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
|
Ghosh M, Lingaraju SM, C R K, Balaram G, Kodandapani R, E V, K V, N S, H D, Patil S, Thungappa SC, Bhattacharjee S, P S S, Dasgupta R, Naseer M, B J S, Rao V, Ramaswamy V, Naik R, Babu G, Ravichandran A, Bahadur U, Murugan K, B M, Reddy L, Basavalinga S A. Comprehensive genomic profiling reveals a unique genomic landscape in solid tumors in an Indian cancer cohort of 1000 patients: a single institutional experience. Sci Rep 2025; 15:12455. [PMID: 40216820 PMCID: PMC11992052 DOI: 10.1038/s41598-025-94762-z] [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: 09/11/2024] [Accepted: 03/17/2025] [Indexed: 04/14/2025] Open
Abstract
The use of Comprehensive Genomic Profiling (CGP) in clinical practice to detect broad-spectrum therapeutic, prognostic, and predictive biomarkers, including tumor mutational burden (TMB), microsatellite instability (MSI), somatic BRCA (sBRCA) and other homologous recombination repair genes (HRRs) provides a more cost-efficient and tissue-preserving approach than serial single-biomarker analysis. A total of 1000 biopsy-proven cancer patients at the HCG cancer center were profiled in an IRB-approved prospective study. The findings were discussed in the multidisciplinary molecular tumor board (MTB), and recommendations were documented in electronic medical records (EMRs) for clinical management and follow-up. A total of 1747 genomic alterations were detected (mean 1.7 mutations/sample), with 80% of patients having genetic alterations with therapeutic and prognostic implications (Tier I-32%, Tier II-50%). CGP revealed a greater number of druggable genes (47%) than did small panels (14%). Tumor-agnostic markers for immunotherapy (IO) were observed in 16% of the current cohort, based on which IO was initiated. In 13.5% of the cohort, alterations in the HRR pathway including sBRCA (5.5%) were detected providing an option for treatment with platinum or PARP inhibitors. Other significant alterations included those in EGFR, KRAS/BRAF, PIK3CA, cKIT, PDGFRA, ARID1A, ARID2, and FGFR. RNA sequencing revealed 55 + RNA alterations, including those in TMPRSS-ERG, RPS6KB1-VMP1, EML4-ALK, NTRK, PDGFRA and EWSR. Clinical outcome data were available via EMR for 618 patients (62%), out of whom 419 patients had druggable mutations (67%; 95% CI 88.9-93.9%) and 39 patients had 1 or more mutations with prognostic implications. However, only 200 patients (44%; 95% CI 39.1-48.1%) were included in the MTB discussion. Based on genomics reports, the treatment regimen was changed for 137 and 61 patients with and without clinical inputs from the MTB, respectively. The overall change in therapy based on CGP in the clinical cohort was 43%, which was greater in patients enrolled for MTB than in patients who had not undergone MTB. At the interim analysis, with a median follow-up of 18 months (range 12-24 months) after the change in therapy as per genomics report, 97 patients (71%) were found to be alive thus establishing the importance of CGP and MTB in personalized genomics-driven treatment.
Collapse
Affiliation(s)
- Mithua Ghosh
- Triesta Sciences, A Unit of HeathCare Global Enterprises Limited, HCG Towers, Tower 1, #8P Kalinga Rao Road, Sampangiram Nagar, Bangalore, Karnataka, 560027, India.
| | - Sheela Mysore Lingaraju
- Triesta Sciences, A Unit of HeathCare Global Enterprises Limited, HCG Towers, Tower 1, #8P Kalinga Rao Road, Sampangiram Nagar, Bangalore, Karnataka, 560027, India
| | - Krishna C R
- Triesta Sciences, A Unit of HeathCare Global Enterprises Limited, HCG Towers, Tower 1, #8P Kalinga Rao Road, Sampangiram Nagar, Bangalore, Karnataka, 560027, India
| | - Gautam Balaram
- Triesta Sciences, A Unit of HeathCare Global Enterprises Limited, HCG Towers, Tower 1, #8P Kalinga Rao Road, Sampangiram Nagar, Bangalore, Karnataka, 560027, India
| | - Ramya Kodandapani
- Triesta Sciences, A Unit of HeathCare Global Enterprises Limited, HCG Towers, Tower 1, #8P Kalinga Rao Road, Sampangiram Nagar, Bangalore, Karnataka, 560027, India
| | - Vijay E
- Triesta Sciences, A Unit of HeathCare Global Enterprises Limited, HCG Towers, Tower 1, #8P Kalinga Rao Road, Sampangiram Nagar, Bangalore, Karnataka, 560027, India
| | - Vijay K
- Triesta Sciences, A Unit of HeathCare Global Enterprises Limited, HCG Towers, Tower 1, #8P Kalinga Rao Road, Sampangiram Nagar, Bangalore, Karnataka, 560027, India
| | - Suhas N
- Triesta Sciences, A Unit of HeathCare Global Enterprises Limited, HCG Towers, Tower 1, #8P Kalinga Rao Road, Sampangiram Nagar, Bangalore, Karnataka, 560027, India
| | - Devika H
- Triesta Sciences, A Unit of HeathCare Global Enterprises Limited, HCG Towers, Tower 1, #8P Kalinga Rao Road, Sampangiram Nagar, Bangalore, Karnataka, 560027, India
| | - Shekar Patil
- HealthCare Global Enterprises Limited, Bangalore, Karnataka, 560027, India
| | | | | | - Sridhar P S
- HealthCare Global Enterprises Limited, Bangalore, Karnataka, 560027, India
| | - Roshni Dasgupta
- HealthCare Global Enterprises Limited, Bangalore, Karnataka, 560027, India
| | - Mohammed Naseer
- HealthCare Global Enterprises Limited, Bangalore, Karnataka, 560027, India
| | - Srinivas B J
- HealthCare Global Enterprises Limited, Bangalore, Karnataka, 560027, India
| | - Vishal Rao
- HealthCare Global Enterprises Limited, Bangalore, Karnataka, 560027, India
| | - Veena Ramaswamy
- Triesta Sciences, A Unit of HeathCare Global Enterprises Limited, HCG Towers, Tower 1, #8P Kalinga Rao Road, Sampangiram Nagar, Bangalore, Karnataka, 560027, India
| | - Radheshyam Naik
- HealthCare Global Enterprises Limited, Bangalore, Karnataka, 560027, India
| | - Govind Babu
- HealthCare Global Enterprises Limited, Bangalore, Karnataka, 560027, India
| | | | | | - Krithika Murugan
- HealthCare Global Enterprises Limited, Bangalore, Karnataka, 560027, India
| | - Mahesh B
- HealthCare Global Enterprises Limited, Bangalore, Karnataka, 560027, India
| | - Lohith Reddy
- HealthCare Global Enterprises Limited, Bangalore, Karnataka, 560027, India
| | | |
Collapse
|
3
|
Chen H, An Y, Wang C, Zhou J. Circulating tumor DNA in colorectal cancer: biology, methods and applications. Discov Oncol 2025; 16:439. [PMID: 40167831 PMCID: PMC11961841 DOI: 10.1007/s12672-025-02220-z] [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: 05/28/2024] [Accepted: 03/24/2025] [Indexed: 04/02/2025] Open
Abstract
In the practice of colorectal cancer (CRC), traditional tumor tissue analysis is limited by intratumoral and intertumoral heterogeneity and its invasive nature. Circulating tumor DNA (ctDNA) analysis, a promising liquid biopsy approach, has been increasingly explored in clinical studies. Biologically, ctDNA is characterized by tumor-specific diversity and rapid clearance from circulation, enabling real-time, dynamic, and repeatable assessments. Technologically, PCR- and NGS-based downstream analysis methods have been developed and validated. However, variables in pre-analytical and analytical procedures underscores the need for standardized protocols. Compared with clinicopathology-based risk stratification, ctDNA-based molecular residual disease detection has demonstrated significant potential in guiding treatment decisions. Qualitative and quantitative changes in ctDNA have also shown predictive and prognostic value during neoadjuvant or adjuvant treatment, as well as in later-line treatment for metastatic CRC. Specific molecular aberrations in ctDNA can not only assist in identifying candidates for targeted therapies but also reveal resistance mechanisms. Additionally, emerging research is exploring the potential of ctDNA in early cancer detection. Overall, as a novel biomarker, ctDNA holds substantial promise in advancing clinical practice. This review focuses on the biological characteristics, pre-analytical variables, and downstream analysis methods of ctDNA and summarizes its role across various clinical scenarios in CRC.
Collapse
Affiliation(s)
- Han Chen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1, Shuaifuyuan, Beijing, 100730, China
| | - Yang An
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1, Shuaifuyuan, Beijing, 100730, China
| | - Chentong Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1, Shuaifuyuan, Beijing, 100730, China
| | - Jiaolin Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1, Shuaifuyuan, Beijing, 100730, China.
| |
Collapse
|
4
|
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
|
5
|
Yu B, Shao S, Ma W. Frontiers in pancreatic cancer on biomarkers, microenvironment, and immunotherapy. Cancer Lett 2025; 610:217350. [PMID: 39581219 DOI: 10.1016/j.canlet.2024.217350] [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: 09/20/2024] [Revised: 11/06/2024] [Accepted: 11/21/2024] [Indexed: 11/26/2024]
Abstract
Pancreatic cancer remains one of the most challenging malignancies to treat due to its late-stage diagnosis, aggressive progression, and high resistance to existing therapies. This review examines the latest advancements in early detection, and therapeutic strategies, with a focus on emerging biomarkers, tumor microenvironment (TME) modulation, and the integration of artificial intelligence (AI) in data analysis. We highlight promising biomarkers, including microRNAs (miRNAs) and circulating tumor DNA (ctDNA), that offer enhanced sensitivity and specificity for early-stage diagnosis when combined with multi-omics panels. A detailed analysis of the TME reveals how components such as cancer-associated fibroblasts (CAFs), immune cells, and the extracellular matrix (ECM) contribute to therapy resistance by creating immunosuppressive barriers. We also discuss therapeutic interventions that target these TME components, aiming to improve drug delivery and overcome immune evasion. Furthermore, AI-driven analyses are explored for their potential to interpret complex multi-omics data, enabling personalized treatment strategies and real-time monitoring of treatment response. We conclude by identifying key areas for future research, including the clinical validation of biomarkers, regulatory frameworks for AI applications, and equitable access to innovative therapies. This comprehensive approach underscores the need for integrated, personalized strategies to improve outcomes in pancreatic cancer.
Collapse
Affiliation(s)
- Baofa Yu
- Taimei Baofa Cancer Hospital, Dongping, Shandong, 271500, China; Jinan Baofa Cancer Hospital, Jinan, Shandong, 250000, China; Beijing Baofa Cancer Hospital, Beijing, 100010, China; Immune Oncology Systems, Inc, San Diego, CA, 92102, USA.
| | - Shengwen Shao
- Institute of Microbiology and Immunology, Huzhou University School of Medicine, Huzhou, Zhejiang, 313000, China.
| | - Wenxue Ma
- Department of Medicine, Sanford Stem Cell Institute, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
6
|
Suzuki S, Saito Y, Saito K, Yamada Y, Takahashi K, Kumanishi R, Fukui T, Yoshioka T. Limited Efficacy of Anti-EGFR Monoclonal Antibodies in Colorectal Cancer Patients with Rare RAS Variants: Analysis of the C-CAT Database. Curr Issues Mol Biol 2024; 46:14476-14486. [PMID: 39727997 DOI: 10.3390/cimb46120869] [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: 11/30/2024] [Revised: 12/20/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024] Open
Abstract
Epidermal growth factor receptor (EGFR) inhibition is crucial in treating RAS wild-type metastatic colorectal cancer, yet current testing methods may miss rare RAS variants affecting treatment efficacy. We analyzed 4122 colorectal cancer patients receiving anti-EGFR antibodies from the Center for Cancer Genomics and Advanced Therapeutics database, identifying 54 patients (1.3%) with rare RAS variants undetectable by standard testing. These patients showed significantly lower response rates to anti-EGFR therapy (28.3%) compared to RAS wild-type cases (44.6%, p = 0.003). Disease control rates were also lower in rare variant cases (60.9%) versus wild-type cases (80.0%). Most common rare variants included KRAS Q22K, A59E, and A11_G12insGA. Comprehensive genomic profiling revealed additional alterations in TP53 (90.7%), APC (87.0%), and non-V600E BRAF mutations (25.9%). Our findings suggest that rare RAS variants predict poor anti-EGFR therapy response, highlighting the potential benefit of comprehensive genomic profiling before treatment initiation. This study provides real-world evidence supporting the clinical relevance of rare RAS variants in treatment decision-making for colorectal cancer. Future studies should focus on developing cost-effective comprehensive testing strategies and evaluating alternative treatment approaches for patients with rare RAS variants.
Collapse
Affiliation(s)
- Shuhei Suzuki
- Yamagata Hereditary Tumor Research Center, Yamagata University, 1-4-12 Kojirakawa, Yamagata 990-8560, Japan
- Department of Clinical Oncology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Yosuke Saito
- Department of Gastroenterology, Yamagata City Hospital Saiseikan, 1-2-26 Nanokamachi, Yamagata 990-0042, Japan
| | - Koki Saito
- Graduate School of Medicine, Tokyo Medical University, 6-1-1 Shinjuku, Tokyo 160-8402, Japan
| | - Yuta Yamada
- Department of Clinical Oncology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Koshi Takahashi
- Department of Clinical Oncology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Ryosuke Kumanishi
- Department of Clinical Oncology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Tadahisa Fukui
- Department of Clinical Oncology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Takashi Yoshioka
- Department of Clinical Oncology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| |
Collapse
|
7
|
Iwasawa O, Ikegami M, Miyagawa T, Morita H, Saito H, Omori I, Awaji K, Omatsu J, Yamada D, Kage H, Oda K, Sato S, Sumida H. Association of genetic alterations with prognosis in extramammary Paget disease: insights into the involvement of somatic CDKN2A variants in patients with a poor prognosis. Br J Dermatol 2024; 192:46-54. [PMID: 39172540 DOI: 10.1093/bjd/ljae337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/27/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND Previous studies have reported the mutational landscape in extramammary Paget disease (EMPD); however, the prognostic implications of genetic alterations remain unexplored. While CDKN2A loss is known to be associated with tumour progression or poor prognosis in some types of cancer, its significance in EMPD has not been investigated. OBJECTIVES To examine the association between common genetic alterations and prognosis in EMPD. METHODS A retrospective cohort study was carried out to analyse the data of patients with EMPD registered up to January 2024 in the Center for Cancer Genomics and Advanced Therapeutics database, a nationwide database that records clinical data and comprehensive genomic profiling (CGP) test results in Japan. RESULTS A total of 167 patients with EMPD were recorded in the database, with CDKN2A loss being the most frequent genetic variant. Survival analysis was conducted on data from 127 patients. Survival from the initiation of chemotherapy was analysed, adjusting for length bias inherent in the database with the Kaplan-Meier estimator, an established method of adjustment. Patients with BRCA2-mutant tumours (n = 18) had a worse prognosis than those with BRCA2 wildtype (WT) tumours [n = 109; hazard ratio (HR) 2.97, 95% confidence interval (CI) 1.46-6.01 (P = 0.003)]. Additionally, patients in the CDKN2A mutant group (n = 72) had a significantly worse prognosis compared with those in the CDKN2A WT group [n = 55; HR 1.81, 95% CI 1.06-3.07 (P = 0.029)]. Most CDKN2A variants were pathogenic, primarily characterized by loss, while most BRCA2 variants were variants of uncertain significance. In the survival analysis of CGP enrolment based on Eastern Cooperative Oncology Group performance status (ECOG-PS), patients with an ECOG-PS of 1 at the time of CGP enrolment had a significantly poorer prognosis compared with those with an ECOG-PS of 0 (P = 0.034; median survival time 531 vs. 259 days). CONCLUSIONS A somatic CDKN2A variant, mainly exhibiting loss, may be associated with a poor prognosis in EMPD. Patients with EMPD with BRCA2-mutant disease might also have a worse prognosis. In addition, CGP testing before ECOG-PS deteriorates is preferable, considering that the observed median survival of individuals undergoing CGP tests in an ECOG-PS 1 condition was < 9 months.
Collapse
Affiliation(s)
- Okuto Iwasawa
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masachika Ikegami
- Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Takuya Miyagawa
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiromichi Morita
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hinako Saito
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Issei Omori
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kentaro Awaji
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Jun Omatsu
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Daisuke Yamada
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hidenori Kage
- Department of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Katsutoshi Oda
- Division of Integrative Genomics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shinichi Sato
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hayakazu Sumida
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Scleroderma Center, The University of Tokyo Hospital, Tokyo, Japan
| |
Collapse
|
8
|
Fan F, Jiang G, Lv J, Wang H, Li W, Liu C, Zhao Y, Zhang Z, Du H, Zhang Z, Li X, Li W. Analytical and clinical validation of a NGS panel in detecting targetable variants from ctDNA of metastatic NSCLC patients. Cancer Med 2024; 13:e70078. [PMID: 39385536 PMCID: PMC11464656 DOI: 10.1002/cam4.70078] [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: 04/29/2024] [Revised: 07/15/2024] [Accepted: 07/24/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Circulating tumor DNA (ctDNA) has emerged as a promising biomarker for noninvasive cancer diagnostics, particularly in the context of metastatic non-small-cell lung cancer (NSCLC). Detecting targetable variants through ctDNA analysis offers the potential to guide treatment decisions, especially in cases where tissue samples are insufficient or unavailable. METHOD In this study, we developed and validated a next-generation sequencing panel targeting 101 cancer-related genes (101-test) to detect somatic variants in ctDNA from a large cohort of Chinese patients with metastatic NSCLC. The performance of the 101-test was assessed by evaluating its limit of detection (LOD), accuracy, and precision in identifying molecular variants. Additionally, the concordance between ctDNA and tissue samples for detecting targetable variants was analyzed in 904 patients. RESULTS The 101-test demonstrated a LOD of 0.38% for single-nucleotide variants (SNVs), 0.33% for insertions and deletions (InDels), and 0.33% for fusions. Sensitivity was 98.3% for SNVs, 100% for InDels, and 100% for fusions when compared to digital droplet PCR (ddPCR)/breakpoint PCR reference methods. The by-variant sensitivity for somatic variants was 97.5%, with a specificity of 99.9% between tumor-only and tumor-normal analyses. In a real-world cohort, the concordance between ctDNA and tissue samples for identifying targetable variants was 72.2% (457/633). Notably, the EGFR S768I variant, recently recommended by clinical guidelines, achieved an 80% concordance rate. Furthermore, 4.3% of patients (27/633) with targetable variants were identified exclusively through ctDNA testing. CONCLUSION The ctDNA-based 101-test is a highly sensitive and specific tool for detecting targetable variants in metastatic NSCLC, particularly in cases with insufficient tissue samples. The findings support the use of ctDNA testing as a reliable and complementary method to traditional tissue-based molecular analysis, enhancing the precision of treatment strategies for NSCLC patients.
Collapse
Affiliation(s)
- Feifei Fan
- Department of Respiratory MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Guozhong Jiang
- Department of PathologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Juan Lv
- Burning Rock BiotechGuangzhouChina
| | - Hongmin Wang
- Department of Respiratory MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | | | | | - Yu Zhao
- Burning Rock BiotechGuangzhouChina
| | | | | | | | - Xiangnan Li
- Department of Thoracic Surgery and Lung TransplantationThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Wen‐cai Li
- Department of PathologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
9
|
Hung LJ, Huang CY, Tung KC, Chen JS, Huang WK, Hsu CC, Fang YF, Wang CL, Liu PC, Yeh KY, Chang PH, Chang JWC, Lin YC, Huang SF, Chou WC. Comprehensive genomic profiling in multiple cancer types: A comparative analysis of the National Biobank Consortium of Taiwan and clinical practice cohorts. J Formos Med Assoc 2024:S0929-6646(24)00405-4. [PMID: 39244401 DOI: 10.1016/j.jfma.2024.09.001] [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: 05/08/2024] [Revised: 07/30/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND This retrospective study analyzed tumor tissue profiling data to assess the potential of comprehensive genomic profiling (CGP) for patient care across diverse solid tumors. MATERIAL AND METHODS Patients with newly diagnosed or recurrent stage IIIB or IV lung adenocarcinoma with a null immunophenotype and esophageal, gastric, pancreatic, or bile duct cancer between January 2020 and July 2023 at two medical centers in Taiwan were included. One cohort was a part of the National Biobank Consortium of Taiwan project, whereas the other consisted of patients undergoing routine clinical practice. Tumor samples were subjected to CGP using FoundationOne®CDx, with therapeutic implications determined using OncoKB classification. RESULTS FoundationOne®CDx testing of 574 patients was successful in 456 (79.4%) patients. Clinically actionable genomic alterations were detected in 21.1% (96/456) of the patients, including 17.5%, 2.9%, and 0.7% of patients with evidence levels 1, 2, and 3, respectively. Lung adenocarcinoma accounted for the largest proportion of samples with at least one actionable gene alteration (63.2%), followed by bile duct (26.9%), gastric (17.6%), esophageal (4.0%), and pancreatic (3.1%) cancers. Based on CGP results, 43 patients (9.4%) received matched targeted therapy. The median overall survival of patients who received matched therapy or not was 26.1 months (95% confidence interval (CI), 16.7-35.5 months) and 10.6 months (95% CI, 8.1-13.1 months; hazard ratio, 0.28, 95% CI, 0.14-0.55, p < 0.001), respectively. CONCLUSIONS This study provides comprehensive insights into the genomic profiles of diverse cancers in Taiwan, highlighting the crucial role of CGP in identifying actionable genomic alterations and guiding effective therapeutic strategies in real-world practice.
Collapse
Affiliation(s)
- Ling-Jen Hung
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan; Division of Hematology-Oncology, Department of Internal Medicine, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan
| | - Chen-Yang Huang
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Kai-Che Tung
- Patient Journey Solution Division, Roche Products Ltd., Taipei, Taiwan; Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Jen-Shi Chen
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Wen-Kuan Huang
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Chih-Chung Hsu
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Yueh-Fu Fang
- Division of Thoracic Oncology, Department of Thoracic Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Chih-Liang Wang
- Division of Thoracic Oncology, Department of Thoracic Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Ping-Chi Liu
- Division of Thoracic Oncology, Department of Thoracic Medicine, Keelung Chang Gung Memorial Hospital and Chang Gung University, Keelung, Taiwan
| | - Kun-Yun Yeh
- Division of Hematology-Oncology, Department of Internal Medicine, Keelung Chang Gung Memorial Hospital and Chang Gung University, Keelung, Taiwan
| | - Pei-Hung Chang
- Division of Hematology-Oncology, Department of Internal Medicine, Keelung Chang Gung Memorial Hospital and Chang Gung University, Keelung, Taiwan
| | - John Wen-Cheng Chang
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Yung-Chang Lin
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Shiu-Feng Huang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Wen-Chi Chou
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
10
|
Hayashi R. Identification of patients with NSCLC undergoing multiplex gene analysis that also require comprehensive genomic profiling. Respir Investig 2024; 62:744-745. [PMID: 38905994 DOI: 10.1016/j.resinv.2024.04.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 06/23/2024]
|
11
|
Krämer A, Bochtler T, Pauli C, Shiu KK, Cook N, de Menezes JJ, Pazo-Cid RA, Losa F, Robbrecht DG, Tomášek J, Arslan C, Özgüroğlu M, Stahl M, Bigot F, Kim SY, Naito Y, Italiano A, Chalabi N, Durán-Pacheco G, Michaud C, Scarato J, Thomas M, Ross JS, Moch H, Mileshkin L. Molecularly guided therapy versus chemotherapy after disease control in unfavourable cancer of unknown primary (CUPISCO): an open-label, randomised, phase 2 study. Lancet 2024; 404:527-539. [PMID: 39096924 DOI: 10.1016/s0140-6736(24)00814-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/12/2024] [Accepted: 04/16/2024] [Indexed: 08/05/2024]
Abstract
BACKGROUND Patients with unfavourable subset cancer of unknown primary (CUP) have a poor prognosis when treated with standard platinum-based chemotherapy. Whether first-line treatment guided by comprehensive genomic profiling (CGP) can improve outcomes is unknown. The CUPISCO trial was designed to inform a molecularly guided treatment strategy to improve outcomes over standard platinum-based chemotherapy in patients with newly diagnosed, unfavourable, non-squamous CUP. The aim of the trial was to compare the efficacy and safety of molecularly guided therapy (MGT) versus standard platinum-based chemotherapy in these patients. This was to determine whether the inclusion of CGP in the initial diagnostic work-up leads to improved outcomes over the current standard of care. We herein report the primary analysis. METHODS CUPISCO was a phase 2, prospective, randomised, open-label, active-controlled, multicentre trial done at 159 sites in 34 countries outside the USA. Patients with central eligibility review-confirmed disease (acceptable histologies included adenocarcinoma and poorly differentiated carcinoma) and an Eastern Cooperative Oncology Group performance status of 0 or 1, evaluated by CGP, who reached disease control after three cycles of standard first-line platinum-based chemotherapy were randomly assigned 3:1 via a block-stratified randomisation procedure to MGT versus chemotherapy continuation for at least three further cycles. The primary endpoint was investigator-assessed progression-free survival in the intention-to-treat population. The study is registered with ClinicalTrials.gov, NCT03498521, and follow-up is ongoing. FINDINGS From July 10, 2018, to Dec 9, 2022, 636 (42%) of 1505 screened patients were enrolled. Median follow-up in the treatment period was 24·1 months (IQR 11·6-35·6). Of 438 patients who reached disease control after induction chemotherapy, 436 were randomly assigned: 326 (75%) to the MGT group and 110 (25%) to the chemotherapy group. Median progression-free survival in the intention-to-treat population was 6·1 months (95% CI 4·7-6·5) in the MGT group versus 4·4 months (4·1-5·6) in the chemotherapy group (hazard ratio 0·72 [95% CI 0·56-0·92]; p=0·0079). Related adverse event rates per 100-patient-years at risk were generally similar or lower with MGT versus chemotherapy. INTERPRETATION In patients with previously untreated, unfavourable, non-squamous CUP who reached disease control after induction chemotherapy, CGP with subsequent MGTs resulted in longer progression-free survival than standard platinum-based chemotherapy. On the basis of these results, we recommend that CGP is performed at initial diagnosis in patients with unfavourable CUP. FUNDING F Hoffmann-La Roche.
Collapse
Affiliation(s)
- Alwin Krämer
- Clinical Cooperation Unit Molecular Hematology-Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany.
| | - Tilmann Bochtler
- Clinical Cooperation Unit Molecular Hematology-Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; Department of Medical Oncology, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Chantal Pauli
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland; Department of Pathology and Molecular Pathology, University Hospital of Zurich, Zurich, Switzerland; Medical Faculty, University of Zurich, Zurich, Switzerland
| | - Kai-Keen Shiu
- UCLH Gastrointestinal Oncology Service, Cancer of Unknown Primary Service, University College London, Cancer Institute, London, UK
| | - Natalie Cook
- The Christie NHS Foundation Trust and Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | | | - Roberto A Pazo-Cid
- Medical Oncology Department, Miguel Servet University Hospital, Zaragoza, Spain
| | - Ferran Losa
- Medical Oncology Department, Hospital de Sant Joan Despí Moisès Broggi, ICO Hospitalet, Barcelona, Spain
| | - Debbie Gj Robbrecht
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jiří Tomášek
- Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Cagatay Arslan
- Izmir University of Economics Medical Point Hospital, Izmir, Türkiye
| | - Mustafa Özgüroğlu
- Istanbul University Cerrahpasa, Cerrahpasa Faculty of Medicine, Department of Internal Medicine, Division of Oncology, Istanbul, Türkiye
| | | | - Frédéric Bigot
- Department of Medical Oncology, Institut de Cancérologie de l'Ouest, Angers, France
| | - Sun Young Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Yoichi Naito
- Department of General Internal Medicine, National Cancer Center Hospital East, Kashiwa, Japan
| | - Antoine Italiano
- Institut Bergonie, Early Phase Trials and Sarcoma Units, Bordeaux, France
| | - Nasséra Chalabi
- Global Product Development Medical Affairs, F Hoffmann-La Roche, Basel, Switzerland
| | | | - Chantal Michaud
- Global Product Development Medical Affairs, F Hoffmann-La Roche, Basel, Switzerland
| | - Jeremy Scarato
- Global Product Development Medical Affairs, F Hoffmann-La Roche, Basel, Switzerland
| | - Marlene Thomas
- Global Product Development Medical Affairs, F Hoffmann-La Roche, Basel, Switzerland
| | - Jeffrey S Ross
- Pathology Group, Foundation Medicine, Cambridge, MA, USA; Upstate Medical University Departments of Pathology, Urology and Medicine (Oncology), Syracuse, NY, USA
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland; Department of Pathology and Molecular Pathology, University Hospital of Zurich, Zurich, Switzerland; Medical Faculty, University of Zurich, Zurich, Switzerland
| | - Linda Mileshkin
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| |
Collapse
|
12
|
Pecci F, Nakazawa S, Ricciuti B, Harada G, Lee JK, Alessi JV, Barrichello A, Vaz VR, Lamberti G, Di Federico A, Gandhi MM, Gazgalis D, Feng WW, Jiang J, Baldacci S, Locquet MA, Gottlieb FH, Chen MF, Lee E, Haradon D, Smokovich A, Voligny E, Nguyen T, Goel VK, Zimmerman Z, Atwal S, Wang X, Bahcall M, Heist RS, Iqbal S, Gandhi N, Elliott A, Vanderwalde AM, Ma PC, Halmos B, Liu SV, Che J, Schrock AB, Drilon A, Jänne PA, Awad MM. Activating Point Mutations in the MET Kinase Domain Represent a Unique Molecular Subset of Lung Cancer and Other Malignancies Targetable with MET Inhibitors. Cancer Discov 2024; 14:1440-1456. [PMID: 38564707 PMCID: PMC11294820 DOI: 10.1158/2159-8290.cd-23-1217] [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: 10/16/2023] [Revised: 02/23/2024] [Accepted: 04/01/2024] [Indexed: 04/04/2024]
Abstract
Activating point mutations in the MET tyrosine kinase domain (TKD) are oncogenic in a subset of papillary renal cell carcinomas. Here, using comprehensive genomic profiling among >600,000 patients, we identify activating MET TKD point mutations as putative oncogenic driver across diverse cancers, with a frequency of ∼0.5%. The most common mutations in the MET TKD defined as oncogenic or likely oncogenic according to OncoKB resulted in amino acid substitutions at positions H1094, L1195, F1200, D1228, Y1230, M1250, and others. Preclinical modeling of these alterations confirmed their oncogenic potential and also demonstrated differential patterns of sensitivity to type I and type II MET inhibitors. Two patients with metastatic lung adenocarcinoma harboring MET TKD mutations (H1094Y, F1200I) and no other known oncogenic drivers achieved confirmed partial responses to a type I MET inhibitor. Activating MET TKD mutations occur in multiple malignancies and may confer clinical sensitivity to currently available MET inhibitors. Significance: The identification of targetable genomic subsets of cancer has revolutionized precision oncology and offers patients treatments with more selective and effective agents. Here, we demonstrate that activating, oncogenic MET tyrosine kinase domain mutations are found across a diversity of cancer types and are responsive to MET tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Federica Pecci
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Seshiru Nakazawa
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Biagio Ricciuti
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Guilherme Harada
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| | | | - Joao V Alessi
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Adriana Barrichello
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Victor R Vaz
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Giuseppe Lamberti
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Malini M Gandhi
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Dimitris Gazgalis
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - William W Feng
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jie Jiang
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Simon Baldacci
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Marie-Anaïs Locquet
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Felix H Gottlieb
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Monica F Chen
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| | - Elinton Lee
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Danielle Haradon
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Anna Smokovich
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Emma Voligny
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Tom Nguyen
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Vikas K Goel
- Turning Point Therapeutics, Bristol Myers Squibb Company, San Diego, California
| | - Zachary Zimmerman
- Turning Point Therapeutics, Bristol Myers Squibb Company, San Diego, California
| | - Sumandeep Atwal
- Turning Point Therapeutics, Bristol Myers Squibb Company, San Diego, California
| | - Xinan Wang
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Magda Bahcall
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Sumaiya Iqbal
- The Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts
| | | | | | | | - Patrick C Ma
- Penn State Cancer Institute, Penn State College of Medicine, Penn State University, Hershey, Pennsylvania
| | | | | | - Jianwei Che
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Alexander Drilon
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| | - Pasi A Jänne
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mark M Awad
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
13
|
Shinomiya R, Sato Y, Yoshimoto T, Kawaguchi T, Hirao A, Okamoto K, Kawano Y, Sogabe M, Miyamoto H, Takayama T. A case of treatment-resistant advanced gastric cancer with FGFR2 gene alteration successfully treated with pemigatinib. Int Cancer Conf J 2024; 13:240-244. [PMID: 38962030 PMCID: PMC11217231 DOI: 10.1007/s13691-024-00669-3] [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: 01/18/2024] [Accepted: 02/17/2024] [Indexed: 07/05/2024] Open
Abstract
Comprehensive genome profiling (CGP) is expected to widen the scope of cancer drug options by identifying the genes involved in carcinogenesis. However, a few patients can access recommended treatments following CGP. Herein, we report a case in which pemigatinib, a selective fibroblast growth factor receptor (FGFR) inhibitor, was used as last-line therapy to treat a patient with advanced gastric cancer exhibiting FGFR2 genomic alterations, as determined by CGP testing. The patient (male, 52 years old) was diagnosed with advanced gastric cancer (cStage IV, cT4aN3M1 [LYM], por, HER2 0, microsatellite stable) and received docetaxel + cisplatin + S-1 (7 cycles), irinotecan + ramucirumab (11 cycles), and nivolumab (3 cycles), but experienced progressive disease (PD). Subsequently, FoundationOne Liquid CDx testing was conducted, revealing FGFR2 rearrangement and amplification; however, no clinical trials on genotype-matched therapies for FGFR2 alterations were available. After three cycles of TAS-102, the patient experienced PD and provided consent for the off-label use of pemigatinib. The Cancer Genomics Medical Committee of our hospital approved the self-funded treatment. The patient had markedly decreased CEA and CA19-9 levels after treatment initiation, but experienced PD after five courses. Over the treatment course, grade 1 hyperphosphatemia and onychomadesis were observed. To the best of our knowledge, this is the first reported case of pemigatinib therapy employed in a patient with advanced gastric cancer exhibiting FGFR2 gene alterations. This case could serve as a notable example of tumor-agnostic therapy to broaden treatment options for gastric cancer patients with rare genetic alterations.
Collapse
Affiliation(s)
- Ryo Shinomiya
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Medical Science, 3-18-15, Kuramotocho, Tokushima, 770-8503 Japan
| | - Yasushi Sato
- Department of Community Medicine for Gastroenterology and Oncology, Tokushima University Graduate School of Medical Science, 3-18-15, Kuramoto-cho, Tokushima, 770-8503 Japan
| | - Takanori Yoshimoto
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Medical Science, 3-18-15, Kuramotocho, Tokushima, 770-8503 Japan
| | - Tomoyuki Kawaguchi
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Medical Science, 3-18-15, Kuramotocho, Tokushima, 770-8503 Japan
| | - Akihiro Hirao
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Medical Science, 3-18-15, Kuramotocho, Tokushima, 770-8503 Japan
| | - Koichi Okamoto
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Medical Science, 3-18-15, Kuramotocho, Tokushima, 770-8503 Japan
| | - Yutaka Kawano
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Medical Science, 3-18-15, Kuramotocho, Tokushima, 770-8503 Japan
| | - Masahiro Sogabe
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Medical Science, 3-18-15, Kuramotocho, Tokushima, 770-8503 Japan
| | - Hiroshi Miyamoto
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Medical Science, 3-18-15, Kuramotocho, Tokushima, 770-8503 Japan
| | - Tetsuji Takayama
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Medical Science, 3-18-15, Kuramotocho, Tokushima, 770-8503 Japan
| |
Collapse
|
14
|
Ntowe KW, Lee MS, Plichta JK. Clinical genetics in breast cancer. J Surg Oncol 2024; 130:16-22. [PMID: 38557982 PMCID: PMC11246818 DOI: 10.1002/jso.27630] [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: 01/29/2024] [Accepted: 03/11/2024] [Indexed: 04/04/2024]
Abstract
As genetic testing becomes increasingly more accessible and more applicable with a broader range of clinical implications, it may also become more challenging for breast cancer providers to remain up-to-date. This review outlines some of the current clinical guidelines and recent literature surrounding germline genetic testing, as well as genomic testing, in the screening, prevention, diagnosis, and treatment of breast cancer, while identifying potential areas of further research.
Collapse
Affiliation(s)
- Koumani W. Ntowe
- Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Michael S. Lee
- Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Jennifer K. Plichta
- Department of Surgery, Duke University Medical Center, Durham, North Carolina
- Duke Cancer Institute, Duke University, Durham, North Carolina
- Department of Population Health Sciences, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
15
|
Nannya Y. Addressing Information Gaps and Revising Coverage Terms and Conditions for Cancer Panel Testing. JMA J 2024; 7:267-268. [PMID: 38721075 PMCID: PMC11074507 DOI: 10.31662/jmaj.2024-0014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 02/05/2024] [Indexed: 01/06/2025] Open
Affiliation(s)
- Yasuhito Nannya
- Division of Hematopoietic Disease Control, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
16
|
Yamamoto N, Tolcher A, Hafez N, Lugowska I, Ramlau R, Macarulla T, Geng J, Li J, Teufel M, Märten A, LoRusso P. Efficacy and Safety of the MDM2-p53 Antagonist Brigimadlin (BI 907828) in Patients with Advanced Biliary Tract Cancer: A Case Series. Onco Targets Ther 2024; 17:267-280. [PMID: 38567193 PMCID: PMC10986405 DOI: 10.2147/ott.s440979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 03/15/2024] [Indexed: 04/04/2024] Open
Abstract
Background In patients with advanced biliary tract cancer (BTC), first-line chemotherapy plus immunotherapy has improved outcomes; however, second-line options that reflect the disease's molecular heterogeneity are still needed. One emerging target is MDM2, amplified in ~5-8% of BTC cases. Methods This is a subset analysis of two ongoing Phase Ia/Ib trials assessing patients treated with brigimadlin (BI 907828; a highly potent, oral MDM2-p53 antagonist) ± ezabenlimab (PD-1 inhibitor) ± BI 754111 (anti-LAG-3; n = 1). Results Results from 12 patients with BTC are shown (monotherapy: n = 6/combination: n = 6). Six patients achieved partial response (monotherapy: n = 2/combination: n = 4), four had stable disease; responses were durable. Brigimadlin had a manageable safety profile. Seven patients had dose reductions due to adverse events, but no treatment-related adverse events led to treatment discontinuation. Conclusion Brigimadlin demonstrated anti-tumor activity in patients with advanced MDM2-amplified BTC, and warrants further investigation.
Collapse
Affiliation(s)
- Noboru Yamamoto
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | | | - Navid Hafez
- Yale Comprehensive Cancer Center, Yale School of Medicine, New Haven, CT, USA
- The Angeles Clinic and Research Institute, A Cedars-Sinai Affiliate, Los Angeles, CA, USA
| | - Iwona Lugowska
- Early Phase Clinical Trials Unit, Maria Skłodowska Curie National Research Institute of Oncology, Warsaw, Poland
| | - Rodryg Ramlau
- Institute of Oncology, Poznan University of Medical Sciences, Poznan, Poland
| | - Teresa Macarulla
- Vall d’Hebrón University Hospital, Barcelona, Spain
- Vall d’Hebrón Institute of Oncology (VHIO), Barcelona, Spain
| | - Junxian Geng
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| | - Jian Li
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| | - Michael Teufel
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| | - Angela Märten
- Boehringer Ingelheim International GmbH, Ingelheim am Rhein, Germany
| | - Patricia LoRusso
- Yale Comprehensive Cancer Center, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
17
|
Kage H, Akiyama N, Chang H, Shinozaki‐Ushiku A, Ka M, Kawata J, Muto M, Okuma Y, Okita N, Tsuchihara K, Kikuchi J, Shirota H, Hayashi H, Kokuryo T, Yachida S, Hirasawa A, Kubo M, Kenmotsu H, Tanabe M, Ushiku T, Muto K, Seto Y, Oda K. Patient survey on cancer genomic medicine in Japan under the national health insurance system. Cancer Sci 2024; 115:954-962. [PMID: 38273803 PMCID: PMC10920978 DOI: 10.1111/cas.16065] [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: 09/06/2023] [Revised: 12/09/2023] [Accepted: 12/17/2023] [Indexed: 01/27/2024] Open
Abstract
In Japan, comprehensive genomic profiling (CGP) tests have been reimbursed under the national health care system for solid cancer patients who have finished standard treatment. More than 50,000 patients have taken the test since June 2019. We performed a nation-wide questionnaire survey between March 2021 and July 2022. Questionnaires were sent to 80 designated Cancer Genomic Medicine Hospitals. Of the 933 responses received, 370 (39.7%) were web based and 563 (60.3%) were paper based. Most patients (784, 84%) first learned about CGP tests from healthcare professionals, and 775 (83.1%) gave informed consent to their treating physician. At the time of informed consent, they were most worried about test results not leading to novel treatment (536, 57.4%). On a scale of 0-10, 702 respondents (75.2%) felt that the explanations of the test result were easy to understand (7 or higher). Ninety-one patients (9.8%) started their recommended treatment. Many patients could not receive recommended treatment because no approved drugs or clinical trials were available (102/177, 57.6%). Ninety-eight patients (10.5%) did not wish their findings to be disclosed. Overall satisfaction with the CGP test process was high, with 602 respondents (64.5%) giving a score of 7-10. The major reason for choosing 0-6 was that the CGP test result did not lead to new treatment (217/277, 78.3%). In conclusion, satisfaction with the CGP test process was high. Patients and family members need better access to information. More patients need to be treated with genomically matched therapy.
Collapse
Affiliation(s)
- Hidenori Kage
- Department of Clinical GenomicsThe University of Tokyo HospitalTokyoJapan
- Next‐Generation Precision Medicine Development Laboratory, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Nana Akiyama
- Department of Clinical GenomicsThe University of Tokyo HospitalTokyoJapan
| | - Hyangri Chang
- Department of Clinical GenomicsThe University of Tokyo HospitalTokyoJapan
| | - Aya Shinozaki‐Ushiku
- Department of Clinical GenomicsThe University of Tokyo HospitalTokyoJapan
- Division of Integrative Genomics, Graduate School of MedicineThe University of TokyoTokyoJapan
- Department of Pathology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Mirei Ka
- Division of Integrative Genomics, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Junichi Kawata
- Department of Public PolicyThe Institute of Medical Science, The University of TokyoTokyoJapan
| | - Manabu Muto
- Department of Therapeutic OncologyKyoto University Graduate School of MedicineKyotoJapan
| | - Yusuke Okuma
- Center for Cancer Genomics and Advanced TherapeuticsNational Cancer CenterTokyoJapan
| | - Natsuko Okita
- Center for Cancer Genomics and Advanced TherapeuticsNational Cancer CenterTokyoJapan
| | - Katsuya Tsuchihara
- Department of Genetic Medicine and ServicesNational Cancer Center Hospital EastChibaJapan
| | - Junko Kikuchi
- Division of Clinical Cancer GenomicsHokkaido University HospitalSapporoJapan
| | - Hidekazu Shirota
- Department of Clinical OncologyTohoku University HospitalSendaiJapan
| | - Hideyuki Hayashi
- Genomics Unit, Keio Cancer CenterKeio University School of MedicineTokyoJapan
| | - Toshio Kokuryo
- Division of Surgical Oncology, Department of SurgeryNagoya University Graduate School of MedicineNagoyaJapan
| | - Shinichi Yachida
- Department of Cancer Genome Informatics, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Akira Hirasawa
- Department of Clinical Genomic MedicineOkayama University Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayamaJapan
| | - Makoto Kubo
- Department of Breast Surgical OncologyKyushu University HospitalFukuokaJapan
| | | | - Masahiko Tanabe
- Department of Breast and Endocrine Surgery, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Kaori Muto
- Department of Public PolicyThe Institute of Medical Science, The University of TokyoTokyoJapan
| | - Yasuyuki Seto
- Department of Gastrointestinal Surgery, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Katsutoshi Oda
- Department of Clinical GenomicsThe University of Tokyo HospitalTokyoJapan
- Division of Integrative Genomics, Graduate School of MedicineThe University of TokyoTokyoJapan
| |
Collapse
|
18
|
Damaraju N, Miller AL, Miller DE. Long-Read DNA and RNA Sequencing to Streamline Clinical Genetic Testing and Reduce Barriers to Comprehensive Genetic Testing. J Appl Lab Med 2024; 9:138-150. [PMID: 38167773 DOI: 10.1093/jalm/jfad107] [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: 08/18/2023] [Accepted: 10/24/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Obtaining a precise molecular diagnosis through clinical genetic testing provides information about disease prognosis or progression, allows accurate counseling about recurrence risk, and empowers individuals to benefit from precision therapies or take part in N-of-1 trials. Unfortunately, more than half of individuals with a suspected Mendelian condition remain undiagnosed after a comprehensive clinical evaluation, and the results of any individual clinical genetic test ordered during a typical evaluation may take weeks or months to return. Furthermore, commonly used technologies, such as short-read sequencing, are limited in the types of disease-causing variation they can identify. New technologies, such as long-read sequencing (LRS), are poised to solve these problems. CONTENT Recent technical advances have improved accuracy, increased throughput, and decreased the costs of commercially available LRS technologies. This has resolved many historical concerns about the use of LRS in the clinical environment and opened the door to widespread clinical adoption of LRS. Here, we review LRS technology, how it has been used in the research setting to clarify complex variants or identify disease-causing variation missed by prior clinical testing, and how it may be used clinically in the near future. SUMMARY LRS is unique in that, as a single data source, it has the potential to replace nearly every other clinical genetic test offered today. When analyzed in a stepwise fashion, LRS will simplify laboratory processes, reduce barriers to comprehensive genetic testing, increase the rate of genetic diagnoses, and shorten the amount of time required to make a molecular diagnosis.
Collapse
Affiliation(s)
- Nikhita Damaraju
- Institute for Public Health Genetics, University of Washington, Seattle, WA 98195, United States
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, United States
| | - Angela L Miller
- Department of Pediatrics, University of Washington, Seattle, WA 98195, United States
| | - Danny E Miller
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, United States
- Department of Pediatrics, University of Washington, Seattle, WA 98195, United States
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA 98195, United States
| |
Collapse
|
19
|
Olsen S, Nakamura Y. Serial comprehensive genomic profiling by next-generation sequencing for patients with metastatic colorectal cancer. Cancer Sci 2024; 115:321-323. [PMID: 37950591 PMCID: PMC10823270 DOI: 10.1111/cas.16016] [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: 08/08/2023] [Revised: 10/17/2023] [Accepted: 10/28/2023] [Indexed: 11/12/2023] Open
Abstract
In a real‐world database of metastatic colorectal cancer (mCRC) in the United States, comprehensive genomic profiling (CGP) using plasma‐based next‐generation sequencing identified potentially actionable profiles for approximately one‐third of the patients with both the first test and again with a second test after disease progression, with around 60% of all patients tested receiving therapy consistent with the CGP results at each line of treatment. This suggests a role for CGP prior to more than one line of mCRC treatment.
Collapse
Affiliation(s)
- Steven Olsen
- Department of Clinical and Medical AffairsGuardant Health JapanTokyoJapan
| | - Yoshiaki Nakamura
- Department of Gastroenterology and Gastrointestinal OncologyNational Cancer Center Hospital EastChibaJapan
| |
Collapse
|