed, and published decision model. The scenarios concerned "WGS results used for treatment selection" (scenario 1), "WGS-based biomarker for immunotherapy" (scenario 2), and "off-label drug approval for WGS results" (scenario 3). Two diagnostic strategies of the original model, "SoC" and "WGS as a diagnostic test" (base model), were used to compare our scenarios with. Outcomes were reported for the base model, all scenarios separately, combined (combined unweighted), and weighted by likelihood (combined weighted). Cost-effectiveness, BI, and value of information analyses were performed for WGS compared with SoC. RESULTS Total costs and quality-adjusted life-years for SoC in metastatic non-small cell lung cancer were €149 698 and 1.235. Incremental outcomes of WGS were €1529/0.002(base model), -€222/0.020(scenario 1), -€2576/0.023(scenario 2), €388/0.024(scenario 3), -€5041/0.060(combined unweighted), and -€1715/0.029(combined weighted). The annual BI for adopting WGS for this population in The Netherlands ranged between €682 million (combined unweighted) and €714 million (base model). The consequences of uncertainty amounted to €3.4 million for all scenarios (combined weighted) and to €699 000 for the diagnostic yield of WGS alone (combined weighted). CONCLUSIONS Our findings suggest that it is likely for WGS to become cost-effective within the near future if it identifies more patients with actionable targets and show the impact of uncertainty regarding its diagnostic yield. Modeling future scenarios can be useful to consider early adoption of WGS while timely anticipating on unforeseen developments before final conclusions are reached.
Collapse
Affiliation(s)
- Martijn J H G Simons
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Centre, Maastricht, The Netherlands; Care And Public Health Research Institute, Maastricht University, Maastricht, The Netherlands
| | - Carin A Uyl-de Groot
- Erasmus School of Health Policy and Management/Institute for Medical Technology Assessment, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Valesca P Retèl
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek hospital, Amsterdam, The Netherlands; Department of Health Technology and Services Research, University of Twente, Enschede, The Netherlands
| | - Joanne M Mankor
- Department of Pulmonary Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Bram L T Ramaekers
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Centre, Maastricht, The Netherlands; Care And Public Health Research Institute, Maastricht University, Maastricht, The Netherlands
| | - Manuela A Joore
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Centre, Maastricht, The Netherlands; Care And Public Health Research Institute, Maastricht University, Maastricht, The Netherlands.
| | - Wim H van Harten
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek hospital, Amsterdam, The Netherlands; Department of Health Technology and Services Research, University of Twente, Enschede, The Netherlands
| |
Collapse
|
45
|
Debien V, Vignot S, Massard C, Malouf G, Hollebecque A, Scoazec JY, Michiels S, Verlingue L. Molecular analysis for refractory rare cancers: Sequencing battle continues - learnings for the MOSCATO-01 study. Crit Rev Oncol Hematol 2023; 181:103888. [PMID: 36460264 DOI: 10.1016/j.critrevonc.2022.103888] [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: 09/22/2022] [Revised: 11/18/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND For patients with metastatic rare cancers, treatments are limited. How systematic tumor sequencing can improve therapeutic possibilities in this population? PATIENTS AND METHODS Patients with rare cancer were identified in the MOSCATO-01 trial. Patients' outcome was measured by progression-free survival (PFS) and overall survival (OS). RESULTS The most frequently identified histologic subypes were ovarian adenocarcinoma (N = 13), carcinoma of unknown primary (N = 11), and leiomyosarcoma (N = 10). Ninety-nine (39%) of them had at least one targetable cancer molecular alteration Forty-nine patients (50%) received the therapy proposed by the molecular tumor board, and 13 patients (26%, 95%CI 15-41%) achieved a PFS2/PFS1 > 1.3. The median PFS2 on matched treatment subgroup was 2.3 months (95% CI 1.8-3.6) and the median OS was 11.4 months (95% CI 9-15.5). CONCLUSIONS The molecular screening of patients with refractory, metastatic rare cancers might increase the therapeutic options. Facilitating access strategy to molecular-driven clinical trials or agnostic-approved treatment is crucial.
Collapse
Affiliation(s)
- Véronique Debien
- Department of oncology, Institut de Cancérologie Strasbourg Europe, ICANS, Strasbourg, France; Drug Development Department (DITEP), Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France; Institut Jules Bordet, Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | | | - Christophe Massard
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France; Department of Oncology, Institut Eugène Marquis, Rennes, France
| | - Gabriel Malouf
- Department of oncology, Institut de Cancérologie Strasbourg Europe, ICANS, Strasbourg, France
| | - Antoine Hollebecque
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Jean-Yves Scoazec
- Pathology Department, Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France; AMMICa, CNRS UAR3655 INSERM US23, Université Paris Saclay, Villejuif, France
| | - Stefan Michiels
- Service de Biostatistique et d'Epidémiologie, Oncostat, CESP, Inserm U1018, Université Paris-Saclay, Equipe labellisée Ligue Contre le Cancer, Institut Gustave Roussy, Villejuif, France
| | - Loïc Verlingue
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France; Unité de Phase 1, Centre Léon Bérard, Lyon, France.
| |
Collapse
|
46
|
Trapani D, Crimini E, Sandoval J, Curigliano G. Next-Generation Sequencing for Advanced Breast Cancer: What the Way to Go? Cancer Treat Res 2023; 188:343-351. [PMID: 38175352 DOI: 10.1007/978-3-031-33602-7_13] [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: 01/05/2024]
Abstract
The rapid implementation of precision medicine tools in diagnosing and treating breast cancer (BC) has widened the potential therapeutic options for patients. The applications of gene sequencing, including next-generation gene sequencing (NGS), have led to numerous questions on how to validate, implement, interpret, prioritize and operationalize precision medicine tools to deliver meaningful and impactful interventions. Limited benefit has been portended with earlier experiences of NGS-driven treatment, in BC. However, the development and use of frameworks of clinical actionability of genomic alterations, for example, detected with NGS, has resulted in better patient selection, and potentially higher therapeutic value. The European Society for Medical Oncology Scale for Clinical Actionability of molecular Targets (ESCAT) is a framework that includes five tiers of clinical actionability, with tier 1 reserved for approved drugs with demonstrated benefits for targetable genomic alterations. The re-analysis of clinical studies by grouping the genomic alterations and matched drugs with ESCAT, in high vs lower tiers has demonstrated a significant benefit portended by high tiers alterations, with the availability of efficacious treatments. As a result, frameworks for actionability, like ESCAT, should be fundamental in developing and implementing NGS-driven, and broadly, precision medicine research and treatments.
Collapse
Affiliation(s)
- Dario Trapani
- Division of New Drug Development for innovative therapies, European Institute of Oncology IRCCS, Milan, Italy.
| | - Edoardo Crimini
- Division of New Drug Development for innovative therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology, University of Milan, Milan, Italy
| | - José Sandoval
- Department of Oncology, Geneva University Hospitals, Geneva, Switzerland
- Unit of Population Epidemiology, Division and Department of Primary Care Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Giuseppe Curigliano
- Division of New Drug Development for innovative therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology, University of Milan, Milan, Italy
| |
Collapse
|
47
|
Heinrich K, Westphalen CB. Quo vadis precision oncology? J Cancer Res Clin Oncol 2023; 149:3-4. [PMID: 36637503 PMCID: PMC9889488 DOI: 10.1007/s00432-022-04478-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 11/09/2022] [Indexed: 01/14/2023]
Affiliation(s)
- Kathrin Heinrich
- Comprehensive Cancer Center Munich & Department of Medicine III, University Hospital Munich, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany ,German Cancer Consortium (DKTK Partner Site Munich), Heidelberg, Germany
| | - C. Benedikt Westphalen
- Comprehensive Cancer Center Munich & Department of Medicine III, University Hospital Munich, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany ,German Cancer Consortium (DKTK Partner Site Munich), Heidelberg, Germany
| |
Collapse
|
48
|
Pinet S, Durand S, Perani A, Darnaud L, Amadjikpe F, Yon M, Darbas T, Vergnenegre A, Egenod T, Simonneau Y, Le Brun-Ly V, Pestre J, Venat L, Thuillier F, Chaunavel A, Duchesne M, Fermeaux V, Guyot A, Lacorre S, Bessette B, Lalloué F, Durand K, Deluche E. Clinical management of molecular alterations identified by high throughput sequencing in patients with advanced solid tumors in treatment failure: Real-world data from a French hospital. Front Oncol 2023; 13:1104659. [PMID: 36923436 PMCID: PMC10009270 DOI: 10.3389/fonc.2023.1104659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/07/2023] [Indexed: 03/03/2023] Open
Abstract
Background In the context of personalized medicine, screening patients to identify targetable molecular alterations is essential for therapeutic decisions such as inclusion in clinical trials, early access to therapies, or compassionate treatment. The objective of this study was to determine the real-world impact of routine incorporation of FoundationOne analysis in cancers with a poor prognosis and limited treatment options, or in those progressing after at least one course of standard therapy. Methods A FoundationOneCDx panel for solid tumor or liquid biopsy samples was offered to 204 eligible patients. Results Samples from 150 patients were processed for genomic testing, with a data acquisition success rate of 93%. The analysis identified 2419 gene alterations, with a median of 11 alterations per tumor (range, 0-86). The most common or likely pathogenic variants were on TP53, TERT, PI3KCA, CDKN2A/B, KRAS, CCDN1, FGF19, FGF3, and SMAD4. The median tumor mutation burden was three mutations/Mb (range, 0-117) in 143 patients with available data. Of 150 patients with known or likely pathogenic actionable alterations, 13 (8.6%) received matched targeted therapy. Sixty-nine patients underwent Molecular Tumor Board, which resulted in recommendations in 60 cases. Treatment with genotype-directed therapy had no impact on overall survival (13 months vs. 14 months; p = 0.95; hazard ratio = 1.04 (95% confidence interval, 0.48-2.26)]. Conclusions This study highlights that an organized center with a Multidisciplinary Molecular Tumor Board and an NGS screening system can obtain satisfactory results comparable with those of large centers for including patients in clinical trials.
Collapse
Affiliation(s)
- Sandra Pinet
- Medical Oncology Department, Dupuytren University Hospital, Limoges, France
| | - Stéphanie Durand
- The National Institute for Health and Medical Research (INSERM) U1308 - CAPTuR "Control Of Cell Activation, Tumor Progression and Therapeutic Resistance", Faculty of Medicine, University of Limoges, Limoges, France
| | - Alexandre Perani
- Cytogenetic, Medical Genetic and Reproductive Biology, Dupuytren University Hospital, Limoges, France
| | - Léa Darnaud
- Department of Pathology, Dupuytren University Hospital, Limoges, France
| | - Fifame Amadjikpe
- Department of Pathology, Dupuytren University Hospital, Limoges, France
| | - Mathieu Yon
- Department of Pathology, Dupuytren University Hospital, Limoges, France
| | - Tiffany Darbas
- Medical Oncology Department, Dupuytren University Hospital, Limoges, France
| | | | - Thomas Egenod
- Chest Department, Dupuytren University Hospital, Limoges, France
| | | | - Valérie Le Brun-Ly
- Medical Oncology Department, Dupuytren University Hospital, Limoges, France
| | - Julia Pestre
- Medical Oncology Department, Dupuytren University Hospital, Limoges, France
| | - Laurence Venat
- Medical Oncology Department, Dupuytren University Hospital, Limoges, France
| | - Frédéric Thuillier
- Medical Oncology Department, Dupuytren University Hospital, Limoges, France
| | - Alain Chaunavel
- The National Institute for Health and Medical Research (INSERM) U1308 - CAPTuR "Control Of Cell Activation, Tumor Progression and Therapeutic Resistance", Faculty of Medicine, University of Limoges, Limoges, France.,Department of Pathology, Dupuytren University Hospital, Limoges, France
| | - Mathilde Duchesne
- Department of Pathology, Dupuytren University Hospital, Limoges, France.,Research Unit (UR) 20218 - NEURIT "Neuropathies et Innovations Thérapeutiques", Faculty of Medicine, University of Limoges, Limoges, France
| | | | - Anne Guyot
- Department of Pathology, Dupuytren University Hospital, Limoges, France
| | - Sylvain Lacorre
- Department of Pathology, Dupuytren University Hospital, Limoges, France
| | - Barbara Bessette
- The National Institute for Health and Medical Research (INSERM) U1308 - CAPTuR "Control Of Cell Activation, Tumor Progression and Therapeutic Resistance", Faculty of Medicine, University of Limoges, Limoges, France
| | - Fabrice Lalloué
- The National Institute for Health and Medical Research (INSERM) U1308 - CAPTuR "Control Of Cell Activation, Tumor Progression and Therapeutic Resistance", Faculty of Medicine, University of Limoges, Limoges, France
| | - Karine Durand
- The National Institute for Health and Medical Research (INSERM) U1308 - CAPTuR "Control Of Cell Activation, Tumor Progression and Therapeutic Resistance", Faculty of Medicine, University of Limoges, Limoges, France.,Department of Pathology, Dupuytren University Hospital, Limoges, France
| | - Elise Deluche
- Medical Oncology Department, Dupuytren University Hospital, Limoges, France.,The National Institute for Health and Medical Research (INSERM) U1308 - CAPTuR "Control Of Cell Activation, Tumor Progression and Therapeutic Resistance", Faculty of Medicine, University of Limoges, Limoges, France
| |
Collapse
|
49
|
Martin-Romano P, Mezquita L, Hollebecque A, Lacroix L, Rouleau E, Gazzah A, Bahleda R, Planchard D, Varga A, Baldini C, Postel-Vinay S, Friboulet L, Loriot Y, Verlingue L, Geraud A, Camus MN, Nicotra C, Soria JC, André F, Besse B, Massard C, Italiano A. Implementing the European Society for Medical Oncology Scale for Clinical Actionability of Molecular Targets in a Comprehensive Profiling Program: Impact on Precision Medicine Oncology. JCO Precis Oncol 2022; 6:e2100484. [DOI: 10.1200/po.21.00484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
Abstract
PURPOSE To facilitate implementation of precision medicine in clinical management of cancer, the European Society of Medical Oncology proposed in 2018 a new scale to harmonize and standardize the reporting and interpretation of clinically relevant genomics data (ESMO Scale of Actionability of molecular Targets [ESCAT]). This study aims to characterize the clinical impact of matching targetable genomic alterations (GAs) in patients with advanced cancer according to ESCAT. MATERIAL AND METHODS Analysis of next-generation sequencing results from 552 patients is included in two prospective precision medicine studies at Gustave Roussy. End points included objective response rates, progression-free survival, and overall survival according to ESCAT. RESULTS Molecular data from 516 patients were available and discussed within a Molecular Tumor Board. The most common tumor types were GI (n = 164; 30%), lung (n = 137; 25%), and urologic tumors (n = 68; 13%). Overall, 379 GAs were considered as actionable targets according to ESCAT in 348 (67%) patients. In 31 (6%) patients, two concomitant actionable targets were identified. On the basis of ESCAT, GAs were considered to be classified as tier I in 120 patients (29%), II in 25 patients (5%), III in 80 patients (16%), and IV in 153 patients (30%). A total of 136 patients (27%) received a matched therapy. ESCAT was significantly associated with objective response rates and clinical benefit rates. The median progression-free survival was 6.5 months (95% CI, 4.2 to 8.9), 3 months (95% CI, 1 to not available), 3 months (95% CI, 2.2 to 3.8), and 4 months (95% CI, 2.8 to 6.3) for ESCAT I, II, III, and IV, respectively ( P = .0125). CONCLUSION Implementation of ESCAT classification for clinical decision making by Molecular Tumor Board is feasible and useful to better tailor therapies in patients with cancer.
Collapse
Affiliation(s)
- Patricia Martin-Romano
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
- Precision Medicine Group, Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Laura Mezquita
- Precision Medicine Group, Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
- Gustave Roussy, Department of Medical Oncology, Villejuif, France
| | - Antoine Hollebecque
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
- Precision Medicine Group, Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Ludovic Lacroix
- Department of Medical Biology and Pathology, Translational Research Laboratory and Biobank, Gustave Roussy, Villejuif, France
| | - Etienne Rouleau
- Department of Medical Biology and Pathology, Translational Research Laboratory and Biobank, Gustave Roussy, Villejuif, France
| | - Anas Gazzah
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Rastilav Bahleda
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - David Planchard
- Precision Medicine Group, Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Andrea Varga
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Capucine Baldini
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Sophie Postel-Vinay
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
- INSERM Unit U981, Gustave Roussy, Villejuif, France
| | | | - Yohann Loriot
- Precision Medicine Group, Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
- Gustave Roussy, Department of Medical Oncology, Villejuif, France
| | - Loic Verlingue
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
- Precision Medicine Group, Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Arthur Geraud
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
- Precision Medicine Group, Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Maud Ngo Camus
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Claudio Nicotra
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Jean Charles Soria
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
- INSERM Unit U981, Gustave Roussy, Villejuif, France
| | - Fabrice André
- Precision Medicine Group, Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
- INSERM Unit U981, Gustave Roussy, Villejuif, France
| | - Benjamin Besse
- Precision Medicine Group, Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
- INSERM Unit U981, Gustave Roussy, Villejuif, France
| | - Christophe Massard
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
- Precision Medicine Group, Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
- INSERM Unit U981, Gustave Roussy, Villejuif, France
| | - Antoine Italiano
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
- Precision Medicine Group, Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
50
|
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
|