1
|
Pizzamiglio M, Soulabaille A, Lahlou W, Pilla L, Zaanan A, Taieb J. Advances and challenges in targeted therapies for HER2-amplified colorectal cancer. Eur J Cancer 2025; 222:115471. [PMID: 40311507 DOI: 10.1016/j.ejca.2025.115471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/16/2025] [Accepted: 04/18/2025] [Indexed: 05/03/2025]
Abstract
Colorectal cancer is the third most common cancer in terms of incidence rate in adults and the second most common cause of cancer-related death in Europe. Despite an increase in overall survival throughout the years, the prognosis of metastatic colorectal cancer remains poor. Until recently, its treatment was based on the use of standard chemotherapy combined with, anti-epidermal growth factor receptor (for RAS wild-type tumors) or anti-vascular endothelial growth factor, or immunotherapy for tumors with mismatch repair deficiency. Over the last years, precision medicine has become a challenge in oncology and there has been an increasing development of biomarker-driven therapies for metastatic colorectal cancer leading to better outcomes for specific molecular subgroups of patients. Human epidermal growth factor receptor 2 (HER2) amplification/overexpression has been identified in about 6 % of patients with RAS wild-type metastatic CRC and established as an important and drugable biomarker. Its prognostic and predictive implications are still debated but HER2 becoming a therapeutic target with promising results of anti-HER2 therapies for HER2-positive metastatic CRC. Multiple HER2-targeted regimens are now part of National Comprehensive Cancer Network and European Society for Medical Oncology guidelines with two recent Food and Drug Administration approvals for previously treated HER2-positive metastatic colorectal cancer for tucatinib (in combination with trastuzumab) and for trastuzumab-deruxtecan in patients with previously treated HER2-positive metastatic colorectal cancer. This review explores the prognostic and predictive value of HER2 as a biomarker in CRC, describing its molecular structure, the clinical characteristics of patients with HER2 alterations, diagnostic approaches and the most relevant clinical trials assessing its current and future role as a therapeutic target in metastatic colorectal cancer.
Collapse
Affiliation(s)
- Margot Pizzamiglio
- Université Paris Cité, Assistance Publique - Hôpitaux de Paris, Department of Digestive Oncology, Hôpital européen Georges Pompidou, Paris, France
| | - Audrey Soulabaille
- Université Paris Cité, Assistance Publique - Hôpitaux de Paris, Department of Digestive Oncology, Hôpital européen Georges Pompidou, Paris, France
| | - Widad Lahlou
- Université Paris Cité, Assistance Publique - Hôpitaux de Paris, Department of Digestive Oncology, Hôpital européen Georges Pompidou, Paris, France
| | - Lorenzo Pilla
- Université Paris Cité, Assistance Publique - Hôpitaux de Paris, Department of Digestive Oncology, Hôpital européen Georges Pompidou, Paris, France
| | - Aziz Zaanan
- Université Paris Cité, Assistance Publique - Hôpitaux de Paris, Department of Digestive Oncology, Hôpital européen Georges Pompidou, Paris, France
| | - Julien Taieb
- Université Paris Cité, Assistance Publique - Hôpitaux de Paris, Department of Digestive Oncology, Hôpital européen Georges Pompidou, Paris, France.
| |
Collapse
|
2
|
Liu WM, Li XB. East meets West: The winning combination against BRAF V600E metastatic colorectal cancer. World J Clin Oncol 2025; 16:102223. [DOI: 10.5306/wjco.v16.i5.102223] [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: 10/12/2024] [Revised: 02/10/2025] [Accepted: 02/19/2025] [Indexed: 05/19/2025] Open
Abstract
Metastatic colorectal cancer (mCRC) patients with BRAF V600E mutation have a poor prognosis despite the implementation of multiple treatment strategies. The integration of traditional Chinese medicine with Western medicine in treating BRAF mutant mCRC has garnered increasing attention. Recent studies indicate that combining traditional Chinese and modern Western medical approaches not only extend survival but also reduces the risk of mortality in patients with BRAF V600E mutant mCRC. This approach is particularly effective for colorectal cancer patients who have right-sided colon involvement, liver metastasis, or a history of radiotherapy or chemotherapy. In this treatment combination, traditional Chinese medicine may offer symptomatic relief and improve quality of life, while Western medicine targets the disease more aggressively with advanced pharmacological agents. Ongoing research is crucial to further elucidate the mechanisms underlying these benefits and to optimize treatment protocols.
Collapse
Affiliation(s)
- Wen-Ming Liu
- Department of Gastrointestinal Surgery, The First People’s Hospital of Tianmen, Tianmen 431700, Hubei Province, China
| | - Xiao-Bing Li
- Department of Thoracic Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430079, Hubei Province, China
| |
Collapse
|
3
|
Meng X, Lu Z, Mi F, Sha S, Li T. Research hotspots and emerging trends in targeted therapy for colorectal cancer: a bibliometric analysis (2000-2023). Discov Oncol 2025; 16:789. [PMID: 40380023 PMCID: PMC12084209 DOI: 10.1007/s12672-025-02632-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 05/08/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND Targeted therapy has significantly transformed the treatment landscape of colorectal cancer (CRC), enabling personalized treatment approaches and improving patient prognosis. This study employs bibliometric analysis to explore the research hotspots and development trends in the field of CRC-targeted therapy from 2000 to 2023. METHODS Based on the Web of Science Core Collection, this study collected literature related to CRC-targeted therapy published between 2000 and 2023. CiteSpace and VOSviewer were used for data analysis, with a focus on publication trends, key contributors, and keyword co-occurrence patterns. RESULTS A total of 2252 relevant articles were included, demonstrating a steady growth trend in research output. China ranked first in terms of the number of publications, while the University of Texas MD Anderson Cancer Center was identified as the institution with the highest research output. Josep Tabernero was the most prolific author in this field. Among journals, Cancers had the highest impact, while Clinical Cancer Research held a significant advantage in citation frequency. Keyword co-occurrence and clustering analysis indicated that research primarily focused on treatment strategies and precision medicine, with emerging technologies such as cell therapy and liquid biopsy garnering increasing attention. CONCLUSION This study reveals the research trends, core hotspots, and emerging directions in the field of CRC-targeted therapy, providing valuable insights for future research.
Collapse
Affiliation(s)
- Xiangnv Meng
- General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Second Department of Medical Oncology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Zhongting Lu
- General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Fu Mi
- Second Department of Medical Oncology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Sha Sha
- Second Department of Medical Oncology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Tao Li
- Department of Surgical Oncology, Tumor Hospital, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China.
| |
Collapse
|
4
|
Taieb J, Souglakos J, Boukovinas I, Falcoz A, Pages F, Messaritakis I, Bennouna J, Artru P, Louvet C, Lepere C, Emile JF, Bouche O, Mazard T, Vernerey D, Vogiatzoglou K, Tzardi M, Sharma S, Liu MC, Sethi H, André T, Galon J, Laurent-Puig P. Combined Analyses of Circulating Tumor DNA and Immunoscore in Patients With Stage III Colon Cancer: A Post Hoc Analysis of the PRODIGE-GERCOR IDEA-France/HORG-IDEA-Greece Trials. J Clin Oncol 2025; 43:1564-1577. [PMID: 39903903 PMCID: PMC12054978 DOI: 10.1200/jco.24.00648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 10/08/2024] [Accepted: 12/02/2024] [Indexed: 02/06/2025] Open
Abstract
PURPOSE Immunoscore (IS) and circulating tumor DNA (ctDNA) are two emerging technologies in improving prognostication and tailoring adjuvant treatments in patients resected from a stage III colon cancer (CC). Here, we analyzed the prognostic value of the two biomarkers in patients who participated in the randomized phase III IDEA-France and HORG trials. METHODS Plasma samples were collected after surgery and before adjuvant chemotherapy. ctDNA analysis was performed using a clinically validated, personalized, tumor-informed 16-plex protein chain reaction assay. Multivariable analyses for time to recurrence (TTR; patients without recurrence or death due to CC) and overall survival (OS) were performed using ctDNA and IS results, along with other parameters including treatment duration and disease risk group. RESULTS Of the 554 patients with available ctDNA results, 445 were ctDNA-negative (80.3%) and 109 were ctDNA-positive (19.7%); baseline characteristics showed more T4/N2 and venous embolism/lymphatic invasion/perineural invasion+ in ctDNA-positive patients. With a median follow-up of 6.7 years, the 2-year TTR rate was 43.5% (95% CI, 34.1 to 52.6) for ctDNA-positive patients and 88.1% (95% CI, 84.7 to 90.8) for ctDNA-negative patients (P < .0001). ctDNA was confirmed as an independent prognostic marker for both TTR (adjusted hazard ratio [adjHR], 5.21 [95% CI, 3.59 to 7.58]; P < .001) and OS (adjHR, 4.84 [95% CI, 3.40 to 6.89]; P < .001). ctDNA remained the most significant prognostic factor irrespective of disease stage, treatment duration, and IS results. IS was not prognostic in ctDNA-positive patients but remained a significant prognostic tool for ctDNA-negative patients. CONCLUSION In this combined analysis of two adjuvant trials dedicated to patients with stage III CC after surgery, ctDNA was detectable in 19.7% of the patients and was confirmed as a major independent prognostic biomarker. IS seems to bring additional prognostic information in the 80.3% of patients who are ctDNA-negative.
Collapse
Affiliation(s)
- Julien Taieb
- Department of Gastroenterology and Gastrointestinal Oncology, CARPEM Comprehensive Cancer Center, Georges-Pompidou European Hospital, AP-HP, Paris, France
- SIRIC CARPEM, Université Paris-Cité, Paris, France
- Centre de Recherche des Cordeliers, INSERM, CNRS, Université Paris-Cité, Sorbonne Université, USPC, Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - John Souglakos
- Department of Medical Oncology, University Hospital of Heraklion, Crete, Greece
- Laboratory of Translational Oncology, Medical School, University of Crete, Heraklion, Greece
| | | | - Antoine Falcoz
- Methodology and Quality of Life in Oncology Unit, Besançon University Hospital, Besançon, France
- INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Bourgogne Franche-Comté University, Besançon, France
| | - Franck Pages
- Department of Immunology, Georges-Pompidou European Hospital, AP-HP, INSERM, Laboratory of Integrative Cancer Immunology, Equipe Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers, SIRIC CARPEM, Université Paris-Cité, Paris, France
| | - Ippokratis Messaritakis
- Laboratory of Translational Oncology, Medical School, University of Crete, Heraklion, Greece
| | - Jaafar Bennouna
- Department of Medical Oncology, Hospital Foch, Suresnes, France
| | - Pascal Artru
- Department of Medical Oncology, Private Hospital Jean Mermoz—Ramsay Santé, Lyon, France
| | - Christophe Louvet
- Department of Medical Oncology, Institut Mutualiste Montsouris, Paris, France
| | - Celine Lepere
- Department of Gastroenterology and Gastrointestinal Oncology, CARPEM Comprehensive Cancer Center, Georges-Pompidou European Hospital, AP-HP, Paris, France
- SIRIC CARPEM, Université Paris-Cité, Paris, France
| | - Jean Francois Emile
- Laboratory of Integrative Cancer Immunology, Equipe Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Paris, France
| | - Olivier Bouche
- Department of Digestive Oncology, Hospital Robert Debré, Reims, France
| | - Thibault Mazard
- Department of Medical Oncology, Montpellier Cancer Institute (ICM), Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, University of Montpellier, Montpellier, France
| | - Dewi Vernerey
- Methodology and Quality of Life in Oncology Unit, Besançon University Hospital, Besançon, France
- INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Bourgogne Franche-Comté University, Besançon, France
| | | | - Maria Tzardi
- Laboratory of Pathology, University Hospital of Heraklion, Crete, Greece
| | | | | | | | - Thierry André
- Sorbonne Université, Paris, France
- Department of Medical Oncology, Hôpital Saint Antoine, Paris, France
| | - Jérome Galon
- Laboratory of Integrative Cancer Immunology, Equipe Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers, INSERM, SIRIC CARPEM, Sorbonne Université, Université Paris Cité, Paris, France
| | - Pierre Laurent-Puig
- Centre de Recherche des Cordeliers, INSERM, CNRS, Université Paris-Cité, Sorbonne Université, USPC, Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
- Department of Biology, Assistance Publique—Hôpitaux de Paris, European Georges Pompidou Hospital, Paris, France
| |
Collapse
|
5
|
Zhang J, Zhu H, Liu W, Miao J, Mao Y, Li Q. Prognostic and predictive molecular biomarkers in colorectal cancer. Front Oncol 2025; 15:1532924. [PMID: 40308511 PMCID: PMC12040681 DOI: 10.3389/fonc.2025.1532924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/27/2025] [Indexed: 05/02/2025] Open
Abstract
Precision medicine has brought revolutionary changes to the diagnosis and treatment of cancer patients, and is currently a hot and challenging research topic. Currently, the treatment regimens for most colorectal cancer (CRC) patients are mainly determined by several biomakers, including Microsatellite Instability (MSI), RAS, and BRAF. However, the roles of promising biomarkers such as HER-2, consensus molecular subtypes (CMS), and circulating tumor DNA (ctDNA) in CRC are not yet fully clear. Therefore, it is urgent to explore the potential of these emerging biomarkers in the diagnosis and treatment of CRC patients. In this paper, we discuss recent advances in CRC biomarkers, especially clinical data, and focus on the roles of biomarkers in prognosis, prediction, treatment strategies, and the intrinsic connections with clinical pathological features, hoping to promote better precision medicine for colorectal cancer.
Collapse
Affiliation(s)
- Jianzhi Zhang
- Department of General Surgery, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Hao Zhu
- Department of General Surgery, Affiliated Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wentao Liu
- Department of General Surgery, Affiliated Drum Tower Hospital, JiangSu University, Nanjing, China
| | - Ji Miao
- Department of General Surgery, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Yonghuan Mao
- Department of General Surgery, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Qiang Li
- Department of General Surgery, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| |
Collapse
|
6
|
Singh Raghav KP, Guthrie KA, Tan B, Denlinger CS, Fakih M, Overman MJ, Dasari NA, Corum LR, Hicks LG, Patel MS, Esparaz BT, Kazmi SM, Alluri N, Colby S, Gholami S, Gold PJ, Chiorean EG, Kopetz S, Hochster HS, Philip PA. Trastuzumab Plus Pertuzumab Versus Cetuximab Plus Irinotecan in Patients With RAS/BRAF Wild-Type, HER2-Positive, Metastatic Colorectal Cancer (S1613): A Randomized Phase II Trial. J Clin Oncol 2025; 43:1348-1357. [PMID: 39761503 PMCID: PMC11975499 DOI: 10.1200/jco-24-01710] [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: 08/04/2024] [Revised: 10/14/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025] Open
Abstract
PURPOSE ERBB2 overexpression/amplification in RAS/BRAF wild-type (WT) metastatic colorectal cancer (mCRC; human epidermal growth factor receptor 2 [HER2]-positive mCRC) appears to be associated with limited benefit from anti-EGFR antibodies and promising responses to dual-HER2 inhibition; however, comparative efficacy has not been investigated. We conducted a randomized phase II trial to evaluate efficacy and safety of dual-HER2 inhibition against standard-of-care anti-EGFR antibody-based therapy as second/third-line treatment in HER2-positive mCRC. METHODS Patients with RAS/BRAF-WT mCRC after central confirmation of HER2 positivity (immunohistochemistry 3+ or 2+ and in situ hybridization amplified [HER2/CEP17 ratio >2.0]) were assigned (1:1) to either trastuzumab plus pertuzumab (TP; trastuzumab 6 mg/kg and pertuzumab 420 mg once every 3 weeks) or cetuximab plus irinotecan (CETIRI; cetuximab 500 mg/m2 and irinotecan 180 mg/m2 once every 2 weeks) until progression or unacceptable toxicity. Crossover to TP was allowed after progression on CETIRI. The primary end point was progression-free survival (PFS). Secondary end points included objective response rate (ORR), overall survival, safety, and HER2 gene copy number (GCN ≥20/<20) as a predictive factor. RESULTS Between October 2017 and March 2022, 54 participants were assigned to TP (n = 26) and CETIRI (n = 28). Median PFS did not vary significantly by treatment: 4.7 (95% CI, 1.9 to 7.6) and 3.7 (95% CI, 1.6 to 6.7) months in the TP and CETIRI groups, respectively. Efficacy of TP versus CETIRI differed significantly by HER2 GCN (median PFS, GCN ≥20 [9.9 v 2.9 months] and GCN <20 [3.0 v 4.2 months], respectively; P interaction = .003). On TP, ORR was 34.6% (57.1% with GCN ≥20 v 9.1% with GCN <20) with median GCN of 29.7 versus 13.2 for responders and nonresponders, respectively (P = .004). Grade ≥3 adverse events occurred in 23.1% and 46.1% of participants with TP and CETIRI, respectively. CONCLUSION TP appears to be a safe and effective cytotoxic chemotherapy-free option for patients with RAS/BRAF-WT, HER2-positive mCRC. Higher levels of HER2 amplification were associated with greater degree of clinical benefit from TP vis-à-vis CETIRI.
Collapse
MESH Headings
- Humans
- Female
- Colorectal Neoplasms/drug therapy
- Colorectal Neoplasms/genetics
- Colorectal Neoplasms/pathology
- Colorectal Neoplasms/enzymology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/antagonists & inhibitors
- Irinotecan/administration & dosage
- Irinotecan/adverse effects
- Irinotecan/therapeutic use
- Middle Aged
- Male
- Aged
- Cetuximab/administration & dosage
- Cetuximab/adverse effects
- Cetuximab/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Proto-Oncogene Proteins B-raf/genetics
- Trastuzumab/administration & dosage
- Trastuzumab/adverse effects
- Trastuzumab/therapeutic use
- Adult
- Aged, 80 and over
Collapse
Affiliation(s)
| | - Katherine A. Guthrie
- SWOG Statistics and Data Management Center, Seattle, WA
- Fred Hutchinson Cancer Center, Seattle, WA
| | - Benjamin Tan
- Washinton University Siteman Cancer Center, St. Louis, MO
| | | | | | | | | | - Larry R. Corum
- University of Kansas Cancer Center - MCA Rural MU NCORP/Olathe Health Cancer Center, Olathe, KS
| | - Lee G. Hicks
- Baptist Health Cancer Research Network/Baptist Health Lexington, Lexington, KY
| | - Mital S. Patel
- CORA NCORP, CommonSpirit Health Research Institute/ Cancer Center at Saint Joseph’s, Phoenix, AZ
| | - Benjamin T. Esparaz
- Heartland Cancer Research NCORP/Cancer Care Specialists of Illinois, Decatur, IL
| | - Syed M. Kazmi
- University of Texas Southwestern Medical Center/Parkland Memorial Hospital, Dallas, TX
| | - Nitya Alluri
- Pacific Cancer Research Consortium NCORP/St. Luke’s Cancer Institute, Boise, ID
| | - Sarah Colby
- SWOG Statistics and Data Management Center, Seattle, WA
- Fred Hutchinson Cancer Center, Seattle, WA
| | | | | | - E. Gabriela Chiorean
- Fred Hutchinson Cancer Center, Seattle, WA
- University of Washington School of Medicine, Seattle, WA
| | | | | | - Philip A. Philip
- MD Anderson Cancer Center, Houston, TX
- University of Kansas Cancer Center - MCA Rural MU NCORP/Olathe Health Cancer Center, Olathe, KS
| |
Collapse
|
7
|
Inoue K, Nakamura Y, Caughey B, Zheng-Lin B, Ueno M, Furukawa M, Kawamoto Y, Itoh S, Umemoto K, Sudo K, Satoh T, Mizuno N, Kajiwara T, Fujisawa T, Bando H, Yoshino T, Strickler JH, Morizane C, Bekaii-Saab T, Ikeda M. Clinicomolecular Profile and Efficacy of Human Epidermal Growth Factor Receptor 2 (HER2)-Targeted Therapy for HER2-Amplified Advanced Biliary Tract Cancer. JCO Precis Oncol 2025; 9:e2400718. [PMID: 40209139 PMCID: PMC12005869 DOI: 10.1200/po-24-00718] [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: 10/07/2024] [Revised: 12/17/2024] [Accepted: 02/19/2025] [Indexed: 04/12/2025] Open
Abstract
PURPOSE This study aimed to investigate the clinicomolecular profiles and the efficacy of human epidermal growth factor receptor 2 (HER2)-targeted therapy in HER2-amplified biliary tract cancer (BTC). METHODS This study was an international collaboration that used combined data from the prospective SCRUM-Japan GOZILA and MONSTAR-SCREEN in Japan and retrospective reviews in the United States; patients with advanced BTC who had received systemic therapy were included. The clinicomolecular profiles were evaluated in an exploratory cohort, whereas the efficacy of HER2-targeted therapy was assessed in a biomarker-selected cohort. RESULTS Of the 439 patients in the exploratory cohort, 43 (10%) had HER2 amplification. The frequencies of coalterations were higher in patients with HER2 amplification versus patients without HER2 amplification including HER2 mutations (26% v 5%, P < .001), TP53 mutations (84% v 61%, P = .003), and BRAF amplification (9% v 2%, P = .030). There were no KRAS mutations identified in patients with HER2-amplified BTC. No significant difference in overall survival (OS) was observed between patients with and without HER2 amplification (median, 17.7 v 16.9 months; hazard ratio [HR], 0.95 [95% CI, 0.65 to 1.40]). Of the 60 patients with HER2-amplified BTC in the biomarker-selected cohort (43 from Japan and 17 from the United States), the OS was significantly longer in 29 patients who received HER2-targeted therapy than in those who did not receive HER2-targeted therapy (median, 24.3 v 12.1 months; HR, 0.39 [95% CI, 0.23 to 0.82]). Multivariate analysis identified HER2-targeted therapy as an independent prognostic factor for OS (HR, 0.29 [95% CI, 0.14 to 0.58]; P < .001). CONCLUSION HER2 amplification was found in 10% of advanced BTC and was not identified as an independent prognostic factor for OS. Patients with HER2-amplified BTC derive significant benefit from HER2-targeted therapy.
Collapse
Affiliation(s)
- Kanae Inoue
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshiaki Nakamura
- Translational Research Support Office, Division of Drug and Diagnostic Development Promotion, Department for the Promotion of Drug and Diagnostic Development, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Bennett Caughey
- Division of Hematology/Oncology, Massachusetts General Hospital, Boston, MA
| | - Binbin Zheng-Lin
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Masayuki Furukawa
- Department of Hepato-Biliary-Pancreatology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Yasuyuki Kawamoto
- Division of Cancer Center, Hokkaido University Hospital, Sapporo, Japan
| | - Shinji Itoh
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kumiko Umemoto
- Department of Clinical Oncology, St Marianna University School of Medicine, Kawasaki, Japan
| | - Kentaro Sudo
- Department of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Taroh Satoh
- Center for Cancer Genomics and Precision Medicine, Osaka University Hospital, Osaka, Japan
| | - Nobumasa Mizuno
- Department of Gastroenterology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Takeshi Kajiwara
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Takao Fujisawa
- Translational Research Support Office, Division of Drug and Diagnostic Development Promotion, Department for the Promotion of Drug and Diagnostic Development, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Head and Neck Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hideaki Bando
- Translational Research Support Office, Division of Drug and Diagnostic Development Promotion, Department for the Promotion of Drug and Diagnostic Development, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | | | - Chigusa Morizane
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | | | - Masafumi Ikeda
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
8
|
Fujisawa T, Nakamura Y, Bando H, Morizane C, Ikeda M, Nonomura N, Matsubara N, Iwata H, Naito Y, Okano S, Aoki D, Harano K, Yamazaki N, Namikawa K, Ueno M, Kadowaki S, Oki E, Kato K, Komatsu Y, Satoh T, Esaki T, Denda T, Hamaguchi T, Yamazaki K, Matsuhashi N, Yasui H, Satake H, Nishina T, Takahashi N, Goto M, Sunakawa Y, Kato T, Otsuka T, Abutani H, Tukachinsky H, Lee JK, Oxnard GR, Kuramoto N, Horasawa S, Sakamoto Y, Taniguchi H, Yoshino T. Benefits of Combining Circulating Tumor DNA With Tissue and Longitudinal Circulating Tumor DNA Genotyping in Advanced Solid Tumors: SCRUM-Japan MONSTAR-SCREEN-1 Study. JCO Precis Oncol 2025; 9:e2400283. [PMID: 40209142 PMCID: PMC12005867 DOI: 10.1200/po.24.00283] [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: 04/29/2024] [Revised: 12/23/2024] [Accepted: 02/21/2025] [Indexed: 04/12/2025] Open
Abstract
PURPOSE The utility of capturing heterogeneity by circulating tumor DNA (ctDNA) genotyping combined with tissue analysis or applying it in a sequential manner remains uncertain. METHODS We assessed the clinical value of ctDNA genotyping using data from 2,187 patients with advanced solid tumors enrolled in SCRUM-Japan MONSTAR-SCREEN-1, a nationwide cancer genome screening project, which examined ctDNA from longitudinally collected blood samples and tumor tissue samples (UMIN 000036749). RESULTS Among 667 patients with both baseline ctDNA and tissue genotyping results, 51 (7.6%) had actionable biomarkers identified exclusively through ctDNA genotyping. The most frequent targets of genotype-matched therapy guided by solely ctDNA were immune checkpoint, estrogen receptor, and poly(ADP-ribose) polymerase (PARP). Comparison of objective response rates (ORRs) and progression-free survival (PFS) between patients treated based on tissue versus ctDNA alone showed no significant difference, with ORRs of 34.0% versus 23.1% (P = .54) and a median PFS of 11.5 versus 13.8 months (hazard ratio [HR], 1.4 [95% CI, 0.72 to 2.80]), respectively. Among 924 patients undergoing sequential ctDNA genotyping, the detection of actionable biomarkers increased from 63.2% to 72.5% following subsequent ctDNA. Targets for genotype-matched therapy guided by subsequent ctDNA alone commonly included PARP, immune checkpoint, and BRAF. The ORR was 23.2% and 26.7% (P = .75), and the median PFS was 5.2 and. 3.7 months (HR, 1.5 [95% CI, 0.79 to 2.80]) for genotype-matched therapy based on initial versus subsequent ctDNA alone, respectively. CONCLUSION Combining ctDNA with tissue analysis, followed by sequential ctDNA assessments, effectively enhances the identification of actionable biomarkers. This strategy facilitates clinically beneficial, genetically informed therapies, underscoring its significant value in precision oncology.
Collapse
Affiliation(s)
- Takao Fujisawa
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Head and Neck Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Course of Advanced Clinical Research of Cancer, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshiaki Nakamura
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- International Research Promotion Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hideaki Bando
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Chigusa Morizane
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Masafumi Ikeda
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Nobuaki Matsubara
- Department of Medical Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Hiroji Iwata
- Department of Breast Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Yoichi Naito
- Department of Medical Oncology, National Cancer Center Hospital East, Chiba, Japan
- Department of General Internal Medicine, National Cancer Center Hospital East, Chiba, Japan
- Department of Experimental Therapeutics, National Cancer Center Hospital East, Chiba, Japan
| | - Susumu Okano
- Department of Head and Neck Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Daisuke Aoki
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Kenichi Harano
- Department of Medical Oncology, National Cancer Center Hospital East, Chiba, Japan
- Department of Experimental Therapeutics, National Cancer Center Hospital East, Chiba, Japan
| | - Naoya Yamazaki
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kenjiro Namikawa
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Shigenori Kadowaki
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Eiji Oki
- Department of Surgery and Science, Kyushu University, Fukuoka, Japan
| | - Ken Kato
- Department of Head and Neck, Esophageal Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yoshito Komatsu
- Department of Cancer Center, Hokkaido University Hospital, Sapporo, Japan
| | - Taroh Satoh
- Center for Cancer Genomics and Precision Medicine, Osaka University Hospital, Suita, Japan
| | - Taito Esaki
- Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Tadamichi Denda
- Division of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Tetsuya Hamaguchi
- Department of Gastroenterological Oncology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Kentaro Yamazaki
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shunto-gun, Japan
| | - Nobuhisa Matsuhashi
- Department of Gastroenterological Surgery and Pediatric Surgery, Center for One Medicine Innovative Translational Research, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hisateru Yasui
- Department of Medical Oncology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Hironaga Satake
- Cancer Center, Kansai Medical University Hospital, Hirakata, Japan
- Department of Medical Oncology, Kochi Medical School, Kochi, Japan
| | - Tomohiro Nishina
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Naoki Takahashi
- Department of Gastroenterology, Saitama Cancer Center, Kitaadachi-gun, Japan
| | - Masahiro Goto
- Cancer Chemotherapy Center, Osaka Medical and Pharmaceutical University Hospital, Takatsuki, Japan
| | - Yu Sunakawa
- Department of Clinical Oncology, St Marianna University School of Medicine, Kawasaki, Japan
| | - Takeshi Kato
- Department of Surgery, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Tomoyuki Otsuka
- Department of Medical Oncology, Osaka International Cancer Institute, Osaka, Japan
| | | | | | | | | | - Naomi Kuramoto
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Satoshi Horasawa
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yasutoshi Sakamoto
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hiroya Taniguchi
- Department of Surgery and Science, Kyushu University, Fukuoka, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Department of the Promotion of Drug and Diagnostic Development, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
9
|
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
|
10
|
Xiong Y, Cheng L, Zhou YJ, Ge WH, Qian M, Yang H. Diagnosis and treatment of lung cancer: A molecular perspective. World J Clin Oncol 2025; 16:100361. [PMID: 40130062 PMCID: PMC11866089 DOI: 10.5306/wjco.v16.i3.100361] [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: 08/15/2024] [Revised: 12/23/2024] [Accepted: 01/09/2025] [Indexed: 01/21/2025] Open
Abstract
This editorial comments on the review by Da Silva et al, published in the World Journal of Clinical Oncology which focuses on the molecular perspectives of lung cancer. With the rapid development of molecular technology, new diagnostic methods are constantly emerging, including liquid biopsy, the identification of gene mutations, and the monitoring biomarkers, thus providing precise information with which to identify the occurrence and development of lung cancer. Biomarkers, such as circulating tumor cells, circulating tumor DNA, and circulating RNA can provide helpful information for clinical application. Common types of genetic mutations and immune checkpoints include epidermal growth factor receptor, anaplastic lymphoma kinase, c-ROS proto-oncogene 1, programmed death-1 and cytotoxic T-lymphocyte-associated protein. According to specific biomarkers, targeted therapy and immunotherapy can improve survival outcomes based on the types of gene mutation and immune checkpoints. The application of molecular approaches can facilitate our ability to control the progression of disease and select appropriate therapeutic strategies for patients with lung cancer.
Collapse
Affiliation(s)
- Yuan Xiong
- Department of Pharmacy, Nanjing Drum Tower Hospital, Nanjing 210008, Jiangsu Province, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu Province, China
| | - Long Cheng
- Department of Pharmacy, Nanjing Drum Tower Hospital, Nanjing 210008, Jiangsu Province, China
- Nanjing Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, Nanjing 210008, Jiangsu Province, China
| | - Yu-Jie Zhou
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Nanjing 210008, Jiangsu Province, China
| | - Wei-Hong Ge
- Department of Pharmacy, Nanjing Drum Tower Hospital, Nanjing 210008, Jiangsu Province, China
| | - Ming Qian
- Department of Pharmacy, Nanjing Drum Tower Hospital, Nanjing 210008, Jiangsu Province, China
| | - Hui Yang
- Department of Pharmacy, Nanjing Drum Tower Hospital, Nanjing 210008, Jiangsu Province, China
| |
Collapse
|
11
|
Bartolomucci A, Nobrega M, Ferrier T, Dickinson K, Kaorey N, Nadeau A, Castillo A, Burnier JV. Circulating tumor DNA to monitor treatment response in solid tumors and advance precision oncology. NPJ Precis Oncol 2025; 9:84. [PMID: 40122951 PMCID: PMC11930993 DOI: 10.1038/s41698-025-00876-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: 10/02/2024] [Accepted: 03/11/2025] [Indexed: 03/25/2025] Open
Abstract
Circulating tumor DNA (ctDNA) has emerged as a dynamic biomarker in cancer, as evidenced by its increasing integration into clinical practice. Carrying tumor specific characteristics, ctDNA can be used to inform treatment selection, monitor response, and identify drug resistance. In this review, we provide a comprehensive, up-to-date summary of ctDNA in monitoring treatment response with a focus on lung, colorectal, and breast cancers, and discuss current challenges and future directions.
Collapse
Affiliation(s)
- Alexandra Bartolomucci
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Monyse Nobrega
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Tadhg Ferrier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Kyle Dickinson
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Nivedita Kaorey
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Amélie Nadeau
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Alberto Castillo
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Julia V Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
- Department of Pathology, McGill University, Montreal, QC, Canada.
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada.
| |
Collapse
|
12
|
Yan HC, Liu Y, Feng Y, Li JM, Sheng LM, Chen X, Xie YP, Li N. Efficacy of disitamab vedotin-containing therapy in metastatic colorectal cancer: A case report. World J Clin Oncol 2025; 16:99527. [PMID: 40130050 PMCID: PMC11866092 DOI: 10.5306/wjco.v16.i3.99527] [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: 07/24/2024] [Revised: 11/10/2024] [Accepted: 12/30/2024] [Indexed: 01/21/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a leading cause of cancer-related mortality worldwide. In cases of metastatic CRC (mCRC) that are resistant to conventional chemotherapy-based treatments, the efficacy of available therapeutic options is typically low. CRC exhibiting overexpression or amplification of the human epidermal growth factor receptor 2 (HER2) gene has shown responsiveness to HER2-targeted therapies. CASE SUMMARY We present the case of a 69-year-old woman diagnosed with mCRC with an NRAS p.G12V mutation and microsatellite stability, identified through tumor sequencing, along with HER2 overexpression detected by immunohistochemistry. She exhibited an excellent response to disitamab vedotin-containing therapy. To our knowledge, this is the first reported case of mCRC with HER2 overexpression and an NRAS p.G12V mutation achieving a remarkable clinical response to anti-HER2 therapy. CONCLUSION Disitamab vedotin demonstrates promising anti-tumor effects in HER2-overexpressing mCRC, offering patients an additional treatment option.
Collapse
Affiliation(s)
- Hu-Cheng Yan
- Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan Province, China
- Department of Oncology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yan Liu
- Department of Oncology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - You Feng
- Department of Oncology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Jun-Ming Li
- Department of Radiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Lei-Ming Sheng
- Department of Oncology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Xin Chen
- Department of Oncology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yu-Ping Xie
- Department of Oncology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Na Li
- Department of Oncology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
13
|
Cohen R, Raeisi M, Chibaudel B, Yothers G, Goldberg RM, Bachet JB, Wolmark N, Yoshino T, Schmoll HJ, Haller DG, Kerr R, Lonardi S, George TJ, Shacham-Shmueli E, Shi Q, André T, de Gramont A. Impact of tumor and node stages on the efficacy of adjuvant oxaliplatin-based chemotherapy in stage III colon cancer patients: an ACCENT pooled analysis. ESMO Open 2025; 10:104481. [PMID: 40043353 PMCID: PMC11928968 DOI: 10.1016/j.esmoop.2025.104481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/30/2025] [Accepted: 02/04/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Standard adjuvant treatment of stage III colon cancer (CC) is fluoropyrimidine with oxaliplatin. Recently, stage III was subdivided into low-risk (T1-3, N1) and high-risk (T4 and/or N2), with the benefit of adding oxaliplatin varying across these substages. In this study, we aimed to assess the impact of oxaliplatin on survival outcomes in subdividing stage III CC patients based on T and N staging. PATIENTS AND METHODS A total of 4942 stage III CC patients were pooled from the three randomized pivotal trials of oxaliplatin. Kaplan-Meier curves, Cox models stratified by study, and interaction tests were used to assess the oxaliplatin effect across subgroups based on T and N stages. The primary endpoint was overall survival (OS). RESULTS The prevalence of tumor stages was T1-2 12.4%, T3 74.4%, and T4 13.1%; nodal stages were N1 64.7% and N2 35.3%. A significant OS benefit from oxaliplatin was seen only in T3 (5-year OS = 77.2% versus 73.0%, P < 0.001): T3N1 (hazard ratio 0.72, 95% confidence interval 0.62-0.85, P < 0.001) and T3N2 (hazard ratio 0.81, 95% confidence interval 0.69-0.95, P = 0.010). No benefit was observed for T1-2 (5-year OS = 87.8% versus 88.7%, P = 0.644) or T4 patients (5-year OS = 62.6% versus 60.2%, P = 0.648). Subgroup analysis revealed a significant interaction between T stage and the effect of oxaliplatin treatment on OS, whereas no such interaction was observed for N stage. CONCLUSIONS Our analysis revealed that oxaliplatin-based chemotherapy offers a significant survival benefit in stage III CC patients with T3 tumors. In contrast, no survival benefit was observed for T1-2 or T4 patients. These results suggested that T stage plays a more crucial role than N stage in predicting treatment benefit, highlighting the need for tailored treatment strategies based on tumor characteristics.
Collapse
Affiliation(s)
- R Cohen
- Sorbonne University, Department of Medical Oncology, Hôpital Saint-Antoine, AP-HP, and INSERM UMRS 938, Équipe Instabilité des Microsatellites et Cancer, Équipe Labellisée par la Ligue Nationale Contre le Cancer, SIRIC CURAMUS, Centre de recherche Saint Antoine, Paris, France
| | - M Raeisi
- Statistical Unit, ARCAD Foundation, Paris, France.
| | - B Chibaudel
- Department of Medical Oncology, Franco-British Hospital, Levallois-Perret, France
| | - G Yothers
- Department of Biostatistics, University of Pittsburgh and NRG Oncology, Pittsburgh, USA. https://twitter.com/GregYothers
| | - R M Goldberg
- Department of Oncology, West Virginia University Cancer Institute, Morgantown, USA
| | - J-B Bachet
- Hepato-gastroenterology and Digestive Oncology Department, Sorbonne University, Pitié Salpêtrière Hospital, APHP, Paris, France
| | | | - T Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - H-J Schmoll
- Division of Clinical Research in Oncology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - D G Haller
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, USA
| | - R Kerr
- Department of Oncology, University of Oxford, Oxford, UK
| | - S Lonardi
- Medical Oncology, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy. https://twitter.com/sara_lonardi1
| | - T J George
- Division of Hematology and Oncology, University of Florida, Gainesville, USA. https://twitter.com/TGeorgeMD
| | - E Shacham-Shmueli
- Sheba Medical Center at Tel-Hashomer, Tel Aviv University, Tel Aviv, Israel
| | - Q Shi
- Department of Quantitative Health Science, Mayo Clinic, Rochester, USA
| | - T André
- Sorbonne University, Department of Medical Oncology, Hôpital Saint-Antoine, AP-HP, and INSERM UMRS 938, Équipe Instabilité des Microsatellites et Cancer, Équipe Labellisée par la Ligue Nationale Contre le Cancer, SIRIC CURAMUS, Centre de recherche Saint Antoine, Paris, France; ARCAD Foundation, Paris, France
| | - A de Gramont
- Department of Medical Oncology, Franco-British Hospital, Levallois-Perret, France; ARCAD Foundation, Paris, France
| |
Collapse
|
14
|
Okines AFC, Curigliano G, Mizuno N, Oh DY, Rorive A, Soliman H, Takahashi S, Bekaii-Saab T, Burkard ME, Chung KY, Debruyne PR, Fox JR, Gambardella V, Gil-Martin M, Hamilton EP, Monk BJ, Nakamura Y, Nguyen D, O'Malley DM, Olawaiye AB, Pothuri B, Reck M, Sudo K, Sunakawa Y, Van Marcke C, Yu EY, Ramos J, Tan S, Bieda M, Stinchcombe TE, Pohlmann PR. Tucatinib and trastuzumab in HER2-mutated metastatic breast cancer: a phase 2 basket trial. Nat Med 2025; 31:909-916. [PMID: 39825152 PMCID: PMC11922774 DOI: 10.1038/s41591-024-03462-0] [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: 04/24/2024] [Accepted: 12/11/2024] [Indexed: 01/20/2025]
Abstract
Human epidermal growth factor receptor 2 (HER2, also known as ERBB2) signaling promotes cell growth and differentiation, and is overexpressed in several tumor types, including breast, gastric and colorectal cancer. HER2-targeted therapies have shown clinical activity against these tumor types, resulting in regulatory approvals. However, the efficacy of HER2 therapies in tumors with HER2 mutations has not been widely investigated. SGNTUC-019 is an open-label, phase 2 basket study evaluating tucatinib, a HER2-targeted tyrosine kinase inhibitor, in combination with trastuzumab in patients with HER2-altered solid tumors. The study included a cohort of 31 heavily pretreated female patients with HER2-mutated metastatic breast cancer who were also HER2 negative per local testing. Hormone receptor (HR)-positive patients also received fulvestrant. The overall response rate (primary endpoint) was 41.9% (90% confidence interval (CI): 26.9-58.2). Secondary endpoints of duration of response and progression-free survival were 12.6 months (90% CI: 4.7 to not estimable) and 9.5 months (90% CI: 5.4-13.8), respectively. No new safety signals were detected. Responses were observed across various HER2 mutations, including mutations in the tyrosine kinase and extracellular domains. The chemotherapy-free regimen of tucatinib and trastuzumab showed clinically meaningful antitumor activity with durable responses and favorable tolerability in heavily pretreated patients with HER2 mutations. These data support further investigation of HER2-targeted therapies in this patient population. ClinicalTrials.gov registration: NCT04579380 .
Collapse
Affiliation(s)
| | - Giuseppe Curigliano
- Istituto Europeo di Oncologia, IRCCS, Milan, Italy
- University of Milano, Milan, Italy
| | | | - Do-Youn Oh
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, South Korea
| | - Andree Rorive
- CHU Sart Tilman Liège, University of Liège, Liège, Belgium
| | - Hatem Soliman
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | | | | | - Mark E Burkard
- UW Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Ki Y Chung
- Prisma Health Institute, Greenville, SC, USA
| | - Philip R Debruyne
- Kortrijk Cancer Centre, General Hospital AZ Groeninge, Kortrijk, Belgium
- Medical Technology Research Centre (MTRC), School of Life Sciences, Anglia Ruskin University, Cambridge, UK
- School of Nursing and Midwifery, University of Plymouth, Plymouth, UK
| | - Jenny R Fox
- Rocky Mountain Cancer Center, Boulder, CO, USA
| | | | - Marta Gil-Martin
- Institut Català d'Oncologia L'Hospitalet-IDIBELL, Hospitalet de Llobregat, Spain
| | | | - Bradley J Monk
- Florida Cancer Specialists and Research Institute, West Palm Beach, FL, USA
| | | | - Danny Nguyen
- City of Hope National Medical Center, Duarte, CA, USA
| | - David M O'Malley
- The Ohio State University and James Comprehensive Cancer Center, Columbus, OH, USA
| | | | - Bhavana Pothuri
- Laura & Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Martin Reck
- Department of Thoracic Oncology, Airway Research Center North, Germany Center for Lung Disease, Grosshansdorf, Germany
| | | | - Yu Sunakawa
- St. Marianna University Hospital, Kawasaki, Japan
| | | | - Evan Y Yu
- Fred Hutchinson Cancer Center/University of Washington, Seattle, WA, USA
| | | | | | | | | | - Paula R Pohlmann
- University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
15
|
Haynes J, Manogaran P. Mechanisms and Strategies to Overcome Drug Resistance in Colorectal Cancer. Int J Mol Sci 2025; 26:1988. [PMID: 40076613 PMCID: PMC11901061 DOI: 10.3390/ijms26051988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Colorectal cancer (CRC) is a major cause of cancer-related mortality worldwide, with a significant impact on public health. Current treatment options include surgery, chemotherapy, radiotherapy, molecular-targeted therapy, and immunotherapy. Despite advancements in these therapeutic modalities, resistance remains a significant challenge, often leading to treatment failure, poor progression-free survival, and cancer recurrence. Mechanisms of resistance in CRC are multifaceted, involving genetic mutations, epigenetic alterations, tumor heterogeneity, and the tumor microenvironment. Understanding these mechanisms at the molecular level is crucial for identifying novel therapeutic targets and developing strategies to overcome resistance. This review provides an overview of the diverse mechanisms driving drug resistance in sporadic CRC and discusses strategies currently under investigation to counteract this resistance. Several promising strategies are being explored, including targeting drug transport, key signaling pathways, DNA damage response, cell death pathways, epigenetic modifications, cancer stem cells, and the tumor microenvironment. The integration of emerging therapeutic approaches that target resistance mechanisms aims to enhance the efficacy of current CRC treatments and improve patient outcomes.
Collapse
Affiliation(s)
- Jennifer Haynes
- Department of Clinical and Translational Sciences, Joan C. Edwards School of Medicine, Marshall University, 1600 Medical Center Drive, Huntington, WV 25701, USA;
| | | |
Collapse
|
16
|
Sakai SA, Nomura R, Nagasawa S, Chi S, Suzuki A, Suzuki Y, Imai M, Nakamura Y, Yoshino T, Ishikawa S, Tsuchihara K, Kageyama SI, Yamashita R. SpatialKNifeY (SKNY): Extending from spatial domain to surrounding area to identify microenvironment features with single-cell spatial omics data. PLoS Comput Biol 2025; 21:e1012854. [PMID: 39965034 PMCID: PMC11849985 DOI: 10.1371/journal.pcbi.1012854] [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: 10/07/2024] [Revised: 02/24/2025] [Accepted: 02/03/2025] [Indexed: 02/20/2025] Open
Abstract
Single-cell spatial omics analysis requires consideration of biological functions and mechanisms in a microenvironment. However, microenvironment analysis using bioinformatic methods is limited by the need to detect histological morphology and extend it to the surrounding area. In this study, we developed SpatialKNifeY (SKNY), an image-processing-based toolkit that detects spatial domains that potentially reflect histology and extends these domains to the microenvironment. Using spatial transcriptomic data from breast cancer, we applied the SKNY algorithm to identify tumor spatial domains, followed by clustering of the domains, trajectory estimation, and spatial extension to the tumor microenvironment (TME). The results of the trajectory estimation were consistent with the known mechanisms of cancer progression. We observed tumor vascularization and immunodeficiency at mid- and late-stage progression in TME. Furthermore, we applied the SKNY to integrate and cluster the spatial domains of 14 patients with metastatic colorectal cancer, and the clusters were divided based on the TME characteristics. In conclusion, the SKNY facilitates the determination of the functions and mechanisms in the microenvironment and cataloguing of the features.
Collapse
Affiliation(s)
- Shunsuke A. Sakai
- Division of Translational Informatics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Ryosuke Nomura
- Division of Translational Informatics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Satoi Nagasawa
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
- Department of Breast Surgery, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - SungGi Chi
- Division of Translational Informatics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| | - Ayako Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Mitsuho Imai
- Translational Research Support Office, National Cancer Center Hospital East, Chiba, Japan
- Department of Genetic Medicine and Services, National Cancer Center Hospital East, Chiba, Japan
| | - Yoshiaki Nakamura
- Translational Research Support Office, National Cancer Center Hospital East, Chiba, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Takayuki Yoshino
- Translational Research Support Office, National Cancer Center Hospital East, Chiba, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Shumpei Ishikawa
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Division of Pathology, National Cancer Center Exploratory Oncology Research & Clinical Trial Center, Kashiwa, Chiba, Japan
| | - Katsuya Tsuchihara
- Division of Translational Informatics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Shun-Ichiro Kageyama
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
- Division of Radiation Oncology and Particle Therapy, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| | - Riu Yamashita
- Division of Translational Informatics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| |
Collapse
|
17
|
Strickler JH, Bekaii-Saab T, Cercek A, Heinemann V, Nakamura Y, Raghav K, Siena S, Tabernero J, Van Cutsem E, Yoshino T, Ramos J, Guan X, Andre T. MOUNTAINEER-03 phase III study design: first-line mFOLFOX6 + tucatinib + trastuzumab for HER2+ metastatic colorectal cancer. Future Oncol 2025; 21:303-311. [PMID: 39723627 PMCID: PMC11792820 DOI: 10.1080/14796694.2024.2441101] [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: 06/20/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024] Open
Abstract
Patients diagnosed with metastatic colorectal cancer (mCRC) have a poor prognosis with survival ranging 2-3 years. The prevalence of human epidermal growth factor receptor 2 (HER2) amplification is approximately 3-4% in mCRC and increases up to 8% in patients with KRAS/NRAS/BRAF wild-type (WT) CRC tumors. Tucatinib is a highly selective HER2-directed tyrosine kinase inhibitor that, in combination with trastuzumab, has demonstrated clinically meaningful activity in patients with chemotherapy-refractory, HER2-positive (HER2+), RAS WT mCRC in the MOUNTAINEER trial. The MOUNTAINEER-03 phase III trial is designed to investigate the efficacy and safety of first-line tucatinib in combination with trastuzumab and modified FOLFOX6 (mFOLFOX6) versus standard of care (mFOLFOX6 plus bevacizumab or cetuximab) in patients with untreated HER2+, RAS WT locally advanced unresectable or mCRC. MOUNTAINEER-03 will include two arms of approximately 400 patients randomized 1:1 to either treatment arm. The primary endpoint is progression-free survival per RECIST v1.1 by blinded independent central review (BICR). Key secondary endpoints are overall survival and confirmed objective response rate (according to RECIST v1.1 per BICR). Safety assessments will include surveillance and recording of adverse events, physical examination findings, vital signs, cardiac assessments, Eastern Cooperative Oncology Group performance status, concomitant medications, and laboratory tests.Clinical trial registration: NCT05253651 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- John H Strickler
- Division of Medical Oncology, Duke University Medical Center, Durham, NC, USA
| | - Tanios Bekaii-Saab
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Andrea Cercek
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Volker Heinemann
- Department of Haematology and Oncology, LMU Klinikum, University of Munich, Comprehensive Cancer Center, Munich, Germany
| | - Yoshiaki Nakamura
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital Japan East, Kashiwa, Japan
| | - Kanwal Raghav
- Department of Gastrointestinal Medical Oncology,MD Anderson Cancer Center, Houston, TX,USA
| | - Salvatore Siena
- Department of Hematology and Oncology, Università degli Studi di Milano, and Grande Ospedale Metropolitano Niguard, Milan, Italy
- Niguarda Cancer Center, Università degli Studi di Milano, and Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Josep Tabernero
- Department of Medical Oncology, Vall D’Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain
| | - Eric Van Cutsem
- Department of Digestive Oncology, University Hospital Gasthuisberg and University of Leuven, Leuven, Belgium
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital Japan East, Kashiwa, Japan
| | - Jorge Ramos
- Oncology Research and Development, Pfizer Inc, Bothell, WA, USA
| | - Xuesong Guan
- Oncology Biostatistics, Pfizer Inc, Bothell, WA, USA
| | - Thierry Andre
- Department of Medical Oncology, Sorbonne Université et Hôpital Saint Antoine, Paris, France
| |
Collapse
|
18
|
Ji P, Chen T, Li C, Zhang J, Li X, Zhu H. Comprehensive review of signaling pathways and therapeutic targets in gastrointestinal cancers. Crit Rev Oncol Hematol 2025; 206:104586. [PMID: 39653094 DOI: 10.1016/j.critrevonc.2024.104586] [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: 05/31/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024] Open
Abstract
Targeted therapy, the milestone in the development of human medicine, originated in 2004 when the FDA approved the first targeted agent bevacizumab for colorectal cancer treatment. This new development has resulted from drug developers moving beyond traditional chemotherapy, and several trials have popped up in the last two decades with an unprecedented speed. Specifically, EGF/EGFR, VEGF/VEGFR, HGF/c-MET, and Claudin 18.2 therapeutic targets have been developed in recent years. Some targets previously thought to be undruggable are now being newly explored, such as the RAS site. However, the efficacy of targeted therapy is extremely variable, especially with the emergence of new drugs and the innovative use of traditional targets for other tumors in recent years. Accordingly, this review provides an overview of the major signaling pathway mechanisms and recent advances in targeted therapy for gastrointestinal cancers, as well as future perspectives.
Collapse
Affiliation(s)
- Pengfei Ji
- Department of Thoracic Surgery, West China Hospital, Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China
| | - Tingting Chen
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Chao Li
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Jinyuan Zhang
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Xiao Li
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| |
Collapse
|
19
|
Imai M, Nakamura Y, Shin S, Okamoto W, Kato T, Esaki T, Kato K, Komatsu Y, Yuki S, Masuishi T, Nishina T, Sawada K, Sato A, Kuwata T, Yamashita R, Fujisawa T, Bando H, Ock CY, Fujii S, Yoshino T. Artificial Intelligence-Powered Human Epidermal Growth Factor Receptor 2 and Tumor Microenvironment Analysis in Human Epidermal Growth Factor Receptor 2-Amplified Metastatic Colorectal Cancer: Exploratory Analysis of Phase II TRIUMPH Trial. JCO Precis Oncol 2025; 9:e2400385. [PMID: 39823559 PMCID: PMC11753463 DOI: 10.1200/po-24-00385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/24/2024] [Accepted: 11/27/2024] [Indexed: 01/19/2025] Open
Abstract
PURPOSE Human epidermal growth factor receptor 2 (HER2)-targeted therapies have shown promise in treating HER2-amplified metastatic colorectal cancer (mCRC). Identifying optimal biomarkers for treatment decisions remains challenging. This study explores the potential of artificial intelligence (AI) in predicting treatment responses to trastuzumab plus pertuzumab (TP) in patients with HER2-amplified mCRC from the phase II TRIUMPH trial. MATERIALS AND METHODS AI-powered HER2 quantification continuous score (QCS) and tumor microenvironment (TME) analysis were applied to the prescreening cohort (n = 143) and the TRIUMPH cohort (n = 30). AI analyzers determined the proportions of tumor cells (TCs) with HER2 staining intensity and the densities of various cells in TME, examining their associations with clinical outcomes of TP. RESULTS The AI-powered HER2 QCS for HER2 immunohistochemistry (IHC) achieved an accuracy of 86.7% against pathologist evaluations, with a 100% accuracy for HER2 IHC 3+ patients. Patients with ≥50% of TCs showing HER2 3+ staining intensity (AI-H3-high) exhibited significantly prolonged progression-free survival (PFS; median PFS, 4.4 v 1.4 months; hazard ratio [HR], 0.12 [95% CI, 0.04 to 0.38]) and overall survival (OS; median OS, 16.5 v 4.1 months; HR, 0.13 [95% CI, 0.05 to 0.38]) compared with the AI-H3-low (<50% group). Stratification among patients with AI-H3-high included TME-high (all lymphocyte, fibroblast, and macrophage densities in the cancer stroma above the median) and TME-low (anything below the median), showing a median PFS of 1.3 and 5.6 months for TME-high and TME-low respectively, with an HR of 0.04 (95% CI, 0.01 to 0.19) for AI-H3-high with TME-low compared with AI-H3-low. CONCLUSION AI-powered HER2 QCS and TME analysis demonstrated potential in enhancing treatment response predictions in patients with HER2-amplified mCRC undergoing TP therapy.
Collapse
Affiliation(s)
- Mitsuho Imai
- Translational Research Support Office, National Cancer Center Hospital East, Chiba, Japan
- Department of Genetic Medicine and Services, National Cancer Center Hospital East, Chiba, Japan
| | - Yoshiaki Nakamura
- Translational Research Support Office, National Cancer Center Hospital East, Chiba, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | | | - Wataru Okamoto
- Department of Clinical Oncology, Hiroshima University Hospital, Hiroshima, Japan
| | - Takeshi Kato
- Department of Surgery, NHO Osaka National Hospital, Osaka, Japan
| | - Taito Esaki
- Department of Gastrointestinal and Medical Oncology, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Ken Kato
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yoshito Komatsu
- Department of Cancer Center, Hokkaido University Hospital, Hokkaido, Japan
| | - Satoshi Yuki
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Hokkaido, Japan
| | - Toshiki Masuishi
- Department of Clinical Oncology, Aichi Cancer Center, Nagoya, Japan
| | - Tomohiro Nishina
- Gastrointestinal Medical Oncology, NHO Shikoku Cancer Center, Ehime, Japan
| | - Kentaro Sawada
- Department of Medical Oncology, Kushiro Rosai Hospital, Kushiro, Japan
| | - Akihiro Sato
- Clinical Research Support Office, National Cancer Center Hospital East, Chiba, Japan
| | - Takeshi Kuwata
- Department of Genetic Medicine and Services, National Cancer Center Hospital East, Chiba, Japan
| | - Riu Yamashita
- Division of Translational Informatics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Takao Fujisawa
- Translational Research Support Office, National Cancer Center Hospital East, Chiba, Japan
- Department of Head and Neck Medical Oncology/Translational Research Support Office, National Cancer Center East Hospital, Chiba, Japan
| | - Hideaki Bando
- Translational Research Support Office, National Cancer Center Hospital East, Chiba, Japan
- Department of Genetic Medicine and Services, National Cancer Center Hospital East, Chiba, Japan
| | | | - Satoshi Fujii
- Department of Molecular Pathology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takayuki Yoshino
- Translational Research Support Office, National Cancer Center Hospital East, Chiba, Japan
- Department for the Promotion of Drug and Diagnostic Development, National Cancer Center Hospital East, Chiba, Japan
| |
Collapse
|
20
|
Nakamura Y, Ozaki H, Ueno M, Komatsu Y, Yuki S, Esaki T, Taniguchi H, Sunakawa Y, Yamaguchi K, Kato K, Denda T, Nishina T, Takahashi N, Satoh T, Yasui H, Satake H, Oki E, Kato T, Ohta T, Matsuhashi N, Goto M, Okano N, Ohtsubo K, Yamazaki K, Yamashita R, Iida N, Yuasa M, Bando H, Yoshino T. Targeted therapy guided by circulating tumor DNA analysis in advanced gastrointestinal tumors. Nat Med 2025; 31:165-175. [PMID: 39284955 PMCID: PMC11750700 DOI: 10.1038/s41591-024-03244-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 08/09/2024] [Indexed: 01/23/2025]
Abstract
Although comprehensive genomic profiling has become standard in oncology for advanced solid tumors, the full potential of circulating tumor DNA (ctDNA)-based profiling in capturing tumor heterogeneity and guiding therapy selection remains underexploited, marked by a scarcity of evidence on its clinical impact and the assessment of intratumoral heterogeneity. The GOZILA study, a nationwide, prospective observational ctDNA profiling study, previously demonstrated higher clinical trial enrollment rates using liquid biopsy compared with tissue screening. This updated analysis of 4,037 patients further delineates the clinical utility of ctDNA profiling in advanced solid tumors, showcasing a significant enhancement in patient outcomes with a 24% match rate for targeted therapy. Patients treated with matched targeted therapy based on ctDNA profiling demonstrated significantly improved overall survival compared with those receiving unmatched therapy (hazard ratio, 0.54). Notably, biomarker clonality and adjusted plasma copy number were identified as predictors of therapeutic efficacy, reinforcing the value of ctDNA in reflecting tumor heterogeneity for precise treatment decisions. These new insights into the relationship between ctDNA characteristics and treatment outcomes advance our understanding beyond the initial enrollment benefits. Our findings advocate for the broader adoption of ctDNA-guided treatment, signifying an advancement in precision oncology and improving survival outcomes in advanced solid tumors.
Collapse
Affiliation(s)
- Yoshiaki Nakamura
- International Research Promotion Office, National Cancer Center Hospital East, Kashiwa, Japan
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hiroshi Ozaki
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Yoshito Komatsu
- Department of Cancer Center, Hokkaido University Hospital, Sapporo, Japan
| | - Satoshi Yuki
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Sapporo, Japan
| | - Taito Esaki
- Department of Gastrointestinal and Medical Oncology, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Hiroya Taniguchi
- Department of Clinical Oncology, Aichi Cancer Center, Nagoya, Japan
| | - Yu Sunakawa
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Kensei Yamaguchi
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ken Kato
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Tadamichi Denda
- Division of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Tomohiro Nishina
- Gastrointestinal Medical Oncology, NHO Shikoku Cancer Center, Matsuyama, Japan
| | - Naoki Takahashi
- Department of Gastroenterology, Saitama Cancer Center, Ina, Japan
| | - Taroh Satoh
- Center for Cancer Genomics and Precision Medicine, Osaka University Hospital, Suita, Japan
| | - Hisateru Yasui
- Department of Medical Oncology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Hironaga Satake
- Cancer Treatment Center, Kansai Medical University Hospital, Hirakata, Japan
- Department of Medical Oncology, Kochi Medical School, Nankoku, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeshi Kato
- Department of Surgery, NHO Osaka National Hospital, Osaka, Japan
| | - Takashi Ohta
- Department of Clinical Oncology, Kansai Rosai Hospital, Amagasaki, Japan
| | - Nobuhisa Matsuhashi
- Department of Gastroenterological Surgery and Pediatric Surgery, Center for One Medicine Innovative Translational Research (COMIT), Gifu University Graduate School of Medicine, Gifu, Japan
| | - Masahiro Goto
- Cancer Chemotherapy Center, Osaka Medical and Pharmaceutical University Hospital, Takatsuki, Japan
| | - Naohiro Okano
- Department of Medical Oncology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Koushiro Ohtsubo
- Department of Medical Oncology, Kanazawa University Hospital, Kanazawa, Japan
| | - Kentaro Yamazaki
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Nagaizumi, Japan
| | - Riu Yamashita
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Naoko Iida
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Mihoko Yuasa
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hideaki Bando
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takayuki Yoshino
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
- Kindai University Faculty of Medicine, Osakasayama, Japan.
| |
Collapse
|
21
|
Ohmura H, Hanamura F, Okumura Y, Ando Y, Masuda T, Mimori K, Akashi K, Baba E. Liquid biopsy for breast cancer and other solid tumors: a review of recent advances. Breast Cancer 2025; 32:33-42. [PMID: 38492205 DOI: 10.1007/s12282-024-01556-8] [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: 10/26/2023] [Accepted: 02/13/2024] [Indexed: 03/18/2024]
Abstract
Liquid biopsy using circulating tumor DNA (ctDNA) has been reported to be less invasive and effective for comprehensive genetic analysis of heterogeneous solid tumors, including decision-making for therapeutic strategies, predicting recurrence, and detecting genetic factors related to treatment resistance in various types of cancers. Breast cancer, colorectal cancer, and lung cancer are among the most prevalent malignancies worldwide, and clinical studies of liquid biopsy for these cancers are ongoing. Liquid biopsy has been used as a companion diagnostic tool in clinical settings, and research findings have accumulated, especially in cases of colorectal cancer after curative resection and non-small cell lung cancer (NSCLC) after curative chemoradiotherapy, in which ctDNA detection helps predict eligibility for adjuvant chemotherapy. Liquid biopsy using ctDNA shows promise across a wide range of cancer types, including breast cancer, and its clinical applications are expected to expand further through ongoing research. In this article, studies on liquid biopsy in breast cancer, colorectal cancer, and NSCLC are compared focusing on ctDNA.
Collapse
Affiliation(s)
- Hirofumi Ohmura
- Department of Oncology and Social Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
- Department of Internal Medicine, Kyushu University Beppu Hospital, Oita, Japan
| | - Fumiyasu Hanamura
- Department of Internal Medicine, Kyushu University Beppu Hospital, Oita, Japan
| | - Yuta Okumura
- Department of Internal Medicine, Kyushu University Beppu Hospital, Oita, Japan
- Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Yuki Ando
- Department of Surgery, Kyushu University Beppu Hospital, Oita, Japan
| | - Takaaki Masuda
- Department of Surgery, Kyushu University Beppu Hospital, Oita, Japan
| | - Koshi Mimori
- Department of Surgery, Kyushu University Beppu Hospital, Oita, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eishi Baba
- Department of Oncology and Social Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
22
|
Kou M, Deng Y. Circulating tumor DNA as a predictive biomarker for treatment response and survival in metastatic colorectal cancer. Int J Colorectal Dis 2024; 39:203. [PMID: 39681775 DOI: 10.1007/s00384-024-04785-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/09/2024] [Indexed: 12/18/2024]
Abstract
PURPOSE To explore the potential of circulating tumor DNA (ctDNA) as a prognostic biomarker to predict treatment response and survival outcomes in patients with metastatic colorectal cancer (mCRC). METHODS A retrospective analysis was conducted on 134 patients with mCRC who were treated between January 2020 and December 2021. The patients were classified into ctDNA-negative and ctDNA-positive groups based on plasma ctDNA detection. Demographic, clinical, and laboratory parameters, treatment response, survival outcomes, and adverse events were recorded and analyzed. RESULTS No significant differences were observed in baseline characteristics between the two groups. Compared to the ctDNA-positive patients, ctDNA-negative patients exhibited superior outcomes, including a higher objective response rate (65.22% vs. 46.15%), disease control rate (81.16% vs. 63.08%), progression-free survival (8.24 ± 1.02 vs. 7.86 ± 0.91 months), overall survival (24.58 ± 3.58 vs. 23.27 ± 3.46 months), and 1-year survival rate (73.91% vs. 55.38%). The ctDNA-positive group had a significantly higher incidence of adverse events. Correlation analyses revealed significant associations between ctDNA status, tumor markers, treatment response, and survival outcomes. CONCLUSIONS ctDNA is a promising noninvasive biomarker for predicting treatment response, survival, and adverse events in mCRC, potentially guiding personalized therapeutic strategies.
Collapse
Affiliation(s)
- Mengying Kou
- Department of Oncology, Maoming People's Hospital, Maoming City, Guangdong Province, China
| | - Ying Deng
- Department of Gastroenterology, Maoming People's Hospital, No.101 Weimin Road, Maonan District, Maoming City, Guangdong Province, 525000, China.
| |
Collapse
|
23
|
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: 6] [Impact Index Per Article: 6.0] [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
|
24
|
Ikushima H, Watanabe K, Shinozaki-Ushiku A, Oda K, Kage H. A machine learning-based analysis of nationwide cancer comprehensive genomic profiling data across cancer types to identify features associated with recommendation of genome-matched therapy. ESMO Open 2024; 9:103998. [PMID: 39591805 PMCID: PMC11629217 DOI: 10.1016/j.esmoop.2024.103998] [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: 07/05/2024] [Revised: 10/24/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND The low probability of identifying druggable mutations through comprehensive genomic profiling (CGP) and its financial and time costs hinder its widespread adoption. To enhance the effectiveness and efficiency of cancer precision medicine, it is critical to identify patient characteristics that are most likely to benefit from CGP. PATIENTS AND METHODS This nationwide retrospective study employed machine learning models to predict the identification of genome-matched therapies by CGP, utilizing a national database covering 99.7% of the patients who underwent CGP in Japan from June 2019 to November 2023. Prediction models were constructed for the overall cancer population, specific cancer types, and adolescent and young adult (AYA) group. The SHapley Additive exPlanations (SHAP) algorithm was applied to elucidate clinical features contributing to model predictions. RESULTS This study included 60 655 patients [mean age (standard deviation), 60.8 years (14.5 years); 50.1% males]. CGP identified at least one genome-matched therapy in 11 227 cases (18.5%). The best prediction model was eXtreme Gradient Boosting (XGBoost) with an area under the receiver operating characteristic curve of 0.819. Cancer type was the most important predictor (negative for pancreas and positive for breast and lung), followed by the age, presence of liver metastasis, and number of metastatic sites. Analysis of cancer type-specific models identified several organ-specific features, including the sex, interval between the cancer diagnosis and CGP, sampling site, and CGP panel. Among 3455 AYA patients, genome-matched therapies were identified in 459 patients (13.3%). The AYA-specific model achieved an area under the receiver operating characteristic curve of 0.768, with bone tumor identified as a negative predictor in addition to those identified in the overall cancer population model. CONCLUSION Several factors predicting the identification of genome-matched therapies through CGP were identified for the overall cancer population and cancer type-specific subpopulations. Expedited CGP is recommended for patients who match the identified profile to facilitate early targeted therapy.
Collapse
Affiliation(s)
- H Ikushima
- Department of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - K Watanabe
- Department of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Next-Generation Precision Medicine Development Laboratory, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - A Shinozaki-Ushiku
- Division of Integrative Genomics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - K Oda
- Division of Integrative Genomics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - H Kage
- Department of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
25
|
Venturini J, Massaro G, Lavacchi D, Rossini D, Pillozzi S, Caliman E, Pellegrini E, Antonuzzo L. The emerging HER2 landscape in colorectal cancer: the key to unveil the future treatment algorithm? Crit Rev Oncol Hematol 2024; 204:104515. [PMID: 39304034 DOI: 10.1016/j.critrevonc.2024.104515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 09/03/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024] Open
Abstract
Colorectal cancer (CRC) represents a global health threat, standing as the second leading cause of cancer-related death worldwide. Targeted therapies brought new hope for the metastatic stage, which historically bore a very poor prognosis. Human epidermal growth receptor 2 (HER2) overexpression concerns about 5 % of the metastatic CRC (mCRC) patients, including both gene amplifications and point mutations. Albeit its controversial prognostic role, preclinical and clinical data indicate HER2 as a negative predictive biomarker of response to anti-EGFR therapies. Tissue and plasma-based NGS testing, could permit a precise identification of this resistance mechanism both at baseline and during treatment, thus guiding decision-making. Furthermore, promising results come from completed and ongoing randomized trials, testing HER2 as an actionable target. In this review, we discuss the available evidence on HER2 targeting in advanced CRC, analyzing its possible future role in the treatment algorithm.
Collapse
Affiliation(s)
- Jacopo Venturini
- Clinical Oncology Unit, Careggi University Hospital, Florence 50134, Italy
| | - Giulia Massaro
- Clinical Oncology Unit, Careggi University Hospital, Florence 50134, Italy
| | - Daniele Lavacchi
- Clinical Oncology Unit, Careggi University Hospital, Florence 50134, Italy
| | - Daniele Rossini
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy; Department of Health Science, University of Florence, Florence 50139, Italy
| | - Serena Pillozzi
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy; Medical Oncology Unit, Careggi University Hospital, Florence 50134, Italy
| | - Enrico Caliman
- Clinical Oncology Unit, Careggi University Hospital, Florence 50134, Italy
| | - Elisa Pellegrini
- Medical Oncology Unit, Careggi University Hospital, Florence 50134, Italy
| | - Lorenzo Antonuzzo
- Clinical Oncology Unit, Careggi University Hospital, Florence 50134, Italy; Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy; Medical Oncology Unit, Careggi University Hospital, Florence 50134, Italy.
| |
Collapse
|
26
|
Yagisawa M, Taniguchi H, Satoh T, Kadowaki S, Sunakawa Y, Nishina T, Komatsu Y, Esaki T, Sakai D, Doi A, Kajiwara T, Ono H, Asano M, Hirano N, Odegaard J, Fujii S, Nomura S, Bando H, Sato A, Yoshino T, Nakamura Y. Trastuzumab Deruxtecan in Advanced Solid Tumors With Human Epidermal Growth Factor Receptor 2 Amplification Identified by Plasma Cell-Free DNA Testing: A Multicenter, Single-Arm, Phase II Basket Trial. J Clin Oncol 2024; 42:3817-3825. [PMID: 39088783 PMCID: PMC11542975 DOI: 10.1200/jco.23.02626] [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: 12/05/2023] [Revised: 05/01/2024] [Accepted: 05/10/2024] [Indexed: 08/03/2024] Open
Abstract
PURPOSE HERALD/EPOC1806 was conducted as a multicenter phase II trial assessing trastuzumab deruxtecan (T-DXd) therapy for patients with human epidermal growth factor receptor 2 (HER2)-amplified progressive stage solid tumors detected by cell-free DNA (cfDNA) testing. PATIENTS AND METHODS Patients exhibited advanced solid tumors with HER2 amplification that was identified via next-generation sequencing of cfDNA testing, without the requirement for immunohistochemical HER2 testing. The studied group was administered T-DXd at 5.4 mg/kg once every 3 weeks until onset of disease progression or intolerable toxicity. RESULTS Overall, 4,734 patients underwent cfDNA testing from December 2019 to January 2022, and 252 demonstrated HER2 amplification. Finally, the study included 62 patients with 16 cancer types with a median baseline plasma HER2 copy number (CN) of 8.55 (range, 2.4-73.9). Confirmed overall response rate (ORR) by investigator assessment was 56.5% (95% CI, 43.3 to 69.0), thus showing a value beyond the 5% threshold. Responses were evaluated for 13 cancer types, including KRAS-mutant colorectal (1/3), PIK3CA-mutant endometrial (5/6), and tissue HER2-negative gastric (1/2) cancers. Plasma HER2 CN above versus below the baseline median value did not differ for impact response; however, clearance of HER2 amplification in cfDNA on cycle 2 day 1 had higher response values compared with persistence. Median progression-free survival and response duration were 7.0 (95% CI, 4.9 to 9.7) and 8.8 (95% CI, 5.8 to 11.2) months, respectively, with the majority of complications being mild to moderate. Interstitial lung diseases were identified in 16 (26%) patients, including 14 patients with grade 1 disease, one patient with grade 2 disease, and one patient with grade 3 disease. CONCLUSION T-DXd treatment demonstrated high ORR with durable response in patients with advanced HER2-amplified solid tumors determined with cfDNA testing.
Collapse
Affiliation(s)
- Masataka Yagisawa
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology, Japan Community Health Care Organization Sapporo Hokushin Hospital, Sapporo, Japan
| | - Hiroya Taniguchi
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Taroh Satoh
- Center for Cancer Genomics and Precision Medicine, Osaka University Hospital, Osaka, Japan
| | - Shigenori Kadowaki
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Yu Sunakawa
- Department of Clinical Oncology, St Marianna University School of Medicine, Kawasaki, Japan
| | - Tomohiro Nishina
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Yoshito Komatsu
- Division of Cancer Center, Hokkaido University Hospital, Sapporo, Japan
| | - Taito Esaki
- Gastrointestinal and Medical Oncology Division, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Daisuke Sakai
- Center for Cancer Genomics and Precision Medicine, Osaka University Hospital, Osaka, Japan
| | - Ayako Doi
- Department of Clinical Oncology, St Marianna University School of Medicine, Kawasaki, Japan
| | - Takeshi Kajiwara
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Hiromi Ono
- Clinical Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masatoshi Asano
- Clinical Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Nami Hirano
- Clinical Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | | | - Satoshi Fujii
- Department of Molecular Pathology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shogo Nomura
- Department of Biostatistics and Bioinformatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hideaki Bando
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Translational Research Support Section, National Cancer Center Hospital East, Kashiwa, Japan
| | - Akihiro Sato
- Clinical Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshiaki Nakamura
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Translational Research Support Section, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
27
|
Yamaguchi R, Uchiyama M, Miyazaki M, Hayashi T, Oyabu K, Nakano T, Matsuo K. Factors Associated With Infusion Reactions in Patients With Breast Cancer Receiving Trastuzumab. CANCER DIAGNOSIS & PROGNOSIS 2024; 4:722-728. [PMID: 39502602 PMCID: PMC11534052 DOI: 10.21873/cdp.10387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 11/08/2024]
Abstract
Background/Aim Trastuzumab (TRA) is a key drug in human epidermal growth factor receptor type 2 (HER2)-positive breast cancer treatment. Infusion reactions (IR) with TRA are frequently observed in practice. Although the efficacy of premedication has been previously reported, it remains uncommon. The probability of severe IR due to TRA is low; however, when it does occur, it is associated with patient discomfort and expenditure of medical resources. This study aimed to analyze the factors associated with the occurrence of IR in patients with breast cancer who received TRA. Patients and Methods We retrospectively studied 204 patients who underwent TRA for breast cancer treatment between September 2008 and June 2023, identifying factors influencing the occurrence of IR at the time of TRA administration. Results A total of 182 patients were included in this study, and the incidence of IR was 25.8% (47/182 patients). Multiple logistic regression analysis showed that pertuzumab (PER) use, high alkaline phosphatase (ALP), and low high-density lipoprotein (HDL) cholesterol levels were associated with IR. Conclusion IR should be considered when PER is combined with TRA. ALP and HDL cholesterol levels may be predictive markers of TRA-induced IR in patients with breast cancer.
Collapse
Affiliation(s)
- Ryuji Yamaguchi
- Department of Pharmacy, Kyushu Central Hospital, Fukuoka, Japan
| | - Masanobu Uchiyama
- Department of Pharmacy, Fukuoka University Hospital, Fukuoka, Japan
- Department of Oncology and Infectious Disease Pharmacy, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Motoyasu Miyazaki
- Department of Pharmacy, Fukuoka University Chikushi Hospital, Fukuoka, Japan
- Department of Emergency and Disaster Medical Pharmacy, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Toshinobu Hayashi
- Department of Emergency and Disaster Medical Pharmacy, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Kohei Oyabu
- Department of Drug Informatics and Translational Research, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Takafumi Nakano
- Department of Pharmacy, Fukuoka University Hospital, Fukuoka, Japan
- Department of Oncology and Infectious Disease Pharmacy, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Koichi Matsuo
- Department of Pharmacy, Fukuoka University Hospital, Fukuoka, Japan
- Department of Oncology and Infectious Disease Pharmacy, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
28
|
Smabers LP, Huismans MA, van Nieuwenhuijzen N, Minnema MC, Kranenburg O, Koopman M, Snippert HJG, May AM, Roodhart JML. Efficacy and safety in early-phase clinical trials for refractory colorectal cancer: A meta-analysis. Eur J Cancer 2024; 212:115059. [PMID: 39368225 DOI: 10.1016/j.ejca.2024.115059] [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: 06/05/2024] [Revised: 09/12/2024] [Accepted: 09/22/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND Despite recent metastatic colorectal cancer (mCRC) therapeutic innovations a comprehensive synthesis of patient outcome and risk-benefit assessment of phase 1/2 trials is missing. The aim of this meta-analysis is to assess efficacy, safety, and trends over time for phase 1 and 2 mCRC trials by examining clinical benefit rate (CBR), overall response rate (ORR), grade 3 or higher adverse events (AE), and discontinuation due to AE. METHODS The PRISMA guidelines were followed. We searched PubMed and Embase for publications of phase 1/2 trials between 2010-2021. Trials reporting on new therapies for treatment-refractory mCRC were included. RESULTS The search strategy yielded 4175 unique reports, of which 258 publications were eligible. These publications report data of 277 unique treatment arms. Overall ORR was 6 %, CBR was 27 % in phase 1 and 36 % in phase 2 trials. CBR increased from 23 % in 2010-2012 to 42 % in 2019-2021. Compared to 2010-2012, trials in 2019-2021 more often tested immunomodulators (4 % vs 23 %), included molecularly preselected populations (4 % vs 38 %) and younger patients (median age<60 44 % vs 66 %). Grade 3 + AE occurred in 35 % of patients, most frequently in trials investigating targeted treatments. CONCLUSIONS Treatment efficacy in phase 1/2 trials is modest but improved from 2010 to 2021. This improvement is accompanied by a shift towards testing in a younger, fitter, and more strictly molecularly preselected population, as well as an increased focus on targeted and immunotherapies.
Collapse
Affiliation(s)
- Lidwien P Smabers
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Laboratory of Translational Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Maarten A Huismans
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Niels van Nieuwenhuijzen
- Department of Hematology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Monique C Minnema
- Department of Hematology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Onno Kranenburg
- Laboratory of Translational Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Miriam Koopman
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Hugo J G Snippert
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Anne M May
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, Utrecht University, Utrecht, the Netherlands
| | - Jeanine M L Roodhart
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
29
|
Hashimoto T, Nakamura Y, Fujisawa T, Imai M, Shibuki T, Iida N, Ozaki H, Nonomura N, Morizane C, Iwata H, Okano S, Yamagami W, Yamazaki N, Kadowaki S, Taniguchi H, Ueno M, Boku S, Oki E, Komatsu Y, Yuki S, Makiyama A, Otsuka T, Hara H, Okano N, Nishina T, Sakamoto Y, Miki I, Kobayashi S, Yuda J, Kageyama SI, Nagamine M, Sakashita S, Sakamoto N, Yamashita R, Koga Y, Bando H, Ishii G, Kuwata T, Park WY, Ohtsu A, Yoshino T. The SCRUM-MONSTAR Cancer-Omics Ecosystem: Striving for a Quantum Leap in Precision Medicine. Cancer Discov 2024; 14:2243-2261. [PMID: 39023403 PMCID: PMC11528206 DOI: 10.1158/2159-8290.cd-24-0206] [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: 02/12/2024] [Revised: 04/23/2024] [Accepted: 06/22/2024] [Indexed: 07/20/2024]
Abstract
The SCRUM-Japan MONSTAR-SCREEN consortium is a nationwide molecular profiling project employing artificial intelligence-driven multiomics analyses for patients with advanced malignancies, aiming to develop novel therapeutics and diagnostics and deliver effective drugs to patients. Concurrently, studies assessing molecular residual disease-based precision medicine for resectable solid tumors, including CIRCULATE-Japan, are ongoing. The substantial data generated by these platforms are stored within a state-of-the-art supercomputing infrastructure, VAPOR CONE. Since 2015, our project has registered over 24,000 patients as of December 2023. Among 16,144 patients with advanced solid tumors enrolled in MONSTAR-SCREEN projects, 5.0% have participated in matched clinical trials, demonstrating a 29.2% objective response rate and 14.8-month median survival (95% CI, 13.4-16.3) for patients treated in the matched clinical trials. Notably, patients who received matched therapy demonstrated significantly prolonged overall survival compared with those who did not (hazard ratio 0.77; 95% confidence interval, 0.71-0.83). Significance: Our nationwide molecular profiling initiative played pivotal roles in facilitating the enrollment of patients with advanced solid tumors into matched clinical trials and highlighted the substantial survival benefits of patients treated with matched therapy. We aim to facilitate an industry-academia data-sharing infrastructure ecosystem, fostering new drug discovery paradigms and precision medicine.
Collapse
Affiliation(s)
- Tadayoshi Hashimoto
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshiaki Nakamura
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takao Fujisawa
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Head and Neck Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Mitsuho Imai
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Taro Shibuki
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Naoko Iida
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hiroshi Ozaki
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Chigusa Morizane
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hiroji Iwata
- Department of Breast Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Susumu Okano
- Department of Head and Neck Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Wataru Yamagami
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Naoya Yamazaki
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Shigenori Kadowaki
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Hiroya Taniguchi
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Shogen Boku
- Cancer Treatment Center, Kansai Medical University Hospital, Osaka, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshito Komatsu
- Department of Cancer Center, Hokkaido University Hospital, Sapporo, Japan
| | - Satoshi Yuki
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Sapporo, Japan
| | - Akitaka Makiyama
- Cancer Center, Gifu University Hospital, Gifu, Japan
- Center for One Medicine Innovative Translational Research, Gifu University, Gifu, Japan
| | - Tomoyuki Otsuka
- Department of Medical Oncology, Osaka International Cancer Institute Osaka Prefectural Hospital Organization, Osaka, Japan
| | - Hiroki Hara
- Department of Gastroenterology, Saitama Cancer Center, Saitama, Japan
| | - Naohiro Okano
- Department of Medical Oncology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Tomohiro Nishina
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Yasutoshi Sakamoto
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Izumi Miki
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shin Kobayashi
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Junichiro Yuda
- Department of Hematology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shun-Ichiro Kageyama
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Michiko Nagamine
- TR Sample Management Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shingo Sakashita
- TR Sample Management Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Naoya Sakamoto
- TR Sample Management Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Riu Yamashita
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshikatsu Koga
- Division of Developmental Therapeutics, Research Center for Innovative Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hideaki Bando
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Genichiro Ishii
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takeshi Kuwata
- Department of Genetic Medicine and Services, National Cancer Center Hospital East, Kashiwa, Japan
| | - Woong-Yang Park
- GxD Inc., Kashiwa, Japan
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea
| | - Atsushi Ohtsu
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
30
|
Hu Q, Chen L, Li K, Liu R, Sun L, Han T. Circulating tumor DNA: current implementation issues and future challenges for clinical utility. Clin Chem Lab Med 2024; 62:2094-2110. [PMID: 38109307 DOI: 10.1515/cclm-2023-1157] [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: 10/16/2023] [Accepted: 12/06/2023] [Indexed: 12/20/2023]
Abstract
Over the past decades, liquid biopsy, especially circulating tumor DNA (ctDNA), has received tremendous attention as a noninvasive detection approach for clinical applications, including early diagnosis of cancer and relapse, real-time therapeutic efficacy monitoring, potential target selection and investigation of drug resistance mechanisms. In recent years, the application of next-generation sequencing technology combined with AI technology has significantly improved the accuracy and sensitivity of liquid biopsy, enhancing its potential in solid tumors. However, the increasing integration of such promising tests to improve therapy decision making by oncologists still has complexities and challenges. Here, we propose a conceptual framework of ctDNA technologies and clinical utilities based on bibliometrics and highlight current challenges and future directions, especially in clinical applications such as early detection, minimal residual disease detection, targeted therapy, and immunotherapy. We also discuss the necessities of developing a dynamic field of translational cancer research and rigorous clinical studies that may support therapeutic strategy decision making in the near future.
Collapse
Affiliation(s)
- Qilin Hu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, P.R. China
| | - Lujun Chen
- The General Hospital of Northern Theater Command Training Base for Graduate, China Medical University, Shenyang, P.R. China
| | - Kerui Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, P.R. China
| | - Ruotong Liu
- Clinical Medicine, Shenyang Medical College, Shenyang, P.R. China
| | - Lei Sun
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, P.R. China
| | - Tao Han
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, P.R. China
| |
Collapse
|
31
|
Zhang R, Su C, Jia Y, Xing M, Jin S, Zong H. Molecular mechanisms of HER2-targeted therapy and strategies to overcome the drug resistance in colorectal cancer. Biomed Pharmacother 2024; 179:117363. [PMID: 39236476 DOI: 10.1016/j.biopha.2024.117363] [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: 06/11/2024] [Revised: 08/15/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024] Open
Abstract
HER2 amplification is one of the mechanisms that induce drug resistance to anti-EGFR therapy in colorectal cancer. In recent years, data from several randomized clinical trials show that anti-HER2 therapies improved the prognosis of patients with HER2-positive colorectal cancer. These results indicate that HER2 is a promising therapeutic target in advanced colorectal cancer. Despite the anti-HER2 therapies including monoclonal antibodies, tyrosine kinase inhibitors, and antibody-drug conjugates improving the outcomes, less than 30 % of the patients achieve objective response and eventually have drug resistance. It is necessary to explore the primary and secondary mechanisms for the resistance to anti-HER2 therapies, which will pave the way to overcome the drug resistance. Several studies have reported the potential mechanisms for the resistance to anti-HER2 therapies. In this review, we present a comprehensive overview of the recent advances in clinical research, mechanisms of treatment resistance, and strategies for reversing resistance in HER2-positive colorectal cancer patients.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Chang Su
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Yongliang Jia
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, China.
| | - Menglu Xing
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Shuiling Jin
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Hong Zong
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
32
|
Casas-Arozamena C, Vilar A, Cueva J, Arias E, Sampayo V, Diaz E, Oltra SS, Moiola CP, Cabrera S, Cortegoso A, Curiel T, Abalo A, Pamies Serrano M, Domingo S, Padilla-Iserte P, Arnaez de la Cruz M, Hernández A, García-Pineda V, Ruiz-Bañobre J, López R, Matias-Guiu X, Colás E, Gil-Moreno A, Abal M, Moreno-Bueno G, Muinelo-Romay L. Role of cfDNA and ctDNA to improve the risk stratification and the disease follow-up in patients with endometrial cancer: towards the clinical application. J Exp Clin Cancer Res 2024; 43:264. [PMID: 39304963 DOI: 10.1186/s13046-024-03158-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 08/08/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND There has been a rise in endometrial cancer (EC) incidence leading to increased mortality. To counter this trend, improving the stratification of post-surgery recurrence risk and anticipating disease relapse and treatment resistance is essential. Liquid biopsy analyses offer a promising tool for these clinical challenges, though the best strategy for applying them in EC must be defined. This study was designed to determine the value of cfDNA/ctDNA monitoring in improving the clinical management of patients with localized and recurrent disease. METHODS Plasma samples and uterine aspirates (UA) from 198 EC patients were collected at surgery and over time. The genetic landscape of UAs was characterized using targeted sequencing. Total cfDNA was analyzed for ctDNA presence based on the UA mutational profile. RESULTS High cfDNA levels and detectable ctDNA at baseline correlated with poor prognosis for DFS (p-value < 0.0001; HR = 9.25) and DSS (p-value < 0.0001; HR = 11.20). This remained clinically significant when stratifying tumors by histopathological risk factors. Of note, cfDNA/ctDNA analyses discriminated patients with early post-surgery relapse and the ctDNA kinetics served to identify patients undergoing relapse before any clinical evidence emerged. CONCLUSIONS This is the most comprehensive study on cfDNA/ctDNA characterization in EC, demonstrating its value in improving risk stratification and anticipating disease relapse in patients with localized disease. CtDNA kinetics assessment complements current strategies to monitor the disease evolution and the treatment response. Therefore, implementing cfDNA/ctDNA monitoring in clinical routines offers a unique opportunity to improve EC management. TRANSLATIONAL RELEVANCE The study demonstrates that high levels of cfDNA and detectable ctDNA at baseline are strong indicators of poor prognosis. This enables more accurate risk stratification beyond traditional histopathological factors, allowing clinicians to identify high-risk patients who may benefit from more aggressive treatment and closer monitoring. Moreover, longitudinal analysis of cfDNA/ctDNA can detect disease recurrence months before clinical symptoms or imaging evidence appear. This early warning system offers a significant advantage in clinical practice, providing a window of opportunity for early intervention and potentially improving patient outcomes.
Collapse
Affiliation(s)
- Carlos Casas-Arozamena
- Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela, 15706, Spain
- University of Santiago de Compostela (USC), Praza do Obradoiro, 0, Santiago de Compostela, 15705, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ana Vilar
- Department of Gynecology, University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela, 15706, Spain
| | - Juan Cueva
- Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela, 15706, Spain
- Department of Medical Oncology, University Clinical Hospital of Santiago de Compostela, University of Santiago de Compostela (USC), Santiago de Compostela, 15706, Spain
| | - Efigenia Arias
- Department of Gynecology, University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela, 15706, Spain
| | - Victoria Sampayo
- Department of Gynecology, University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela, 15706, Spain
| | - Eva Diaz
- MD Anderson Cancer Center Foundation, C/Gómez Hemans 2, Madrid, 28033, Spain
| | - Sara S Oltra
- MD Anderson Cancer Center Foundation, C/Gómez Hemans 2, Madrid, 28033, Spain
| | - Cristian Pablo Moiola
- Department of Gynecologic Oncology, Biomedical Research Group in Gynecology, Vall Hebron Research Institute (VHIR), Universitat Autónoma de Barcelona, 119-129 Pg. Vall d'Hebron, Barcelona, 08035, Spain
- Department of Basic Sciences, Faculty of Health Sciences at Manresa, University of Vic - Central University of Catalonia (UVic-UCC), Av. Universitària, 4-6, Manresa, 08242, Spain
| | - Silvia Cabrera
- Department of Gynecologic Oncology, Biomedical Research Group in Gynecology, Vall Hebron Research Institute (VHIR), Universitat Autónoma de Barcelona, 119-129 Pg. Vall d'Hebron, Barcelona, 08035, Spain
| | - Alexandra Cortegoso
- Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela, 15706, Spain
- Department of Medical Oncology, University Clinical Hospital of Santiago de Compostela, University of Santiago de Compostela (USC), Santiago de Compostela, 15706, Spain
| | - Teresa Curiel
- Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela, 15706, Spain
- Department of Medical Oncology, University Clinical Hospital of Santiago de Compostela, University of Santiago de Compostela (USC), Santiago de Compostela, 15706, Spain
| | - Alicia Abalo
- Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela, 15706, Spain
| | - Mónica Pamies Serrano
- Department of Gynecologic Oncology, Biomedical Research Group in Gynecology, Vall Hebron Research Institute (VHIR), Universitat Autónoma de Barcelona, 119-129 Pg. Vall d'Hebron, Barcelona, 08035, Spain
| | - Santiago Domingo
- Department of Gynecologic Oncology, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - Pablo Padilla-Iserte
- Department of Gynecologic Oncology, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - Marta Arnaez de la Cruz
- Department of Gynecologic Oncology, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - Alicia Hernández
- Department of Gynecology, University Hospital "La Paz", Madrid, Spain
| | | | - Juan Ruiz-Bañobre
- Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela, 15706, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Department of Medical Oncology, University Clinical Hospital of Santiago de Compostela, University of Santiago de Compostela (USC), Santiago de Compostela, 15706, Spain
| | - Rafael López
- Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela, 15706, Spain
- University of Santiago de Compostela (USC), Praza do Obradoiro, 0, Santiago de Compostela, 15705, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Department of Medical Oncology, University Clinical Hospital of Santiago de Compostela, University of Santiago de Compostela (USC), Santiago de Compostela, 15706, Spain
| | - Xavier Matias-Guiu
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Department of Pathology, Instituto de Investigación Biomédica de Bellvitge, Centro de Investigación Biomédica en Red de Cáncer, Hospital U de Bellvitge and Hospital U Arnau de Vilanova, Universities of Lleida and Barcelona, Institut de Recerca Biomèdica de Lleida, Barcelona, Spain
| | - Eva Colás
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Department of Gynecologic Oncology, Biomedical Research Group in Gynecology, Vall Hebron Research Institute (VHIR), Universitat Autónoma de Barcelona, 119-129 Pg. Vall d'Hebron, Barcelona, 08035, Spain
| | - Antonio Gil-Moreno
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Department of Gynecologic Oncology, Biomedical Research Group in Gynecology, Vall Hebron Research Institute (VHIR), Universitat Autónoma de Barcelona, 119-129 Pg. Vall d'Hebron, Barcelona, 08035, Spain
| | - Miguel Abal
- Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela, 15706, Spain
- University of Santiago de Compostela (USC), Praza do Obradoiro, 0, Santiago de Compostela, 15705, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Gema Moreno-Bueno
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
- MD Anderson Cancer Center Foundation, C/Gómez Hemans 2, Madrid, 28033, Spain.
- Instituto de Investigaciones Biomedicas "Sols-Morreale" CSIC-UAM, C/Arturo Dupurier 4, Madrid, 28029, Spain.
| | - Laura Muinelo-Romay
- Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela, 15706, Spain.
- University of Santiago de Compostela (USC), Praza do Obradoiro, 0, Santiago de Compostela, 15705, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
| |
Collapse
|
33
|
Iida N, Imai M, Okamoto W, Kato T, Esaki T, Kato K, Komatsu Y, Yuki S, Masuishi T, Nishina T, Ebi H, Taniguchi H, Nonomura N, Sunakawa Y, Shiozawa M, Yamazaki K, Boku S, Bando H, Shiraishi Y, Kobayashi M, Goto H, Sato A, Fujii S, Yoshino T, Nakamura Y. Novel ERBB2 Variant Potentially Associated with Resistance against Anti-HER2 Monoclonal Antibody-Based Therapy in ERBB2-Amplified Metastatic Colorectal Cancer. Clin Cancer Res 2024; 30:4167-4178. [PMID: 39163021 PMCID: PMC11393546 DOI: 10.1158/1078-0432.ccr-24-1023] [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: 03/29/2024] [Revised: 05/26/2024] [Accepted: 07/16/2024] [Indexed: 08/21/2024]
Abstract
PURPOSE HER2-targeted therapies in ERBB2-amplified metastatic colorectal cancer (mCRC) are effective; however, a notable portion of patients do not respond to treatment, and secondary resistance occurs in most patients receiving these treatments. The purpose of this study was to investigate determinants of treatment efficacy and resistance in patients with ERBB2-amplified mCRC who received HER2-targeted therapy by analyzing multiomics data. EXPERIMENTAL DESIGN We investigated genomic data from a nationwide large cancer genomic screening project, the SCRUM-Japan project. We analyzed paired genome and transcriptome data of tissue and genomic data of ctDNA collected pre- and postprogression in patients enrolled in the related trial, TRIUMPH, in ERBB2-amplified mCRC. RESULTS In 155 patients with ERBB2-amplified solid tumors who received HER2-targeted therapy based on the SCRUM-Japan project, the objective response rate was 50%, 51%, and 35% in ERBB2 wild-type, variant of unknown significance, and pathogenic variant groups, respectively. In the paired genome and transcriptome data analyses in TRIUMPH, we identified the novel splicing-associated variant c.644-66_-2del in one of the 11 patients with paired whole-exome sequencing and whole-transcriptome sequencing data sets, which lacks the binding domain of pertuzumab, in progressed metastatic tumor as a variant with potential pathogenicity. The time-course ctDNA analysis detected c.644-66_-2del as an acquired variant. CONCLUSIONS This study highlighted the importance of ERBB2 genomic status when evaluating the efficacy of HER2-targeted therapies in ERBB2-amplified mCRC. The identification of a novel splicing-associated variant may provide insights into potential mechanisms of treatment resistance. Furthermore, we demonstrated the utility of ctDNA to follow the acquired genomic status of mCRC tumors.
Collapse
Affiliation(s)
- Naoko Iida
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan.
| | - Mitsuho Imai
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan.
| | - Wataru Okamoto
- Department of Clinical Oncology, Hiroshima University Hospital, Hiroshima, Japan.
| | - Takeshi Kato
- Department of Surgery, NHO Osaka National Hospital, Osaka, Japan.
| | - Taito Esaki
- Department of Gastrointestinal and Medical Oncology, NHO Kyushu Cancer Center, Fukuoka, Japan.
| | - Ken Kato
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Yoshito Komatsu
- Department of Cancer Center, Hokkaido University Hospital, Sapporo, Japan.
| | - Satoshi Yuki
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Sapporo, Japan.
| | - Toshiki Masuishi
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan.
| | - Tomohiro Nishina
- Gastrointestinal Medical Oncology, NHO Shikoku Cancer Center, Matsuyama, Japan.
| | - Hiromichi Ebi
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, Japan.
| | - Hiroya Taniguchi
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan.
| | - Norio Nonomura
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Japan.
| | - Yu Sunakawa
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan.
| | - Manabu Shiozawa
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan.
| | - Kentaro Yamazaki
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shunto-gun, Japan.
| | - Shogen Boku
- Cancer Treatment Center, Kansai Medical University Hospital, Hirakata, Japan.
| | - Hideaki Bando
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
| | - Yuichi Shiraishi
- Division of Genome Analysis Platform Development, National Cancer Center Research Institute, Tokyo, Japan.
| | - Maki Kobayashi
- Translational Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan.
| | - Hiroki Goto
- Translational Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan.
| | - Akihiro Sato
- Clinical Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan.
| | - Satoshi Fujii
- Division of Pathology, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Tokyo, Japan.
- Department of Molecular Pathology, Yokohama City University Graduate School of Medicine, Kanagawa, Japan.
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
| | - Yoshiaki Nakamura
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan.
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
| |
Collapse
|
34
|
Roodhart JML, Koopman M. Trastuzumab deruxtecan in HER2-positive metastatic colorectal cancer: less is more? Lancet Oncol 2024; 25:1104-1105. [PMID: 39116901 DOI: 10.1016/s1470-2045(24)00397-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 07/14/2024] [Indexed: 08/10/2024]
Affiliation(s)
- Jeanine M L Roodhart
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Miriam Koopman
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.
| |
Collapse
|
35
|
Wang L, Lin Y, Yao Z, Babu N, Lin W, Chen C, Du L, Cai S, Pan Y, Xiong X, Ye Q, Ren H, Zhang D, Chen Y, Yeung SCJ, Bremer E, Zhang H. Targeting undruggable phosphatase overcomes trastuzumab resistance by inhibiting multi-oncogenic kinases. Drug Resist Updat 2024; 76:101118. [PMID: 39094301 DOI: 10.1016/j.drup.2024.101118] [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: 01/23/2024] [Revised: 06/12/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024]
Abstract
AIMS Resistance to targeted therapy is one of the critical obstacles in cancer management. Resistance to trastuzumab frequently develops in the treatment for HER2+ cancers. The role of protein tyrosine phosphatases (PTPs) in trastuzumab resistance is not well understood. In this study, we aim to identify pivotal PTPs affecting trastuzumab resistance and devise a novel counteracting strategy. METHODS Four public datasets were used to screen PTP candidates in relation to trastuzumab responsiveness in HER2+ breast cancer. Tyrosine kinase (TK) arrays were used to identify kinases that linked to protein tyrosine phosphate receptor type O (PTPRO)-enhanced trastuzumab sensitivity. The efficacy of small activating RNA (saRNA) in trastuzumab-conjugated silica nanoparticles was tested for PTPRO upregulation and resistance mitigation in cell models, a transgenic mouse model, and human cancer cell line-derived xenograft models. RESULTS PTPRO was identified as the key PTP which influences trastuzumab responsiveness and patient survival. PTPRO de-phosphorated several TKs, including the previously overlooked substrate ERBB3, thereby inhibiting multiple oncogenic pathways associated with drug resistance. Notably, PTPRO, previously deemed "undruggable," was effectively upregulated by saRNA-loaded nanoparticles. The upregulated PTPRO simultaneously inhibited ERBB3, ERBB2, and downstream SRC signaling pathways, thereby counteracting trastuzumab resistance. CONCLUSIONS Antibody-conjugated saRNA represents an innovative approach for targeting "undruggable" PTPs.
Collapse
Affiliation(s)
- Lu Wang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China; Zhuhai Institute of Jinan University, Zhuhai, China
| | - Yusheng Lin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China; Zhuhai Institute of Jinan University, Zhuhai, China; Department of Thoracic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China; Department of Hematology, University of Groningen, University Medical Center Groningen, the Netherlands; Shantou University Medical College, Shantou, China
| | - Zhimeng Yao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China; Zhuhai Institute of Jinan University, Zhuhai, China; Department of Urology Surgery, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Nipun Babu
- Shantou University Medical College, Shantou, China
| | - Wan Lin
- Shantou University Medical College, Shantou, China
| | | | - Liang Du
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China; Zhuhai Institute of Jinan University, Zhuhai, China
| | - Songwang Cai
- Department of Thoracic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yunlong Pan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiao Xiong
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China; Zhuhai Institute of Jinan University, Zhuhai, China
| | - Qiantao Ye
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China; Zhuhai Institute of Jinan University, Zhuhai, China
| | - Hongzheng Ren
- Department of Pathology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China; Department of Pathology, Heping Hospital, Changzhi Medical College, Changzhi, China
| | - Dianzheng Zhang
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, USA
| | - Yexi Chen
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Sai-Ching Jim Yeung
- Department of Emergency Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Edwin Bremer
- Department of Hematology, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Hao Zhang
- Department of Pathology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China; Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China; Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China.
| |
Collapse
|
36
|
Raghav K, Siena S, Takashima A, Kato T, Van den Eynde M, Pietrantonio F, Komatsu Y, Kawakami H, Peeters M, Andre T, Lonardi S, Yamaguchi K, Tie J, Castro CG, Hsu HC, Strickler JH, Kim TY, Cha Y, Barrios D, Yan Q, Kamio T, Kobayashi K, Boran A, Koga M, Allard JD, Yoshino T. Trastuzumab deruxtecan in patients with HER2-positive advanced colorectal cancer (DESTINY-CRC02): primary results from a multicentre, randomised, phase 2 trial. Lancet Oncol 2024; 25:1147-1162. [PMID: 39116902 DOI: 10.1016/s1470-2045(24)00380-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND Trastuzumab deruxtecan has shown encouraging activity in patients with treatment-refractory HER2-positive, RAS wild-type and BRAF wild-type metastatic colorectal cancer. Dose optimisation and further antitumour assessments in patients with RAS mutations and those with previous anti-HER2 therapy are warranted. We aimed to evaluate two doses of trastuzumab deruxtecan (5·4 mg/kg and 6·4 mg/kg) to establish the recommended dose in patients with pretreated HER2-positive, RAS wild-type or mutant metastatic colorectal cancer. METHODS DESTINY-CRC02 was a multicentre, randomised, two-stage, two-arm, phase 2 study done in 53 research hospitals and medical centres in Australia, Belgium, France, Italy, Japan, South Korea, Spain, Taiwan, the UK, and the USA. Eligible patients were aged 18 years and older or 20 years and older (depending on region) with pretreated pathologically documented, unresectable, recurrent, or metastatic HER2-positive, and RAS wild-type or mutant colorectal cancer. Patients were required to have an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1 and have received previous chemotherapy, and anti-EGFR, anti-VEGF, or anti-PD-L1 therapy, if clinically indicated. In stage 1, patients were randomly assigned (1:1), via a secure interactive response technology system, to receive 5·4 mg/kg or 6·4 mg/kg trastuzumab deruxtecan administered intravenously every 21 days. Stratification factors were ECOG performance status, HER2 status, and RAS status. In stage 2, patients were assigned into the 5·4 mg/kg treatment group only. The primary endpoint was confirmed objective response rate by blinded independent central review, assessed in all patients for whom treatment was assigned (full analysis set). Safety was assessed in all patients who received at least one dose of study drug. This trial is registered with ClinicalTrials.gov, NCT04744831, and is ongoing (not recruiting). FINDINGS Between March 5, 2021, and March 29, 2022, 135 patients were centrally screened, 122 of whom were enrolled. In stage 1, 40 patients each were randomly assigned to receive trastuzumab deruxtecan 5·4 mg/kg and 6·4 mg/kg. In stage 2, an additional 42 patients were enrolled in the 5·4 mg/kg group. 64 (52%) participants were male and 58 (48%) were female. The median duration of follow-up was 8·9 months (IQR 6·7-10·5) in the 5·4 mg/kg group and 10·3 months (5·9-12·7) in the 6·4 mg/kg group. The confirmed objective response rate by blinded independent central review was 37·8% (31/82 [95% CI 27·3-49·2]) in the 5·4 mg/kg group and 27·5% (11/40 [14·6-43·9]) in the 6·4 mg/kg group. 34 (41%) of 83 patients in the 5·4 mg/kg group and 19 (49%) of 39 in the 6·4 mg/kg group had grade 3 or worse drug-related treatment-emergent adverse events. The most common grade 3 or worse drug-related treatment-emergent adverse events were neutrophil count decreased (13 [16%] of 83 patients), anaemia (six [7%]), nausea (six [7%]), and white blood cell count decreased (five [6%]) in the 5·4 mg/kg group; and were neutrophil count decreased (10 [26%] of 39 patients), anaemia (eight [21%]), platelet count decreased (four [10%]), and white blood cell count decreased (four [10%]) in the 6·4 mg/kg group. Drug-related serious adverse events occurred in 11 (13%) of 83 patients in the 5·4 mg/kg group and six (15%) of 39 patients in the 6·4 mg/kg group; the most common in the 5·4 mg/kg group was nausea (three [4%] patients) and the most common in the 6·4 mg/kg group were fatigue (two [5%] patients), neutropenia (two [5%]), and thrombocytopenia (two [5%]). A drug-related treatment-emergent adverse event related to death occurred in one (1%) patient in the 5·4 mg/kg group (due to hepatic failure). Adjudicated drug-related interstitial lung disease or pneumonitis events were observed in seven (8%) patients in the 5·4 mg/kg group (all grade 1 or 2) and in five (13%) patients in the 6·4 mg/kg group (four grade 1 or 2; one grade 5). INTERPRETATION The promising antitumour activity and favourable safety profile support trastuzumab deruxtecan 5·4 mg/kg as the optimal single-agent dose for patients with pretreated HER2-positive metastatic colorectal cancer, including those with RAS mutations, previous anti-HER2 therapy, or both. FUNDING Daiichi Sankyo and AstraZeneca.
Collapse
Affiliation(s)
- Kanwal Raghav
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Salvatore Siena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Atsuo Takashima
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Takeshi Kato
- Department of Surgery, NHO Osaka National Hospital, Osaka, Japan
| | - Marc Van den Eynde
- Institut Roi Albert II, Cliniques Universitaires Saint-Luc, UCLouvain, Brussels, Belgium
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Hisato Kawakami
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Marc Peeters
- Department of Oncology, Antwerp University Hospital, Edegem, Belgium
| | - Thierry Andre
- Department of Medical Oncology, Sorbonne University, Saint-Antoine Hospital, Paris, France
| | - Sara Lonardi
- Oncology Unit 1, Veneto Institute of Oncology, IRCCS, Padua, Italy
| | - Kensei Yamaguchi
- Department of Gastroenterological Chemotherapy, The Cancer Institute Hospital of JFCR, Tokyo, Japan
| | - Jeanne Tie
- Medical Oncology Department, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | | | - Hung-Chih Hsu
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - John H Strickler
- Division of Medical Oncology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Tae-You Kim
- Seoul National University Hospital, Seoul, South Korea
| | - Yongjun Cha
- Division of Medical Oncology, Center for Colorectal Cancer, National Cancer Center, Research Institute and Hospital, Goyang, South Korea
| | | | - Qi Yan
- Daiichi Sankyo, Basking Ridge, NJ, USA
| | | | | | | | | | | | - Takayuki Yoshino
- Department for the Promotion of Drug and Diagnostic Development and Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
37
|
Jones L, Cunningham D, Starling N. HER-2 directed therapies across gastrointestinal tract cancers - A new frontier. Cancer Treat Rev 2024; 129:102789. [PMID: 38959629 DOI: 10.1016/j.ctrv.2024.102789] [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: 05/23/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 07/05/2024]
Abstract
Gastrointestinal (GI) cancers are common and in the metastatic setting they have a poor prognosis. The current mainstay of treatment of GI cancers is chemotherapy; however, the biomarker-directed treatment landscape is evolving. HER-2 is overexpressed in a portion of GI cancers and is an emerging target for therapy, with recent FDA tumor agnostic approval for trastuzumab deruxtecan. Testing for HER-2 expression is not standardized across GI cancers, methodology requires further optimization and standardization as HER-2 targeted therapy emerges into the treatment landscape. There is established rationale for use of HER-2 targeted therapy in first line treatment of metastatic gastric cancer, and emerging evidence with variable benefit in bile duct, pancreatic and colorectal cancers.
Collapse
Affiliation(s)
- Lauren Jones
- Gastrointestinal and Lymphoma Unit, Royal Marsden NHS Foundation Trust, London, Sutton, UK
| | - David Cunningham
- Gastrointestinal and Lymphoma Unit, Royal Marsden NHS Foundation Trust, London, Sutton, UK
| | - Naureen Starling
- Gastrointestinal and Lymphoma Unit, Royal Marsden NHS Foundation Trust, London, Sutton, UK.
| |
Collapse
|
38
|
Mo C, Sterpi M, Jeon H, Bteich F. Resistance to Anti-HER2 Therapies in Gastrointestinal Malignancies. Cancers (Basel) 2024; 16:2854. [PMID: 39199625 PMCID: PMC11352490 DOI: 10.3390/cancers16162854] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024] Open
Abstract
Human epidermal growth factor 2 (HER2) is a tyrosine kinase receptor that interacts with multiple signaling pathways related to cellular growth and proliferation. Overexpression or amplification of HER2 is linked to various malignancies, and there have been decades of research dedicated to targeting HER2. Despite the landmark ToGA trial, progress in HER2-positive gastrointestinal malignancies has been hampered by drug resistance. This review examines current HER2 expression patterns and therapies for gastroesophageal, colorectal, biliary tract, and small bowel cancers, while dissecting potential resistance mechanisms that limit treatment effectiveness.
Collapse
Affiliation(s)
- Christiana Mo
- Department of Medical Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (C.M.); (M.S.); (H.J.)
- Department of Medical Oncology, Montefiore Medical Center, Bronx, NY 10467, USA
| | - Michelle Sterpi
- Department of Medical Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (C.M.); (M.S.); (H.J.)
- Department of Medical Oncology, Montefiore Medical Center, Bronx, NY 10467, USA
| | - Hyein Jeon
- Department of Medical Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (C.M.); (M.S.); (H.J.)
- Department of Medical Oncology, Montefiore Medical Center, Bronx, NY 10467, USA
| | - Fernand Bteich
- Department of Medical Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (C.M.); (M.S.); (H.J.)
- Department of Medical Oncology, Montefiore Medical Center, Bronx, NY 10467, USA
| |
Collapse
|
39
|
Chong X, Madeti Y, Cai J, Li W, Cong L, Lu J, Mo L, Liu H, He S, Yu C, Zhou Z, Wang B, Cao Y, Wang Z, Shen L, Wang Y, Zhang X. Recent developments in immunotherapy for gastrointestinal tract cancers. J Hematol Oncol 2024; 17:65. [PMID: 39123202 PMCID: PMC11316403 DOI: 10.1186/s13045-024-01578-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024] Open
Abstract
The past few decades have witnessed the rise of immunotherapy for Gastrointestinal (GI) tract cancers. The role of immune checkpoint inhibitors (ICIs), particularly programmed death protein 1 (PD-1) and PD ligand-1 antibodies, has become increasingly pivotal in the treatment of advanced and perioperative GI tract cancers. Currently, anti-PD-1 plus chemotherapy is considered as first-line regimen for unselected advanced gastric/gastroesophageal junction adenocarcinoma (G/GEJC), mismatch repair deficient (dMMR)/microsatellite instability-high (MSI-H) colorectal cancer (CRC), and advanced esophageal cancer (EC). In addition, the encouraging performance of claudin18.2-redirected chimeric antigen receptor T-cell (CAR-T) therapy in later-line GI tract cancers brings new hope for cell therapy in solid tumour treatment. Nevertheless, immunotherapy for GI tumour remains yet precise, and researchers are dedicated to further maximising and optimising the efficacy. This review summarises the important research, latest progress, and future directions of immunotherapy for GI tract cancers including EC, G/GEJC, and CRC.
Collapse
Affiliation(s)
- Xiaoyi Chong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Yelizhati Madeti
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Jieyuan Cai
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Wenfei Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Lin Cong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Jialin Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Liyang Mo
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Huizhen Liu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Siyi He
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Chao Yu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Zhiruo Zhou
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Boya Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Yanshuo Cao
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Zhenghang Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Yakun Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| |
Collapse
|
40
|
Osumi H, Shinozaki E, Nakamura Y, Esaki T, Yasui H, Taniguchi H, Satake H, Sunakawa Y, Komatsu Y, Kagawa Y, Denda T, Shiozawa M, Satoh T, Nishina T, Goto M, Takahashi N, Kato T, Bando H, Yamaguchi K, Yoshino T. Clinical features associated with NeoRAS wild-type metastatic colorectal cancer A SCRUM-Japan GOZILA substudy. Nat Commun 2024; 15:5885. [PMID: 39003289 PMCID: PMC11246505 DOI: 10.1038/s41467-024-50026-4] [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: 10/31/2023] [Accepted: 06/11/2024] [Indexed: 07/15/2024] Open
Abstract
"NeoRAS WT" refers to the loss of RAS mutations (MTs) following first-line treatment in metastatic colorectal cancer (mCRC). We evaluate the incidence and clinicopathological characteristics of NeoRAS WT mCRC using next-generation sequencing of plasma circulating tumor DNA. Patients with mCRC enrolled in the GOZILA study initially diagnosed with tissue RAS MT mCRC and received subsequent systemic therapy are eligible. NeoRAS WT is defined as the absence of detectable RAS MT in plasma and assessed in all eligible patients (Group A) and in a subgroup with at least one somatic alteration detected in plasma (Group B). Overall, 478 patients are included. NeoRAS WT prevalence is 19.0% (91/478) in Group A and 9.8% (42/429) in Group B. Absence of liver or lymph node metastasis and tissue RAS MTs other than KRAS exon 2 MTs are significantly associated with NeoRAS WT emergence. Overall, 1/6 and 2/6 patients with NeoRAS WT treated with anti-EGFR monoclonal antibodies (mAbs) show partial response and stable disease for ≥6 months, respectively. NeoRAS WT mCRC is observed at a meaningful prevalence, and anti-EGFR mAb-based therapy may be effective.
Collapse
Affiliation(s)
- Hiroki Osumi
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Eiji Shinozaki
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan.
| | - Yoshiaki Nakamura
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Taito Esaki
- Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Hisateru Yasui
- Department of Medical Oncology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Hiroya Taniguchi
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Hironaga Satake
- Department of Medical Oncology, Kochi Medical School, Kochi, Japan
| | - Yu Sunakawa
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Yoshito Komatsu
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Sapporo, Japan
| | - Yoshinori Kagawa
- Department of Gastroenterological Surgery, Osaka General Medical Center, Osaka, Japan
| | - Tadamichi Denda
- Division of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Manabu Shiozawa
- Department of Gastroenterological Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Taroh Satoh
- Palliative and Supportive Care Center, Osaka University Hospital, Suita, Japan
| | - Tomohiro Nishina
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Ehime, Japan
| | - Masahiro Goto
- Department of Cancer Chemotherapy Center, Osaka Medical and Pharmaceutical University Hospital, Osaka, Japan
| | - Naoki Takahashi
- Department of Gastroenterology, Saitama Cancer Center, Saitama, Japan
| | - Takeshi Kato
- Department of Surgery, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Hideaki Bando
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kensei Yamaguchi
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
| |
Collapse
|
41
|
Napolitano S, Martini G, Ciardiello D, Del Tufo S, Martinelli E, Troiani T, Ciardiello F. Targeting the EGFR signalling pathway in metastatic colorectal cancer. Lancet Gastroenterol Hepatol 2024; 9:664-676. [PMID: 38697174 DOI: 10.1016/s2468-1253(23)00479-x] [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: 10/13/2023] [Revised: 12/21/2023] [Accepted: 12/28/2023] [Indexed: 05/04/2024]
Abstract
Epidermal growth factor receptor (EGFR) and its activated downstream signalling pathways play a crucial role in colorectal cancer development and progression. After four decades of preclinical, translational, and clinical research, it has been shown that blocking the EGFR signalling pathway at different molecular levels represents a fundamental therapeutic strategy for patients with metastatic colorectal cancer. Nevertheless, the efficacy of molecularly targeted therapies is inescapably limited by the insurgence of mechanisms of acquired cancer cell resistance. Thus, in the era of precision medicine, a deeper understanding of the complex molecular landscape of metastatic colorectal cancer is required to deliver the best treatment choices to all patients. Major efforts are currently ongoing to improve patient selection, improve the efficacy of available treatments targeting the EGFR pathway, and develop novel combination strategies to overcome therapy resistance within the continuum of care of metastatic colorectal cancer.
Collapse
Affiliation(s)
- Stefania Napolitano
- Department of Precision Medicine, Università degli studi della Campania Luigi Vanvitelli, Napoli, Italy
| | - Giulia Martini
- Department of Precision Medicine, Università degli studi della Campania Luigi Vanvitelli, Napoli, Italy
| | - Davide Ciardiello
- Department of Precision Medicine, Università degli studi della Campania Luigi Vanvitelli, Napoli, Italy; Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO, IRCCS, Milan, Italy
| | - Sara Del Tufo
- Department of Precision Medicine, Università degli studi della Campania Luigi Vanvitelli, Napoli, Italy
| | - Erika Martinelli
- Department of Precision Medicine, Università degli studi della Campania Luigi Vanvitelli, Napoli, Italy
| | - Teresa Troiani
- Department of Precision Medicine, Università degli studi della Campania Luigi Vanvitelli, Napoli, Italy
| | - Fortunato Ciardiello
- Department of Precision Medicine, Università degli studi della Campania Luigi Vanvitelli, Napoli, Italy.
| |
Collapse
|
42
|
Piazza GA, Chandrasekaran P, Maxuitenko YY, Budhwani KI. Assessment of KRAS G12C inhibitors for colorectal cancer. Front Oncol 2024; 14:1412435. [PMID: 38978742 PMCID: PMC11228624 DOI: 10.3389/fonc.2024.1412435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/06/2024] [Indexed: 07/10/2024] Open
Abstract
Colorectal cancer (CRC) is a highly prevalent and lethal cancer worldwide. Approximately 45% of CRC patients harbor a gain-in-function mutation in KRAS. KRAS is the most frequently mutated oncogene accounting for approximately 25% of all human cancers. Gene mutations in KRAS cause constitutive activation of the KRAS protein and MAPK/AKT signaling, resulting in unregulated proliferation and survival of cancer cells and other aspects of malignant transformation, progression, and metastasis. While KRAS has long been considered undruggable, the FDA recently approved two direct acting KRAS inhibitors, Sotorasib and Adagrasib, that covalently bind and inactivate KRASG12C. Both drugs showed efficacy for patients with non-small cell lung cancer (NSCLC) diagnosed with a KRASG12C mutation, but for reasons not well understood, were considerably less efficacious for CRC patients diagnosed with the same mutation. Thus, it is imperative to understand the basis for resistance to KRASG12C inhibitors, which will likely be the same limitations for other mutant specific KRAS inhibitors in development. This review provides an update on clinical trials involving CRC patients treated with KRASG12C inhibitors as a monotherapy or combined with other drugs. Mechanisms that contribute to resistance to KRASG12C inhibitors and the development of novel RAS inhibitors with potential to escape such mechanisms of resistance are also discussed.
Collapse
Affiliation(s)
- Gary A Piazza
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL, United States
| | | | - Yulia Y Maxuitenko
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL, United States
| | - Karim I Budhwani
- CerFlux, Birmingham, AL, United States
- University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
43
|
Zhou Y, Wu S, Qu FJ. Therapeutic strategies targeting the epidermal growth factor receptor signaling pathway in metastatic colorectal cancer. World J Gastrointest Oncol 2024; 16:2362-2379. [PMID: 38994135 PMCID: PMC11236217 DOI: 10.4251/wjgo.v16.i6.2362] [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: 12/07/2023] [Revised: 02/13/2024] [Accepted: 04/01/2024] [Indexed: 06/14/2024] Open
Abstract
More than 1.9 million new colorectal cancer (CRC) cases and 935000 deaths were estimated to occur worldwide in 2020, representing about one in ten cancer cases and deaths. Overall, colorectal ranks third in incidence, but second in mortality. More than half of the patients are in advanced stages at diagnosis. Treatment options are complex because of the heterogeneity of the patient population, including different molecular subtypes. Treatments have included conventional fluorouracil-based chemotherapy, targeted therapy, immunotherapy, etc. In recent years, with the development of genetic testing technology, more and more targeted drugs have been applied to the treatment of CRC, which has further prolonged the survival of metastatic CRC patients.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Oncology, Affiliated Dalian Third People’s Hospital of Dalian Medical University, Dalian 116033, Liaoning Province, China
| | - Shuang Wu
- Department of Oncology, Affiliated Dalian Third People’s Hospital of Dalian Medical University, Dalian 116033, Liaoning Province, China
| | - Fan-Jie Qu
- Department of Oncology, Affiliated Dalian Third People’s Hospital of Dalian Medical University, Dalian 116033, Liaoning Province, China
| |
Collapse
|
44
|
Bando H, Misumi T, Sakamoto Y, Takeda Y, Nakamura Y, Mizuguchi K, Aoyagi Y, Miki I, Kuroda T, Kasai R, Suzuki T, Yoshino T, Ohtsu A. Appropriate Relevancy and Reliability of Real-World Data for the Utilization of Regulatory Submission. Clin Colorectal Cancer 2024; 23:111-117. [PMID: 38679555 DOI: 10.1016/j.clcc.2024.04.001] [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: 02/16/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 05/01/2024]
Abstract
The extraction of data that contribute to regulatory approval from real-world data (RWD) is difficult because of the lack of a standardized data format and extraction methodology. Additionally, when real-world evidence (RWE) is used as an external control group, the similarity between internal and external control data is not evaluated. To investigate the data extraction methodology for the external control data of rare molecular subtypes, we have initiated the "REALISE" study. In this study, we aim to elucidate the "relevance" and "reliability" of RWD/RWE necessary for regulatory approval. As most databases are not designed for regulatory use in the creation phase, we will investigate retrospective methodologies to ensure RWD/RWE reliability. This study will compare the "relevance" and "reliability" of the ARCAD global database, SCRUM-Japan Registry, SCRUM-Japan observational study, and Flatiron Health RWD, and statistically analyze the differences and similarities among the four databases. We will also examine the methodology for extracting sufficiently relevant data from the SCRUM-Japan observational study. Additionally, if the reliability of the RWD/RWE does not reach the required level for regulatory approval, we will examine the methodologies to ensure the "reliability" of the SCRUM-Japan observational study for regulatory approval. The obtained results will be submitted to the "Consultation for Development of Registry" in the Pharmaceuticals and Medical Devices Agency, and we will discuss the standard methodology. The procedures and findings identified in the REALISE study will be organized from the perspectives of "database construction," "data analysis," and "outcome evaluation" and will be issued as "the draft guidelines."
Collapse
Affiliation(s)
- Hideaki Bando
- Translational Research Support Office, Division of Drug and Diagnostic Development Promotion, Department for the Promotion of Drug and Diagnostic Development, National Cancer Center Hospital East, Kashiwa, Chiba, Japan; Division of Data Science, Department for the Promotion of Drug and Diagnostic Development, National Cancer Center Hospital East, Kashiwa, Chiba, Japan; Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan.
| | - Toshihiro Misumi
- Division of Data Science, Department for the Promotion of Drug and Diagnostic Development, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Yasutoshi Sakamoto
- Translational Research Support Office, Division of Drug and Diagnostic Development Promotion, Department for the Promotion of Drug and Diagnostic Development, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Yuriko Takeda
- Division of Data Science, Department for the Promotion of Drug and Diagnostic Development, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Yoshiaki Nakamura
- Translational Research Support Office, Division of Drug and Diagnostic Development Promotion, Department for the Promotion of Drug and Diagnostic Development, National Cancer Center Hospital East, Kashiwa, Chiba, Japan; Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan; International Research Promotion Office, Division of Drug and Diagnostic Development Promotion, Department for the Promotion of Drug and Diagnostic Development, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Kazuya Mizuguchi
- Department of Medical Information, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Yoshihiro Aoyagi
- Information Technology Management Section, Clinical Research Management Division, Clinical Research Support Office, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Izumi Miki
- Translational Research Support Office, Division of Drug and Diagnostic Development Promotion, Department for the Promotion of Drug and Diagnostic Development, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Tomohiro Kuroda
- Graduate School of Medicine, Kyoto University Hospital, Sakyo-ku, Kyoto, Japan
| | - Ryu Kasai
- Science & Technology Intelligence Department, Chugai Pharmaceutical Co., Ltd., Chuo-ku, Tokyo, Japan
| | - Takuya Suzuki
- Japan and Asia Clinical Development Department, Oncology, Deep Human Biology Learning, Eisai Co., Ltd., Bunkyo-ku, Tokyo, Japan
| | - Takayuki Yoshino
- Translational Research Support Office, Division of Drug and Diagnostic Development Promotion, Department for the Promotion of Drug and Diagnostic Development, National Cancer Center Hospital East, Kashiwa, Chiba, Japan; Division of Data Science, Department for the Promotion of Drug and Diagnostic Development, National Cancer Center Hospital East, Kashiwa, Chiba, Japan; Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Atsushi Ohtsu
- National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| |
Collapse
|
45
|
González NS, Marchese PV, Baraibar I, Ros J, Salvà F, Rodríguez M, Salvà C, Vaghi C, Alcaraz A, García A, Tabernero J, Élez E. Epidermal growth factor receptor antagonists in colorectal cancer: emerging strategies for precision therapy. Expert Opin Investig Drugs 2024; 33:613-625. [PMID: 38775361 DOI: 10.1080/13543784.2024.2349287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/25/2024] [Indexed: 06/25/2024]
Abstract
INTRODUCTION The global prevalence of colorectal cancer highlights the need to enhance treatment strategies for improved patient outcomes. The pivotal role of epidermal growth factor receptor (EGFR) signaling in regulating cellular processes for this disease pinpoints its value as a therapeutic target, despite the emergence of resistance mechanisms over time. AREAS COVERED This review discusses the clinical evidence supporting the use of EGFR inhibitors in molecularly-selected patients based on molecular characteristics (notably BRAF V600E and KRAS G12C) including combination approaches targeting different points in in the signaling pathway, as well as strategies such as EGFR inhibitor rechallenge. The role of HER2 inhibitors and emerging approaches such as bispecific antibodies are also reviewed. EXPERT OPINION Recently, inhibitors targeting the KRAS G12C variant have emerged, albeit with modest monotherapy activity compared to other tumor types, emphasizing the influence of histologic origins on the EGFR signaling pathway. Integration of EGFR inhibitors into precision medicine has facilitated tailored therapies addressing resistance mechanisms. Patient selection for EGFR inhibitor rechallenge guided by ctDNA findings is crucial, with ongoing investigations exploring novel combinations to enhance EGFR blockade, highlighting the transformative potential of precision medicine in shaping the future of mCRC treatment toward personalized and targeted approaches.
Collapse
Affiliation(s)
- Nadia Saoudi González
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Vall d'Hebron Hospital Campus, Barcelona, Spain
| | | | - Iosune Baraibar
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Vall d'Hebron Hospital Campus, Barcelona, Spain
| | - Javier Ros
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Vall d'Hebron Hospital Campus, Barcelona, Spain
| | - Francesc Salvà
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Vall d'Hebron Hospital Campus, Barcelona, Spain
| | - Marta Rodríguez
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Vall d'Hebron Hospital Campus, Barcelona, Spain
| | - Clara Salvà
- Vall d'Hebron Hospital Campus, Barcelona, Spain
| | - Caterina Vaghi
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Adriana Alcaraz
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ariadna García
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Josep Tabernero
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Vall d'Hebron Hospital Campus, Barcelona, Spain
| | - Elena Élez
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Vall d'Hebron Hospital Campus, Barcelona, Spain
| |
Collapse
|
46
|
Nakano T, Takao S, Dairaku K, Uno N, Low S(A, Hashimoto M, Tsuda Y, Hisamatsu Y, Toshima T, Yonemura Y, Masuda T, Eto K, Ikegami T, Fukunaga Y, Niida A, Nagayama S, Mimori K. Implementable assay for monitoring minimum residual disease after radical treatment for colorectal cancer. Cancer Sci 2024; 115:1989-2001. [PMID: 38531808 PMCID: PMC11145105 DOI: 10.1111/cas.16149] [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: 10/28/2023] [Revised: 02/09/2024] [Accepted: 02/29/2024] [Indexed: 03/28/2024] Open
Abstract
Considering the cost and invasiveness of monitoring postoperative minimal residual disease (MRD) of colorectal cancer (CRC) after adjuvant chemoradiotherapy (ACT), we developed a favorable approach based on methylated circulating tumor DNA to detect MRD after radical resection. Analyzing the public database, we identified the methylated promoter regions of the genes FGD5, GPC6, and MSC. Using digital polymerase chain reaction (dPCR), we termed the "amplicon of methylated sites using a specific enzyme" assay as "AMUSE." We examined 180 and 114 pre- and postoperative serial plasma samples from 28 recurrent and 19 recurrence-free pathological stage III CRC patients, respectively. The results showed 22 AMUSE-positive of 28 recurrent patients (sensitivity, 78.6%) and 17 AMUSE-negative of 19 recurrence-free patients (specificity, 89.5%). AMUSE predicted recurrence 208 days before conventional diagnosis using radiological imaging. Regarding ACT evaluation by the reactive response, 19 AMUSE-positive patients during their second or third blood samples showed a significantly poorer prognosis than the other patients (p = 9E-04). The AMUSE assay stratified four groups by the altered patterns of tumor burden postoperatively. Interestingly, only 34.8% of cases tested AMUSE-negative during ACT treatment, indicating eligibility for ACT. The AMUSE assay addresses the clinical need for accurate MRD monitoring with universal applicability, minimal invasiveness, and cost-effectiveness, thereby enabling the timely detection of recurrences. This assay can effectively evaluate the efficacy of ACT in patients with stage III CRC following curative resection. Our study strongly recommends reevaluating the clinical application of ACT using the AMUSE assay.
Collapse
Affiliation(s)
- Takafumi Nakano
- Department of SurgeryKyushu University Beppu HospitalBeppuJapan
- Department of SurgeryThe Jikei University School of MedicineTokyoJapan
| | - Seiichiro Takao
- Department of SurgeryKyushu University Beppu HospitalBeppuJapan
- Department of Clinical Radiology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Katsushi Dairaku
- Department of SurgeryKyushu University Beppu HospitalBeppuJapan
- Department of SurgeryThe Jikei University School of MedicineTokyoJapan
| | - Naoki Uno
- Department of Laboratory MedicineNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| | - Siew‐Kee (Amanda) Low
- Department of Colorectal Surgery, Gastroenterological Cancer CenterCancer Institute Hospital, Japanese Foundation for Cancer ResearchTokyoJapan
| | | | - Yasuo Tsuda
- Department of SurgeryKyushu University Beppu HospitalBeppuJapan
| | | | - Takeo Toshima
- Department of SurgeryKyushu University Beppu HospitalBeppuJapan
| | - Yusuke Yonemura
- Department of SurgeryKyushu University Beppu HospitalBeppuJapan
| | - Takaaki Masuda
- Department of SurgeryKyushu University Beppu HospitalBeppuJapan
| | - Ken Eto
- Department of SurgeryThe Jikei University School of MedicineTokyoJapan
| | - Toru Ikegami
- Department of SurgeryThe Jikei University School of MedicineTokyoJapan
| | - Yosuke Fukunaga
- Department of Colorectal Surgery, Gastroenterological Cancer CenterCancer Institute Hospital, Japanese Foundation for Cancer ResearchTokyoJapan
| | - Atsushi Niida
- Human Genome Center, Institute of Medical ScienceUniversity of TokyoTokyoJapan
| | - Satoshi Nagayama
- Department of Colorectal Surgery, Gastroenterological Cancer CenterCancer Institute Hospital, Japanese Foundation for Cancer ResearchTokyoJapan
- Department of SurgeryUji‐Tokushukai Medical CenterUji, KyotoJapan
| | - Koshi Mimori
- Department of SurgeryKyushu University Beppu HospitalBeppuJapan
| |
Collapse
|
47
|
Spiekman IAC, Zeverijn LJ, Geurts BS, Verkerk K, Haj Mohammad SF, van der Noort V, Roepman P, de Leng WWJ, Jansen AML, Gootjes EC, de Groot DJA, Kerver ED, van Voorthuizen T, Roodhart JML, Valkenburg-van Iersel LBJ, Gelderblom H, Voest EE, Verheul HMW. Trastuzumab plus pertuzumab for HER2-amplified advanced colorectal cancer: Results from the drug rediscovery protocol (DRUP). Eur J Cancer 2024; 202:113988. [PMID: 38471288 DOI: 10.1016/j.ejca.2024.113988] [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: 01/18/2024] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024]
Abstract
BACKGROUND In 2-5% of patients with colorectal cancer (CRC), human epidermal growth factor 2 (HER2) is amplified or overexpressed. Despite prior evidence that anti-HER2 therapy confers clinical benefit (CB) in one-third of these patients, it is not approved for this indication in Europe. In the Drug Rediscovery Protocol (DRUP), patients are treated with off-label drugs based on their molecular profile. Here, we present the results of the cohort 'trastuzumab/pertuzumab for treatment-refractory patients with RAS/BRAF-wild-type HER2amplified metastatic CRC (HER2+mCRC)'. METHODS Patients with progressive treatment-refractory RAS/BRAF-wild-type HER2+mCRC with measurable disease were included for trastuzumab plus pertuzumab treatment. Primary endpoints of DRUP are CB (defined as confirmed objective response (OR) or stable disease (SD) ≥ 16 weeks) and safety. Patients were enrolled using a Simon-like 2-stage model, with 8 patients in stage 1 and 24 patients in stage 2 if at least 1/8 patients had CB. To identify biomarkers for response, whole genome sequencing (WGS) was performed on pre-treatment biopsies. RESULTS CB was observed in 11/24 evaluable patients (46%) with HER2+mCRC, seven patients achieved an OR (29%). Median duration of response was 8.4 months. Patients had undergone a median of 3 prior treatment lines. Median progression-free survival and overall survival were 4.3 months (95% CI 1.9-10.3) and 8.2 months (95% CI 7.2-14.7), respectively. No unexpected toxicities were observed. WGS provided potential explanations for resistance in 3/10 patients without CB, for whom WGS was available. CONCLUSIONS The results of this study confirm a clinically significant benefit of trastuzumab plus pertuzumab treatment in patients with HER2+mCRC.
Collapse
Affiliation(s)
- Ilse A C Spiekman
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, the Netherlands
| | - Laurien J Zeverijn
- Oncode Institute, Utrecht, the Netherlands; Department of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Birgit S Geurts
- Oncode Institute, Utrecht, the Netherlands; Department of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Karlijn Verkerk
- Oncode Institute, Utrecht, the Netherlands; Department of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Soemeya F Haj Mohammad
- Department of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Paul Roepman
- Hartwig Medical Foundation, Amsterdam, the Netherlands
| | - Wendy W J de Leng
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Anne M L Jansen
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Elske C Gootjes
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Derk-Jan A de Groot
- Department of Medical Oncology, University Medical Center Groningen, Groningen, the Netherlands
| | - Emile D Kerver
- Department of Medical Oncology, OLVG, Amsterdam, the Netherlands
| | | | - Jeanine M L Roodhart
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Liselot B J Valkenburg-van Iersel
- Division of Medical Oncology, Department of Internal Medicine, GROW school of Oncology and Development Biology, Maastricht University Center+, Maastricht, the Netherlands
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Emile E Voest
- Oncode Institute, Utrecht, the Netherlands; Department of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Henk M W Verheul
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, the Netherlands.
| |
Collapse
|
48
|
Li Y, Pan C, Gao Y, Zhang L, Ji D, Cui X, Zhang X, Cai Y, Zhang Y, Yao Y, Wang L, Leng J, Zhan T, Wu D, Gao Z, Sun YS, Li Z, Luo H, Wu A. Total Neoadjuvant Therapy With PD-1 Blockade for High-Risk Proficient Mismatch Repair Rectal Cancer. JAMA Surg 2024; 159:529-537. [PMID: 38381429 PMCID: PMC10882505 DOI: 10.1001/jamasurg.2023.7996] [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: 07/05/2023] [Accepted: 11/25/2023] [Indexed: 02/22/2024]
Abstract
Importance Total neoadjuvant therapy (TNT) is the standard treatment for locally advanced rectal cancer, especially for patients with high-risk factors. However, the efficacy of TNT combined with immunotherapy for patients with proficient mismatch repair (pMMR) rectal cancer is unknown. Objectives To evaluate the safety and efficacy of TNT with induction chemoimmunotherapy followed by long-course chemoradiation in patients with high-risk, pMMR rectal cancer and to identify potential molecular biomarkers associated with treatment efficacy. Design, Setting, and Participants This cohort study was a single-arm phase 2 trial conducted at Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, from June 2020 to October 2021. Biopsies and plasma were collected before treatment for whole-exome sequencing and cell-free DNA sequencing, respectively. Data were analyzed from May 2022 to September 2022. Interventions Participants received 3 cycles of induction oxaliplatin and capecitabine combined with camrelizumab and radiotherapy (50.6 Gy in 22 fractions) with concurrent capecitabine. Patients without disease progression received 2 cycles of consolidation oxaliplatin/capecitabine. Main Outcomes and Measures The primary end point was pathologic complete response rate. Results Of 25 patients enrolled (19 men [76%]; 6 women [24%]; median [IQR] age, 58 [48-64] years), 22 patients (88%) completed the TNT schedule. The pathologic complete response rate was 33.3% (7/21). Twelve patients (48%) achieved clinical complete response, and 4 patients (16%) chose to watch and wait. R0 resection was achieved in 21 of 21 patients, and the major pathologic response rate was 38.1% (8/21). The most common adverse event was nausea (80%, 20/25); grade 3 toxic effects occurred in 9 of 25 patients (36%). Patients with tumor shrinkage of 50% or greater after induction oxaliplatin/capecitabine and camrelizumab or clinical complete response had higher percentages of LRP1B mutation. Mutation of LRP1B was associated with high tumor mutation burden and tumor neoantigen burden. Patients with high tumor mutation burden all benefited from therapy. Conclusions and Relevance This study found that TNT with induction chemoimmunotherapy followed by long-course chemoradiation was safe and effective for patients with high-risk rectal cancer with pMMR status. Longer follow-up and larger clinical studies are needed to validate this innovative regimen. There is also an urgent need to further validate the predictive value of LRP1B and discover other novel biomarkers with potential predictive value for rectal cancer.
Collapse
Affiliation(s)
- Yingjie Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Unit III, Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Chaohu Pan
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology, Shenzhen, China
| | - Yuye Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Unit III, Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Li Zhang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Pathology, Beijing Cancer Hospital and Institute, Peking University School of Oncology, Beijing, China
| | - Dengbo Ji
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Unit III, Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaoli Cui
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology, Shenzhen, China
| | - Xiaoyan Zhang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Radiology, Beijing Cancer Hospital and Institute, Peking University School of Oncology, Beijing, China
| | - Yong Cai
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Radiation Oncology, Beijing Cancer Hospital and Institute, Peking University School of Oncology, Beijing, China
| | - Yangzi Zhang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Radiation Oncology, Beijing Cancer Hospital and Institute, Peking University School of Oncology, Beijing, China
| | - Yunfeng Yao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Unit III, Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Lin Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Unit III, Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jiahua Leng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Unit III, Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Tiancheng Zhan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Unit III, Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Dongfang Wu
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology, Shenzhen, China
| | - Zhibo Gao
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology, Shenzhen, China
| | - Ying-Shi Sun
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Radiology, Beijing Cancer Hospital and Institute, Peking University School of Oncology, Beijing, China
| | - Zhongwu Li
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Pathology, Beijing Cancer Hospital and Institute, Peking University School of Oncology, Beijing, China
| | - Haitao Luo
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology, Shenzhen, China
| | - Aiwen Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Unit III, Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
49
|
Nakamura D, Yanagita T, Fujii Y, Watanabe K, Suzuki T, Ushigome H, Nishigaki R, Sugimura N, Tanaka M, Ogawa R, Takahashi H, Shimura T, Hotta Y, Matsuo Y, Kondo M, Furukawa-Hibi Y, Takiguchi S. Unanticipated pathological clearance in two cases of clinical T4b dMMR/MSI-h advanced colorectal cancer: the potential of immune checkpoint inhibitors despite positive positron-emission tomography results. Surg Case Rep 2024; 10:105. [PMID: 38691233 PMCID: PMC11063011 DOI: 10.1186/s40792-024-01894-x] [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: 11/08/2023] [Accepted: 04/08/2024] [Indexed: 05/03/2024] Open
Abstract
BACKGROUND The standard treatment for colorectal cancer consists of surgery and chemotherapy, which can be combined to improve outcomes. Immune checkpoint inhibitors (ICI) are a significant advancement in the standard treatment of metastatic, unresectable colorectal cancer with deficient mismatch repair (dMMR). However, limited data are available about the use of ICI in the neoadjuvant and conversion settings. Here, we present two cases treated with ICI. CASE PRESENTATION Case 1: A 75-year-old male with a large, borderline resectable rectal cancer diagnosed as cT4bN1bM0 who underwent neoadjuvant chemotherapy, followed by combination ICI consisting of ipilimumab and nivolumab. After four courses of ICI, the tumor significantly shrank, but positron emission tomography still showed a positive result and R0 resection was performed. Pathological analysis revealed no residual cancer cells. The patient has been monitored without adjuvant chemotherapy, and no recurrences have occurred after one year. Case 2: A 60-year-old male with locally advanced sigmoid colon cancer who received neoadjuvant treatment with pembrolizumab. The tumor partially shrank after three courses, and continued pembrolizumab monotherapy resulted in further tumor shrinkage which still showed positive positron emission tomography. Curative sigmoidectomy with partial resection of the ileum and bladder was performed, and the pathological outcome was pCR. There was no viable tumor in the specimen. The patient has been monitored without adjuvant chemotherapy for six months, and no recurrence has been observed. CONCLUSIONS The present study reports two cases, including a large, borderline resectable rectal cancer after failure of chemotherapy followed by combination treatment with nivolumab and ipilimumab and one case of sigmoid colon cancer after pembrolizumab treatment, which resulted in pathological complete response. However, it remains unknown whether ICI therapy can replace surgery or diminish the optimal extent of resection, or whether adjuvant chemotherapy is needed after surgery in the case of achieving pCR after ICI therapy. Overall, this case report suggests that ICI before colorectal surgery can be effective and potentially a 'watch-and-wait" strategy could be used for cases in which ICI is effective.
Collapse
Affiliation(s)
- Daigaku Nakamura
- Department of Pharmacy, Nagoya City University Hospital, Kawasumi 1, Mizuho-Cho, Mizuho-Ku, Nagoya, 467-8602, Japan
| | - Takeshi Yanagita
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya City, Aichi, 467-8601, Japan.
| | - Yoshiaki Fujii
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya City, Aichi, 467-8601, Japan
| | - Kaori Watanabe
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya City, Aichi, 467-8601, Japan
| | - Takuya Suzuki
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya City, Aichi, 467-8601, Japan
| | - Hajime Ushigome
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya City, Aichi, 467-8601, Japan
| | - Ruriko Nishigaki
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Kawasumi 1, Mizuho-Cho, Mizuho-Ku, Nagoya, 467-8601, Japan
| | - Naomi Sugimura
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Kawasumi 1, Mizuho-Cho, Mizuho-Ku, Nagoya, 467-8601, Japan
| | - Mamoru Tanaka
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Kawasumi 1, Mizuho-Cho, Mizuho-Ku, Nagoya, 467-8601, Japan
| | - Ryo Ogawa
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya City, Aichi, 467-8601, Japan
| | - Hiroki Takahashi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya City, Aichi, 467-8601, Japan
| | - Takaya Shimura
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Kawasumi 1, Mizuho-Cho, Mizuho-Ku, Nagoya, 467-8601, Japan
| | - Yuji Hotta
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Kawasumi 1, Mizuho-Cho, Mizuho-Ku, Nagoya, 467-8601, Japan
| | - Yoichi Matsuo
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya City, Aichi, 467-8601, Japan
| | - Masahiro Kondo
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Kawasumi 1, Mizuho-Cho, Mizuho-Ku, Nagoya, 467-8601, Japan
| | - Yoko Furukawa-Hibi
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Kawasumi 1, Mizuho-Cho, Mizuho-Ku, Nagoya, 467-8601, Japan
| | - Shuji Takiguchi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya City, Aichi, 467-8601, Japan
| |
Collapse
|
50
|
Hashimoto T, Nakamura Y, Oki E, Kobayashi S, Yuda J, Shibuki T, Bando H, Yoshino T. Bridging horizons beyond CIRCULATE-Japan: a new paradigm in molecular residual disease detection via whole genome sequencing-based circulating tumor DNA assay. Int J Clin Oncol 2024; 29:495-511. [PMID: 38551727 PMCID: PMC11043144 DOI: 10.1007/s10147-024-02493-4] [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: 01/28/2024] [Accepted: 02/16/2024] [Indexed: 04/26/2024]
Abstract
Circulating tumor DNA (ctDNA) is the fraction of cell-free DNA in patient blood that originates from a tumor. Advances in DNA sequencing technologies and our understanding of the molecular biology of tumors have increased interest in exploiting ctDNA to facilitate detection of molecular residual disease (MRD). Analysis of ctDNA as a promising MRD biomarker of solid malignancies has a central role in precision medicine initiatives exemplified by our CIRCULATE-Japan project involving patients with resectable colorectal cancer. Notably, the project underscores the prognostic significance of the ctDNA status at 4 weeks post-surgery and its correlation to adjuvant therapy efficacy at interim analysis. This substantiates the hypothesis that MRD is a critical prognostic indicator of relapse in patients with colorectal cancer. Despite remarkable advancements, challenges endure, primarily attributable to the exceedingly low ctDNA concentration in peripheral blood, particularly in scenarios involving low tumor shedding and the intrinsic error rates of current sequencing technologies. These complications necessitate more sensitive and sophisticated assays to verify the clinical utility of MRD across all solid tumors. Whole genome sequencing (WGS)-based tumor-informed MRD assays have recently demonstrated the ability to detect ctDNA in the parts-per-million range. This review delineates the current landscape of MRD assays, highlighting WGS-based approaches as the forefront technique in ctDNA analysis. Additionally, it introduces our upcoming endeavor, WGS-based pan-cancer MRD detection via ctDNA, in our forthcoming project, SCRUM-Japan MONSTAR-SCREEN-3.
Collapse
Affiliation(s)
- Tadayoshi Hashimoto
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Yoshiaki Nakamura
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shin Kobayashi
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Junichiro Yuda
- Department of Hematology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Taro Shibuki
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hideaki Bando
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan.
| |
Collapse
|