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
|
Kojitani Y, Takeda M. Current Status of Precision Medicine in Colorectal Cancer in Japan. Int J Mol Sci 2025; 26:5029. [PMID: 40507837 PMCID: PMC12155534 DOI: 10.3390/ijms26115029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/25/2025] [Accepted: 05/21/2025] [Indexed: 06/16/2025] Open
Abstract
Colorectal cancer (CRC) remains a major health burden in Japan, with precision medicine playing an increasingly critical role in treatment optimization. Key biomarkers, including RAS, BRAF, microsatellite instability/mismatch repair, and human epidermal growth factor receptor 2, can be used as a guide for molecularly targeted therapies and immunotherapy. Advances in molecular diagnostics, including comprehensive genomic profiling, have enabled more precise treatment selection such as RET and NTRK fusions. Nationwide initiatives, such as c-CAT and SCRUM-Japan, can leverage real-world data to refine clinical strategies. Recent developments in circulating tumor DNA analysis have led to novel approaches for minimal residual disease monitoring, as demonstrated by the CIRCULATE-Japan GALAXY study. However, certain challenges persist, including the time required for genetic testing, the limited availability of targeted therapies, and disparities in access to molecular tumor boards. This review summarizes the current landscape of precision medicine in CRC in Japan, emphasizing key biomarkers, genetic testing strategies, targeted therapies, and emerging technologies. Future research should focus on expanding clinical trial access, accelerating drug approvals, and integrating real-world data into clinical practice to further advance precision medicine.
Collapse
Affiliation(s)
| | - Masayuki Takeda
- Department of Cancer Genomics and Medical Oncology, Nara Medical University, 840 Shijo-Cho, Kashihara 634-8521, Nara, Japan;
| |
Collapse
|
3
|
Liang W, Zhang Y, Li J, Ji C, Jiang X. Her-2 nanobody modified cisplatin nanoparticles for precise chemotherapy of colon cancer. J Microencapsul 2025; 42:230-238. [PMID: 39976420 DOI: 10.1080/02652048.2025.2467060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 02/10/2025] [Indexed: 02/21/2025]
Abstract
Construct a Her-2 nanobody modified nanoplatform as a versatile carrier of cisplatin and evaluate its anti-tumour effects. Size, morphology, cellular uptake, in vitro release, cell viability, bio-distribution and antitumor efficacy were respectively measured by dynamic light scattering, transmission electron microscopy, confocal microscopy, HPLC, MTT assay, ICP-Mass and tumour volume. Nb-CDDP NPs was prepared with average diameter 60.4 ± 8.4 nm, PDI 0.2 ± 0.02, Zeta potential -35.74 mV, entrapment efficiency 89.5%±0.8% and drug loading 28.7%±1.3% (w/w). From which cisplatin could release more rapidly in acidic solution. NPs could be easily phagocytised and exhibited stronger cytotoxic effect in HCT-116 cells with IC50 1.46 ± 0.019 μg/mL. The concentration of Nb-CDDP NPs in tumour and its inhibition ratio on tumour volume were both higher than without Nb modification, with hardly any influence on body weight. This cisplatin nanoplatform exhibits exceptional properties and high targeting anti-tumour efficacy in colon cancer cells and mice, which maybe provide a promising strategy for precise chemotherapy.
Collapse
Affiliation(s)
- Weina Liang
- Yantai Food and Drug Inspection and Testing Center, Yantai, China
| | - Yan Zhang
- Yantai Food and Drug Inspection and Testing Center, Yantai, China
| | - Jianpeng Li
- Yantai Food and Drug Inspection and Testing Center, Yantai, China
| | - Chenglin Ji
- Yantai Food and Drug Inspection and Testing Center, Yantai, China
| | - Xuexin Jiang
- Yantai Food and Drug Inspection and Testing Center, Yantai, China
| |
Collapse
|
4
|
Pilati C, Soulabaille A, Gallois C, Blons H, Cayre A, Sroussi M, Le Corre D, Mouillet-Richard S, Mulot C, Le Malicot K, De Reynies A, Bachet JB, Borg C, Di Fiore F, Guimbaud R, Bennouna J, André T, Taieb J, Penault-Llorca F, Laurent-Puig P. ERBB2 Comprehensive Profiling and Prognostication in Stage III Colon Cancer: Findings From PETACC8 and IDEA-France Cohorts. Gastroenterology 2025; 168:714-724.e4. [PMID: 39612956 DOI: 10.1053/j.gastro.2024.10.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/30/2024] [Accepted: 10/22/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND & AIMS ERBB2 pathway activation, through amplification or activating mutations, represents a new target for colon cancer (CC) treatment. Molecular methods were compared with the gold standard for assessing ERBB2 status, and the prognostic value of ERBB2 amplification, mutations, and expression was determined using data from 2 phase 3 trials involving nearly 3000 patients with stage III CC. METHODS In the PETACC8 trial, immunohistochemistry and fluorescence in situ hybridization, DNA, and RNA analysis were performed on 1813, 1719, and 1733 samples, respectively. In the IDEA-France trial, DNA and RNA sequencing was performed on 1129 and 1263 samples. The breast cancer SCAN-B cohort (N = 3409) served as an external reference. A new molecular ERBB2-amplified status was defined using ERBB2 next-generation sequencing score, RNA sequencing expression, and clustering based on ERBB2 neighboring gene expression. Concordance between diagnostic techniques and the association between time to recurrence (TTR) and ERBB2-status were evaluated. RESULTS The prevalence of the molecular ERBB2-amplified group was 1.85% in PETACC8 and 1.5% in IDEA-France, with a concordance of 0.81 (95% CI, 0.70-0.92) with the gold standard immunohistochemistry and fluorescence in situ hybridization method in PETACC8. A nonlinear relationship was observed between TTR and ERBB2 expression, with extreme groups showing a less favorable prognosis (P < .0001) in both colon and breast cancers. Patients with molecular ERBB2-amplified status or mutations had the poorest prognosis, followed by low-expression and intermediate-expression groups (3-year TTR of 67.0%, 71.2%, and 77.9%, respectively). In multivariate analysis, the low-expression group had a significantly shorter TTR (hazard ratio, 1.28; 95% CI, 1.07-1.52). CONCLUSIONS The molecular definition of ERBB2 status could represent a cost-effective alternative in stage III CC. ERBB2 alterations and low RNA expression significantly reduced TTR, highlighting the complex role of ERBB2.
Collapse
Affiliation(s)
- Camilla Pilati
- Centre de Recherche des Cordeliers, Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Université Paris Cité, Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France.
| | - Audrey Soulabaille
- Centre de Recherche des Cordeliers, Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Université Paris Cité, Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France
| | - Claire Gallois
- Centre de Recherche des Cordeliers, Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Université Paris Cité, Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France; Institut du Cancer Paris, Cancer Research for Personalized Medicine, Assistance Publique-Hôpitaux de Paris Centre, Hôpital Européen Georges Pompidou, Paris, France
| | - Hélène Blons
- Centre de Recherche des Cordeliers, Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Université Paris Cité, Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France; Institut du Cancer Paris, Cancer Research for Personalized Medicine, Assistance Publique-Hôpitaux de Paris Centre, Hôpital Européen Georges Pompidou, Paris, France
| | - Anne Cayre
- Department of Pathology, Centre Jean Perrin, UNICANCER, Institut National de la Santé et de la Recherche Médical, UMR 1240, Imagerie Moléculaire et Stratégies Théranostiques, University Clermont Auvergne, Clermont-Ferrand, France
| | - Marine Sroussi
- Centre de Recherche des Cordeliers, Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Université Paris Cité, Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France; Institut Chimie Biologie Innovation, Laboratoire de BioChimie, École Supérieure de Physique et de Chimie Industriel, UMR 8231, Le Centre National de la Recherche Scientifique, Paris Sciences et Lettres University, Paris, France
| | - Delphine Le Corre
- Centre de Recherche des Cordeliers, Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Université Paris Cité, Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France
| | - Sophie Mouillet-Richard
- Centre de Recherche des Cordeliers, Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Université Paris Cité, Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France
| | - Claire Mulot
- Centre de Recherche des Cordeliers, Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Université Paris Cité, Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France
| | - Karine Le Malicot
- Fédération Francophone de Cancérologie Digestive, EPICAD Institut National de la Santé et de la Recherche Médical Lipides Nutrition Cancer, UMR 1231, University of Burgundy and Franche Comté, Dijon, France
| | - Aurélien De Reynies
- Centre de Recherche des Cordeliers, Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Université Paris Cité, Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France
| | - Jean-Baptiste Bachet
- Centre de Recherche des Cordeliers, Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Université Paris Cité, Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France; Department of Hepato-Gastroenterology, Groupe Hospitalier Pitié Salpêtrière, Paris, France; Association des Gastroentérologues Oncologues, Paris, France
| | - Christophe Borg
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
| | - Frédéric Di Fiore
- Hepatogastroenterology Department, Centre Hospitalier Universitaire Rouen, University of Rouen Normandy, INSERM 1245, Institut de Recherche en Oncologie Group, Normandie University, Rouen, France
| | - Rosine Guimbaud
- Digestive Medical Oncology, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Jaafar Bennouna
- Thoracic Oncology Unit, University Hospital of Nantes and Institut National de la Santé et de la Recherche Médical, Regional Center for Research in Cancerology and Immunology, Nantes, France
| | - Thierry André
- Sorbonne Université and Hôpital Saint Antoine, INSERM 938, Site de Recherche Intégrée sur le Cancer Curamus, Paris, France
| | - Julien Taieb
- Centre de Recherche des Cordeliers, Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Université Paris Cité, Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France; Institut du Cancer Paris, Cancer Research for Personalized Medicine, Assistance Publique-Hôpitaux de Paris Centre, Hôpital Européen Georges Pompidou, Paris, France; Association des Gastroentérologues Oncologues, Paris, France
| | - Frédérique Penault-Llorca
- Department of Pathology, Centre Jean Perrin, UNICANCER, Institut National de la Santé et de la Recherche Médical, UMR 1240, Imagerie Moléculaire et Stratégies Théranostiques, University Clermont Auvergne, Clermont-Ferrand, France
| | - Pierre Laurent-Puig
- Centre de Recherche des Cordeliers, Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Université Paris Cité, Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France; Institut du Cancer Paris, Cancer Research for Personalized Medicine, Assistance Publique-Hôpitaux de Paris Centre, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
5
|
Pang H, Chen X, Yan M, Sun H. Dual anti-HER2 therapy combined with chemotherapy as a novel neoadjuvant treatment option for locally advanced HER2-positive and microsatellite stable colon cancer. PRECISION CLINICAL MEDICINE 2025; 8:pbae033. [PMID: 39790478 PMCID: PMC11715517 DOI: 10.1093/pcmedi/pbae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/03/2024] [Accepted: 12/09/2024] [Indexed: 01/12/2025] Open
Affiliation(s)
- Huayang Pang
- Department of Gastrointestinal Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Xiufeng Chen
- Department of Gastrointestinal Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Menghua Yan
- Department of Gastrointestinal Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Hao Sun
- Department of Gastrointestinal Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| |
Collapse
|
6
|
Evans MG, Krause HB, Xiu J, Elliott A, Lou E, Ghani H, Yantiss RK, Garcia-Buitrago M, Matsubara Y, Nakamura Y, Shia J, Yaeger R, Radovich M, Bryant DA, Oberley MJ, Hechtman JF. Evidence for Unified Assessment Criteria of HER2 Immunohistochemistry in Colorectal Carcinoma. Mod Pathol 2025; 38:100654. [PMID: 39522645 DOI: 10.1016/j.modpat.2024.100654] [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: 05/03/2024] [Revised: 10/22/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Human epidermal growth factor receptor-2 (HER2) expression is an important biomarker for the management of RAS wild-type metastatic colorectal carcinoma (CRC). Immunohistochemistry (IHC) with reflex in situ hybridization (ISH) is accepted as a standard method of assessment, yet there are currently the following 2 sets of criteria used to interpret results: the HER2 Amplification for Colorectal Cancer Enhanced Stratification (HERACLES) criteria and the MyPathway criteria. The HER2 Amplification for Colorectal Cancer Enhanced Stratification criteria require ISH confirmation when IHC staining is 3+ in 10% to 49% of cells, whereas the MyPathway criteria mirror those for gastric HER2 assessment and do not recommend ISH confirmation in the previously referenced scenario. We aimed to assess the prevalence of HER2 3+ heterogeneity and its association with ERBB2 copy number amplification to evaluate the necessity of ISH testing when IHC staining is 3+ in <50% of cells. Next-generation sequencing of DNA (592-gene panel or whole exome sequencing) was performed for 13,208 CRC tumors submitted to Caris Life Sciences. HER2 (4B5) expression was tested using IHC. A subset of tumors was tested for ERBB2 amplification via chromogenic ISH and/or via next-generation sequencing (copy number amplification). χ2 tests or Fisher exact tests were applied where appropriate, with P values adjusted for multiple comparisons (P < .05). Of 13,208 CRCs with HER2 IHC, 87.4% (11,541/13,208) were negative for HER2 expression (≤3+ intensity and <10% tumor-cell staining) and 11.2% (1473/13,208) demonstrated at least low HER2 expression (1 to 2+ and ≥10%). Only 1.5% (194/13,208) of all tested tumors were either positive or heterogeneously positive for HER2 overexpression (3+ and ≥10%). Of these, 14% (28/194) had heterogenous HER2 overexpression (3+ staining of 10%-49% of cells). Among 22 HER2-positive/heterogenous cases with successful ISH testing, 100% (22/22) demonstrated amplification via ISH. Because the classification of tumors as HER2-positive/heterogenous using IHC correlated very closely with ISH positivity, our results suggest that ISH is likely unnecessary for CRCs with 3+ HER2 overexpression in 10% to 49% of neoplastic cells.
Collapse
Affiliation(s)
| | | | | | | | - Emil Lou
- University of Minnesota Masonic Cancer Center, Minneapolis, Minnesota
| | | | | | | | - Yuki Matsubara
- National Cancer Center Hospital East, Kashiwa-shi Chiba, Japan
| | | | - Jinru Shia
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Rona Yaeger
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | | | | | | |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Battaglin F, Ou FS, Qu X, Hochster HS, Niedzwiecki D, Goldberg RM, Mayer RJ, Ashouri K, Zemla TJ, Blanke CD, Venook AP, Kabbarah O, Lenz HJ, Innocenti F. HER2 Gene Expression Levels Are Predictive and Prognostic in Patients With Metastatic Colorectal Cancer Enrolled in CALGB/SWOG 80405. J Clin Oncol 2024; 42:1890-1902. [PMID: 38457761 PMCID: PMC11240881 DOI: 10.1200/jco.23.01507] [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: 07/16/2023] [Revised: 12/18/2023] [Accepted: 01/04/2024] [Indexed: 03/10/2024] Open
Abstract
PURPOSE The phase III Cancer and Leukemia Group B (CALGB)/SWOG 80405 trial found no difference in overall survival (OS) in patients with metastatic colorectal cancer receiving first-line chemotherapy in combination with either bevacizumab or cetuximab. We investigated the potential prognostic and predictive value of HER2 amplification and gene expression using next-generation sequencing (NGS) and NanoString data. PATIENTS AND METHODS Primary tumor DNA from 559 patients was profiled for HER2 amplification by NGS (FoundationOne CDx). Tumor tissue from 925 patients was tested for NanoString gene expression using an 800-gene panel. OS and progression-free survival (PFS) were the time-to-event end points. RESULTS High HER2 expression (dichotomized at median) was associated with longer PFS (11.6 v 10 months, P = .012) and OS (32 v 25.3 months, P = .033), independent of treatment. An OS benefit for cetuximab versus bevacizumab was observed in the high HER2 expression group (P = .02), whereas a worse PFS for cetuximab was seen in the low-expression group (P = .019). When modeled as a continuous variable, increased HER2 expression was associated with longer OS (hazard ratio [HR], 0.83 [95% CI, 0.75 to 0.93]; adjusted P = .0007) and PFS (HR, 0.82 [95% CI, 0.74 to 0.91]; adjusted P = .0002), reaching a plateau effect after the median. In patients with HER2 expression lower than median, treatment with cetuximab was associated with worse PFS (HR, 1.38 [95% CI, 1.12 to 1.71]; adjusted P = .0027) and OS (HR, 1.28 [95% CI, 1.02 to 1.59]; adjusted P = .03) compared with that with bevacizumab. A significant interaction between HER2 expression and the treatment arm was observed for OS (Pintx = .017), PFS (Pintx = .048), and objective response rate (Pintx = .001). CONCLUSION HER2 gene expression was prognostic and predictive in CALGB/SWOG 80405. HER2 tumor expression may inform treatment selection for patients with low HER2 favoring bevacizumab- versus cetuximab-based therapies.
Collapse
Affiliation(s)
- Francesca Battaglin
- University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA
| | - Fang-Shu Ou
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN
| | | | | | | | | | | | - Karam Ashouri
- University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA
| | - Tyler J. Zemla
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN
| | | | - Alan P. Venook
- University of California, San Francisco, San Francisco, CA
| | | | - Heinz-Josef Lenz
- University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA
| | | |
Collapse
|
10
|
Chen N, He L, Zou Q, Deng H. HER2 targeted therapy in colorectal Cancer: Current landscape and future directions. Biochem Pharmacol 2024; 223:116101. [PMID: 38442793 DOI: 10.1016/j.bcp.2024.116101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 02/11/2024] [Accepted: 02/27/2024] [Indexed: 03/07/2024]
Abstract
Colorectal cancer (CRC) is one of the most common causes of tumor-related deaths globally. Despite recent improvements in the comprehensive therapy of malignancy, metastatic CRC continues to have a poor prognosis. Human epidermal growth factor receptor 2 (HER2) is an established oncogenic driver, which is successfully targeted for breast and gastric cancers. Approximately 5% of CRC patients carry somatic HER2 mutations or gene amplification. In 2019, the U.S. Food and Drug Administration have approved trastuzumab and pertuzumab in combination with chemotherapy for the treatment of HER2-positive metastatic CRC. This approval marked a significant milestone in the treatment of CRC, as HER2-positive patients now have access to targeted therapies that can improve their outcomes. Yet, assessment for HER2 overexpression/ amplification in CRC has not been standardized. The resistance mechanisms to anti-HER2 therapy have been not clearly investigated in CRC. Although many unknowns remain, an improved understanding of these anti-HER2 agents will be essential for advanced CRC. In this review, we provide an overview of the role of HER2 in CRC as an oncogenic driver, a prognostic and predictive biomarker, and a clinically actionable target, as well as the current progress and challenges in the field.
Collapse
Affiliation(s)
- Na Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, 610041, China; Center of Science and Research, Chengdu Medical College, Chengdu, 610500, China
| | - Ling He
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Qiang Zou
- Center of Science and Research, Chengdu Medical College, Chengdu, 610500, China.
| | - Hongxin Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
11
|
Odintsov I, Makarem M, Nishino M, Bachert SE, Zhang T, LoPiccolo J, Paweletz CP, Gokhale PC, Ivanova E, Saldanha A, Rudin CM, Lockwood WW, Ladanyi M, Somwar R, Jänne PA, Sholl LM. Prevalence and Therapeutic Targeting of High-Level ERBB2 Amplification in NSCLC. J Thorac Oncol 2024; 19:732-748. [PMID: 38154514 PMCID: PMC11957432 DOI: 10.1016/j.jtho.2023.12.019] [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: 10/21/2023] [Revised: 12/12/2023] [Accepted: 12/16/2023] [Indexed: 12/30/2023]
Abstract
INTRODUCTION ERBB2 amplification in lung cancer remains poorly characterized. HER2 (encoded by ERBB2) is a transmembrane tyrosine kinase capable of ligand-independent dimerization and signaling when overexpressed, and a common cause of HER2 overexpression is ERBB2 amplification. Here, we evaluated the clinicopathologic and genomic characteristics of ERBB2-amplified NSCLC and explored a HER2 antibody-drug conjugate (ADC) therapeutic strategy. METHODS Our institutional next-generation DNA sequencing data (OncoPanel) from 5769 NSCLC samples (5075 patients) were queried for cases having high-level ERBB2 amplification (≥6 copies). Clinical and demographic characteristics were extracted from the electronic medical records. Efficacy of the pan-ERBB inhibitor afatinib or HER2 ADCs (trastuzumab deruxtecan and trastuzumab emtansine) was evaluated in NSCLC preclinical models and patients with ERBB2 amplification. RESULTS High-level ERBB2 amplification was identified in 0.9% of lung adenocarcinomas and reliably predicted overexpression of HER2. ERBB2 amplification events are detected in two distinct clinicopathologic and genomic subsets of NSCLC: as the sole mitogenic driver in tumors arising in patients with a smoking history or as a concomitant alteration with other mitogenic drivers in patients with a light or never smoking history. We further reveal that trastuzumab deruxtecan is effective therapy in in vitro and in vivo preclinical models of NSCLC harboring ERBB2 amplification and report two cases of clinical activity of an anti-HER2 ADC in patients who acquired ERBB2 amplification after previous targeted therapy. CONCLUSIONS High-level ERBB2 amplification reliably predicts HER2 overexpression in patients with NSCLC, and HER2 ADC is effective therapy in this population.
Collapse
Affiliation(s)
- Igor Odintsov
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Maisam Makarem
- Lowe Center for Thoracic Oncology and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mizuki Nishino
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sara Emily Bachert
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky
| | - Tom Zhang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; School of Medicine, New York Medical College, Valhalla, New York
| | - Jaclyn LoPiccolo
- Lowe Center for Thoracic Oncology and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Cloud P Paweletz
- Lowe Center for Thoracic Oncology and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Prafulla C Gokhale
- Lowe Center for Thoracic Oncology and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Elena Ivanova
- Lowe Center for Thoracic Oncology and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Aisha Saldanha
- Lowe Center for Thoracic Oncology and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Charles M Rudin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - William W Lockwood
- Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Marc Ladanyi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Romel Somwar
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Pasi A Jänne
- Lowe Center for Thoracic Oncology and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Lynette M Sholl
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
12
|
Robinson HR, Messersmith WA, Lentz RW. HER2-Positive Metastatic Colorectal Cancer. Curr Treat Options Oncol 2024; 25:585-604. [PMID: 38539034 DOI: 10.1007/s11864-024-01183-7] [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] [Accepted: 01/17/2024] [Indexed: 04/24/2024]
Abstract
OPINION STATEMENT Targeted treatment strategies are available for human epidermal growth factor receptor 2 (HER2)-positive (amplified and/or overexpressed) metastatic colorectal cancer (mCRC), and HER2 testing is indicated in patients with mCRC. At present, standard of care first-line treatment for those with HER2-positive mCRC remains chemotherapy in combination with epidermal growth factor receptor (EGFR) inhibitors or bevacizumab, depending on RAS/BRAF mutational status and tumor sidedness. HER2-targeted agents should be considered for those with RAS/BRAF wild-type disease in subsequent-line treatment and in first-line treatment for patients not appropriate for intensive therapy. While the choice of anti-HER2 therapy is empiric given lack of head-to-head comparisons, the combination of trastuzumab plus tucatinib has received FDA accelerated approval for use in this setting and is generally the authors' preference. Trastuzumab plus lapatinib, trastuzumab plus pertuzumab, and trastuzumab deruxtecan (T-DXd) also have evidence of efficacy in this setting. As T-DXd has demonstrated activity following treatment with other HER2-targeted regimens and carries an increased risk of high-grade toxicities, the authors favor reserving it for use after progression on prior anti-HER2 therapy. HER2-targeted therapies that inhibit signal transduction appear to have limited activity in those with RAS mutations, including trastuzumab-containing regimens. However, the antibody drug conjugate T-DXd has some data showing efficacy in this setting, and the authors would consider T-DXd in subsequent-line therapy for HER2-positive, RAS-mutated mCRC. Several areas of uncertainty remain regarding how to best utilize HER2-targeted therapies in mCRC. These include the optimal sequence of anti-HER2 therapies with chemotherapy and anti-EGFR therapies, the optimal combination partners for anti-HER2 therapies, and the incorporation of predictive biomarkers to guide use of anti-HER2 therapies. Results of ongoing studies may thus alter the treatment paradigm above in the coming years.
Collapse
Affiliation(s)
- Hannah R Robinson
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, MS 8117, 12801 E. 17Th Avenue, Aurora, CO, 80045, USA
| | - Wells A Messersmith
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, MS 8117, 12801 E. 17Th Avenue, Aurora, CO, 80045, USA
| | - Robert W Lentz
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, MS 8117, 12801 E. 17Th Avenue, Aurora, CO, 80045, USA.
| |
Collapse
|
13
|
Singh H, Sahgal P, Kapner K, Corsello SM, Gupta H, Gujrathi R, Li YY, Cherniack AD, El Alam R, Kerfoot J, Andrews E, Lee A, Nambiar C, Hannigan AM, Remland J, Brais L, Leahy ME, Rubinson DA, Schlechter BL, Meyerson M, Kuang Y, Paweletz CP, Lee JK, Quintanilha JC, Aguirre AJ, Perez KJ, Huffman BM, Rossi H, Abrams TA, Kabraji S, Trusolino L, Bertotti A, Sicinska ET, Parikh AR, Wolpin BM, Schrock AB, Giannakis M, Ng K, Meyerhardt JA, Hornick JL, Sethi NS, Cleary JM. RAS/RAF Comutation and ERBB2 Copy Number Modulates HER2 Heterogeneity and Responsiveness to HER2-directed Therapy in Colorectal Cancer. Clin Cancer Res 2024; 30:1669-1684. [PMID: 38345769 PMCID: PMC11018475 DOI: 10.1158/1078-0432.ccr-23-2581] [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: 08/24/2023] [Revised: 11/17/2023] [Accepted: 02/06/2024] [Indexed: 04/16/2024]
Abstract
PURPOSE ERBB2-amplified colorectal cancer is a distinct molecular subtype with expanding treatments. Implications of concurrent oncogenic RAS/RAF alterations are not known. EXPERIMENTAL DESIGN Dana-Farber and Foundation Medicine Inc. Colorectal cancer cohorts with genomic profiling were used to identify ERBB2-amplified cases [Dana-Farber, n = 47/2,729 (1.7%); FMI, n = 1857/49,839 (3.7%)]. Outcomes of patients receiving HER2-directed therapies are reported (Dana-Farber, n = 9; Flatiron Health-Foundation Medicine clinicogenomic database, FH-FMI CGDB, n = 38). Multisite HER2 IHC and genomic profiling were performed to understand HER2 intratumoral and interlesional heterogeneity. The impact of concurrent RAS comutations on the effectiveness of HER2-directed therapies were studied in isogenic colorectal cancer cell lines and xenografts. RESULTS ERBB2 amplifications are enriched in left-sided colorectal cancer. Twenty percent of ERBB2-amplified colorectal cancers have co-occurring oncogenic RAS/RAF alterations. While RAS/RAF WT colorectal cancers typically have clonal ERBB2 amplification, colorectal cancers with co-occurring RAS/RAF alterations have lower level ERRB2 amplification, higher intratumoral heterogeneity, and interlesional ERBB2 discordance. These distinct genomic patterns lead to differential responsiveness and patterns of resistance to HER2-directed therapy. ERBB2-amplified colorectal cancer with RAS/RAF alterations are resistant to trastuzumab-based combinations, such as trastuzumab/tucatinib, but retain sensitivity to trastuzumab deruxtecan in in vitro and murine models. Trastuzumab deruxtecan shows clinical efficacy in cases with high-level ERBB2-amplified RAS/RAF coaltered colorectal cancer. CONCLUSIONS Co-occurring RAS/RAF alterations define a unique subtype of ERBB2-amplified colorectal cancer that has increased intratumoral heterogeneity, interlesional discordance, and resistance to trastuzumab-based combinations. Further examination of trastuzumab deruxtecan in this previously understudied cohort of ERBB2-amplified colorectal cancer is warranted.
Collapse
Affiliation(s)
- Harshabad Singh
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | - Pranshu Sahgal
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
- Broad Institute of Harvard and MIT, Cambridge MA, USA
| | - Kevin Kapner
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | | | - Hersh Gupta
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
- Broad Institute of Harvard and MIT, Cambridge MA, USA
| | - Rahul Gujrathi
- Department of Radiology, Boston Medical Center and Boston University, Boston, MA USA
| | - Yvonne Y. Li
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
- Broad Institute of Harvard and MIT, Cambridge MA, USA
| | - Andrew D. Cherniack
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
- Broad Institute of Harvard and MIT, Cambridge MA, USA
| | - Raquelle El Alam
- Department of Radiology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | - Joseph Kerfoot
- Department of Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | - Elizabeth Andrews
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | - Annette Lee
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | - Chetan Nambiar
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | - Alison M. Hannigan
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | - Joshua Remland
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | - Lauren Brais
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | - Meghan E. Leahy
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | - Douglas A. Rubinson
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | - Benjamin L. Schlechter
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | - Matthew Meyerson
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
- Broad Institute of Harvard and MIT, Cambridge MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA USA
| | - Yanan Kuang
- Belfer Center for Applied Cancer Science, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA USA
| | - Cloud P. Paweletz
- Belfer Center for Applied Cancer Science, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA USA
| | | | | | - Andrew J. Aguirre
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
- Broad Institute of Harvard and MIT, Cambridge MA, USA
| | - Kimberly J. Perez
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | - Brandon M. Huffman
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | - Humberto Rossi
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | - Thomas A. Abrams
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | - Sheheryar Kabraji
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | - Livio Trusolino
- Candiolo Cancer Institute FPO IRCCS, Candiolo, Torino, Italy
- Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Andrea Bertotti
- Candiolo Cancer Institute FPO IRCCS, Candiolo, Torino, Italy
- Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Ewa T. Sicinska
- Department of Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | - Aparna R. Parikh
- Massachusetts General Hospital Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Brian M. Wolpin
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | | | - Marios Giannakis
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | - Kimmie Ng
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | - Jeffrey A. Meyerhardt
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | - Jason L. Hornick
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Nilay S. Sethi
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | - James M. Cleary
- Dana-Farber Brigham and Women’s Cancer Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| |
Collapse
|
14
|
Zheng-Lin B, Bekaii-Saab TS. Treatment options for HER2-expressing colorectal cancer: updates and recent approvals. Ther Adv Med Oncol 2024; 16:17588359231225037. [PMID: 38249334 PMCID: PMC10798128 DOI: 10.1177/17588359231225037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 12/18/2023] [Indexed: 01/23/2024] Open
Abstract
Colorectal cancer is a leading cause of cancer-related mortality worldwide. Approximately 3-5% of colorectal tumors harbor human epidermal growth factor receptor (HER2) amplification which is associated with a higher incidence of intracranial metastasis and overall worse outcomes. In the setting of refractory metastatic RAS wild-type tumors, evidence supports the use of various HER2-blocking agents, including monoclonal antibodies, tyrosine kinase inhibitors, and novel antibody-drug conjugates. With a number of relatively active agents clinically available and even more in development, it is crucial for clinicians to familiarize themselves with the mechanisms of action, efficacy data, and safety profiles of these treatments. In this review, we aim to summarize key findings from past and ongoing trials with anti-HER2 agents in metastatic colorectal cancer.
Collapse
Affiliation(s)
- Binbin Zheng-Lin
- Division of Hematology and Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Tanios S. Bekaii-Saab
- Division of Hematology and Medical Oncology, Mayo Clinic, 5881 East Mayo Blvd, Phoenix, AZ 85054, USA
| |
Collapse
|
15
|
Buchholz SM, Nause N, König U, Reinecke J, Steuber B, Ammer-Herrmenau C, Reuter-Jessen K, Bohnenberger H, Biggemann L, Braulke F, Neesse A, Ellenrieder V, Ströbel P, Adler M, König A. Time to Deliver on Promises: The Role of ERBB2 Alterations as Treatment Options for Colorectal Cancer Patients in the Era of Precision Oncology. J Pers Med 2023; 13:1701. [PMID: 38138928 PMCID: PMC10745079 DOI: 10.3390/jpm13121701] [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: 10/10/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Receptor tyrosine kinase erythroblastic oncogene B2 (ERBB2), also known as human epidermal growth factor receptor 2 (HER2), represents an oncogenic driver and has been effectively targeted in breast and gastric cancer. Recently, next-generation sequencing (NGS) discovered ERBB2 as a promising therapeutic target in metastatic colorectal cancer (mCRC), where it is altered in 3-5% of patients, but no therapies are currently approved for this use. Herein, we present the experience of a single center in diagnosing actionable genetic ERBB2 alterations using NGS and utilizing the latest therapeutic options. Between October 2019 and December 2022, a total of 107 patients with advanced CRC underwent molecular analysis, revealing actionable ERBB2 mutations in two patients and ERBB2 amplifications in two other patients. These findings correlated with immunohistochemical (IHC) staining. Of these four patients, two were treated with trastuzumab-deruxtecan (T-DXd). We present two exemplary cases of patients with actionable ERBB2 alterations to demonstrate the effectiveness of T-DXd in heavily pretreated ERBB2-positive mCRC patients and the need for early molecular profiling. To fully exploit the potential of this promising treatment, earlier molecular profiling and the initiation of targeted therapies are essential.
Collapse
Affiliation(s)
- Soeren M. Buchholz
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Nelia Nause
- Göttingen Comprehensive Cancer Center (G-CCC), University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Ute König
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Johanna Reinecke
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Benjamin Steuber
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Christoph Ammer-Herrmenau
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Kirsten Reuter-Jessen
- Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Hanibal Bohnenberger
- Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Lorenz Biggemann
- Institute of Diagnostic and Interventional Radiology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Friederike Braulke
- Göttingen Comprehensive Cancer Center (G-CCC), University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Albrecht Neesse
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Volker Ellenrieder
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Philipp Ströbel
- Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Marius Adler
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, 37075 Göttingen, Germany
- Department of Gastroenterology, University Hospital Augsburg, 86156 Augsburg, Germany
| | - Alexander König
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
16
|
Baranov E, Nowak JA. Pathologic Evaluation of Therapeutic Biomarkers in Colorectal Adenocarcinoma. Surg Pathol Clin 2023; 16:635-650. [PMID: 37863556 DOI: 10.1016/j.path.2023.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2023]
Abstract
Molecular testing is an essential component of the pathologic evaluation of colorectal carcinoma providing diagnostic, prognostic, and predictive therapeutic information. Mismatch repair status evaluation is required for all tumors. Advanced and metastatic tumors also require determination of tumor mutational burden, KRAS, NRAS, and BRAF mutation status, ERBB2 amplification status, and NTRK and RET gene rearrangement status to guide therapy. Multiple assays, including immunohistochemistry, microsatellite instability testing, MLH1 promoter methylation, and next-generation sequencing, are typically needed. Pathologists must be aware of these requirements to optimally triage tissue. Advances in colorectal cancer molecular diagnostics will continue to drive refinements in colorectal cancer personalized therapy.
Collapse
Affiliation(s)
- Esther Baranov
- Department of Pathology, Brigham & Women's Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Jonathan A Nowak
- Department of Pathology, Brigham & Women's Hospital, 75 Francis Street, Boston, MA 02115, USA.
| |
Collapse
|
17
|
Neupane N, Thapa S, Bhattarai A, Ahuja K, Schlam I, Mittal A, Tolaney SM, Tarantino P. Opportunities and Challenges for a Histology-Agnostic Utilization of Trastuzumab Deruxtecan. Curr Oncol Rep 2023; 25:1467-1482. [PMID: 37938529 DOI: 10.1007/s11912-023-01469-3] [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] [Accepted: 09/27/2023] [Indexed: 11/09/2023]
Abstract
PURPOSE OF REVIEW This review delves into the prospects and challenges offered by a potential pan-histological utilization of trastuzumab deruxtecan (T-DXd) in patients with advanced solid tumors. RECENT FINDINGS The HER2-targeted antibody-drug conjugate (ADC) T-DXd has shown broad activity across cancer types, with current indications for patients with biomarker-selected breast, gastric, and non-small-cell lung cancer and relevant activity observed in multiple histology-specific trials. Moreover, two recently reported phase 2 trials (DESTINY-Pantumor02 and HERALD) have supported the potential for a pan-cancer utilization of this ADC in patients with advanced cancers expressing HER2 or with HER2 amplifications. By improving the delivery of cytotoxic chemotherapy, ADCs have allowed for meaningful clinical advantages in broad populations of cancer patients, often leading to survival advantages over conventional chemotherapy. Notably, the broad spectrum of activity of certain ADCs has led to the hypothesis of a histology-agnostic utilization based on detecting specific biomarkers, similar to what is already established for certain targeted treatments and immunotherapy. To date, T-DXd has shown the broadest activity across cancer types, with current approvals in breast, gastric, and lung cancer, and relevant antitumor activity observed in a multiplicity of additional cancer types. The optimization of the drug dose, identification of predictive biomarkers, and clarification of mechanisms of resistance will be critical steps in view of a pan-histological expansion in the use of T-DXd.
Collapse
Affiliation(s)
| | - Sangharsha Thapa
- Westchester Medical Center, New York Medical College, New York, NY, USA
| | - Abhinav Bhattarai
- Institute of Medicine, Tribhuvan University, Kathmandu, Nepal
- Medical Database, Irvine, CA, USA
| | - Kriti Ahuja
- Internal Medicine, John H Stroger Hospital of Cook County, Chicago, IL, USA
| | - Ilana Schlam
- Division of Hematology and Oncology, Tufts Medical Center, Boston, MA, USA
- Tufts University, Boston, MA, USA
| | - Abhenil Mittal
- Health Sciences North, Northern Ontario School of Medicine, Sudbury, ON, Canada
| | - Sara M Tolaney
- Breast Oncology Program, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA
- Harvard Medical School, Boston, MA, USA
| | - Paolo Tarantino
- Breast Oncology Program, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA.
- Harvard Medical School, Boston, MA, USA.
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| |
Collapse
|
18
|
Jung J, Kim ST, Ko J, Hong JY, Park JO, Ha SY, Lee J. Clinical Implication of HER2 Aberration in Patients With Metastatic Cancer Using Next-Generation Sequencing: A Pan-Tumor Analysis. JCO Precis Oncol 2023; 7:e2200537. [PMID: 37499191 DOI: 10.1200/po.22.00537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 03/13/2023] [Accepted: 05/22/2023] [Indexed: 07/29/2023] Open
Abstract
PURPOSE Human epidermal growth factor receptor 2 (HER2) protein expression or gene amplification is a significant predictive biomarker for identifying patients with cancer, who may benefit from HER2-targeted therapy. The aim of this study was to survey the proportion of patients who had HER2 aberration and to investigate the correlation between HER2 amplification and HER2 overexpression in immunohistochemistry (IHC) as a real-world data. METHODS We surveyed the incidence of HER2 aberration including mutation (single-nucleotide variant [SNV]), amplification (copy-number variation), and fusion by next-generation sequencing (NGS) in 2,119 patients with cancer from Samsung Medical Center in South Korea. RESULTS Of 2,119 patients with cancer, 189 patients (8.9%) had HER2 aberration in their tumor specimen. Of 189 patients, 113 (5.3%) patients had HER2 amplification, 82 (3.9%) patients had HER2 mutations, and 11 (0.5%) patients had HER2 fusion. Of note, 10 patients (0.5%) had concurrent HER2 amplification and HER2 fusion. In addition, we identified that HER2 protein overexpression was strongly related to HER2 amplification by NGS. Of 74 patients with HER2 amplification only by NGS test, 64 patients (86.5%) had HER2 overexpression by IHC. Of 10 patients with concurrent HER2 amplification and fusion, 80% patients were HER2 overexpression. Among 51 patients with only HER2 mutation (SNV), 9 patients (17.6%) were HER2 (+). Interestingly, almost all patients with colorectal cancer (11 of 12) with HER2 amplification had very strong HER2 overexpression (3+) in their tumor specimen. CONCLUSION In conclusion, we showed that when patients with metastatic cancer receive NGS test, approximately 8.9% have HER2 aberrations in their tumor specimen. Most patients have HER2 amplification, and a small percentage of patients have HER2 fusion. A great majority of patients with HER2 amplification and/or HER2 fusion had HER2 (+) tumor by IHC.
Collapse
Affiliation(s)
- Jaeyun Jung
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea
- Experimental Therapeutics Development Center, Samsung Medical Center, Seoul, South Korea
| | - Seung Tae Kim
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea
| | - Jihoon Ko
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea
| | - Jung Yong Hong
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea
| | - Joon Oh Park
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea
| | - Sang Yun Ha
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jeeyun Lee
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea
| |
Collapse
|
19
|
Liu XT, Kou ZY, Zhang H, Dong J, Zhang JH, Peng YJ, Ma SM, Liang L, Meng XY, Zhou Y, Yang J. Somatic mutation profiling, tumor-infiltrating leukocytes, tertiary lymphoid structures and PD-L1 protein expression in HER2-amplified colorectal cancer. PeerJ 2023; 11:e15261. [PMID: 37151285 PMCID: PMC10162038 DOI: 10.7717/peerj.15261] [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: 01/12/2023] [Accepted: 03/28/2023] [Indexed: 05/09/2023] Open
Abstract
The status of human epidermal growth factor receptor 2 (HER2) for the prognosis in colorectal cancer (CRC) is controversial, and the characteristics of the somatic mutation spectrum, tumor-infiltrating leukocytes, tertiary lymphoid structures and PD-L1 protein are unknown in HER2-amplified colorectal cancer (HACC). In order to explore these characteristics along with their correlation with clinicopathological factors and prognosis in HACC. Samples of 812 CRC patients was collected. After immunohistochemistry (IHC), 59 of 812 were found to be HER2-positive, then 26 of 59 samples were further determined to be HER2 amplification by fluorescence in situ hybridization (FISH). Somatic mutation profiling of HACC was analysed using whole exome sequencing (WES). Multiplex fluorescence immunohistochemistry (mIHC) was used for tumor-infiltrating leukocytes and tertiary lymphoid structures (TLSs), while PD-L1 protein was detected by IHC. Our results indicate that the detection rates of HER2 positivity by IHC and FISH were 7.3% and 3.2% respectively, and HER2 amplification is correlated with distant tumour metastasis. The somatic mutation profiling revealed no differences between HACC and HER2-negative CRC. However, TP 53 strongly correlated with poor prognosis in HACC. Furthermore, tumor-infiltrating T cells and TLSs in the tumor immune microenvironment, as well as PD-L1 expression, were higher in HACC than in HER2-negative controls. However, none of them were associated with the prognosis of HACC. In all, HER2 amplification is correlated with distant metastasis and TP53 gene mutation may be a potential protective mechanism of HACC.
Collapse
Affiliation(s)
- Xiao-Ting Liu
- Department of Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zhi-Yong Kou
- Department of Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Hushan Zhang
- Zhaotong Healthy Vocational College, Zhaotong, Yunnan, China
| | - Jian Dong
- Colorectal Cancer Clinical Research Center, Yunnan Cancer Hospital, Kunming, Yunnan, China
| | - Jian-Hua Zhang
- Department of General Surgery, The Third People’s Hospital of Honghe Prefecture, Honghe, Yunnan, China
| | - You-Jun Peng
- Department of General Surgery, The Third People’s Hospital of Honghe Prefecture, Honghe, Yunnan, China
| | - Shu Min Ma
- Department of General Surgery, The Second People’s Hospital of Qujing, Qujing, Yunnan, China
| | - Lei Liang
- Department of General Surgery, The Third People’s Hospital of Honghe Prefecture, Honghe, Yunnan, China
| | - Xuan-Yu Meng
- Department of Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yuan Zhou
- Department of Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jun Yang
- Department of Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
20
|
Yoshikawa A, Nakamura Y. Molecular Basis of HER2-Targeted Therapy for HER2-Positive Colorectal Cancer. Cancers (Basel) 2022; 15:183. [PMID: 36612185 PMCID: PMC9818808 DOI: 10.3390/cancers15010183] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 12/30/2022] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) amplification has emerged as a biomarker in colorectal cancer (CRC), occurring in 1-4% of metastatic CRC (mCRC). In addition to conventional methods, such as immunohistochemistry and fluorescence in situ hybridization, next-generation sequencing-based tissue or circulating tumor DNA analysis has recently been used to identify HER2 amplification and assess HER2 overexpression. Prospective clinical trials have demonstrated the efficacy of HER2-targeted therapies in HER2-positive mCRC. The TRIUMPH study, a phase II study of dual HER2 antibodies, i.e., pertuzumab plus trastuzumab, demonstrated promising efficacy for patients with HER2-positive mCRC confirmed by tissue-and/or blood-based techniques, which led to the regulatory approval of this combination therapy in Japan. The mechanisms associated with efficacy and resistance have also been explored in translational studies that incorporate liquid biopsy in prospective trials. In particular, HER2 copy number and co-alterations have repeatedly been reported as biomarkers related to efficacy. To improve the therapeutic efficacy of the current strategy, many clinical trials with various HER2-targeted agents are ongoing. This review discusses the molecular basis of HER2-targeted therapeutic strategies for patients with HER2-positive mCRC.
Collapse
Affiliation(s)
- Ayumu Yoshikawa
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa 277-0882, Japan
| | - Yoshiaki Nakamura
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa 277-0882, Japan
- International Research Promotion Office, National Cancer Center Hospital East, Kashiwa 277-0882, Japan
- Translational Research Support Section, National Cancer Center Hospital East, Kashiwa 277-0882, Japan
| |
Collapse
|
21
|
Ivanova M, Venetis K, Guerini-Rocco E, Bottiglieri L, Mastropasqua MG, Garrone O, Fusco N, Ghidini M. HER2 in Metastatic Colorectal Cancer: Pathology, Somatic Alterations, and Perspectives for Novel Therapeutic Schemes. Life (Basel) 2022; 12:1403. [PMID: 36143438 PMCID: PMC9502498 DOI: 10.3390/life12091403] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/02/2022] [Accepted: 09/06/2022] [Indexed: 12/03/2022] Open
Abstract
HER2 is an emerging biomarker in colorectal cancer (CRC). This oncogene plays an essential role in regulating cell proliferation, differentiation, migration, and, more in general, tumorigenesis and tumor progression. The most frequent types of HER2 alterations in CRC include gene amplification and missense mutations in 7-8% of CRC, often being mirrored by HER2 protein overexpression, representing founder events in solid tumors, including CRC. There are currently no approved HER2-targeted therapy guidelines for CRC; however, several studies have shown that HER2 can be effectively targeted in meta-static CRC settings. In this review, we discuss the current knowledge of HER2 testing in CRC and the immediate future perspectives for HER2 targeting in the metastatic setting.
Collapse
Affiliation(s)
- Mariia Ivanova
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Konstantinos Venetis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Elena Guerini-Rocco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Luca Bottiglieri
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Mauro Giuseppe Mastropasqua
- Department of Emergency and Organ Transplantation, School of Medicine, University of Bari “Aldo Moro”, Piazza G Cesare, 11, 70124 Bari, Italy
| | - Ornella Garrone
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Michele Ghidini
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| |
Collapse
|
22
|
Roy-Chowdhuri S, Davies KD, Ritterhouse LL, Snow AN. ERBB2 (HER2) Alterations in Colorectal Cancer. J Mol Diagn 2022; 24:1064-1066. [DOI: 10.1016/j.jmoldx.2022.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/03/2022] [Accepted: 07/19/2022] [Indexed: 10/15/2022] Open
|
23
|
Torres-Jiménez J, Esteban-Villarrubia J, Ferreiro-Monteagudo R. Precision Medicine in Metastatic Colorectal Cancer: Targeting ERBB2 (HER-2) Oncogene. Cancers (Basel) 2022; 14:3718. [PMID: 35954382 PMCID: PMC9367374 DOI: 10.3390/cancers14153718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 02/01/2023] Open
Abstract
Colorectal cancer (CRC) 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. The treatment of metastatic CRC (mCRC) is based on the use of chemotherapy, anti-vascular endothelial growth factor (VEGF), and anti-epidermal growth factor receptor (EGFR) for RAS wild-type tumors. Precision medicine tries to identify molecular alterations that could be treated with targeted therapies. ERBB2 amplification (also known as HER-2) has been identified in 2-3% of patients with mCRC, but there are currently no approved ERBB2-targeted therapies for mCRC. The purpose of this review is to describe the molecular structure of ERBB2, clinical features of these patients, diagnosis of ERBB2 alterations, and the most relevant clinical trials with ERBB2-targeted therapies in mCRC.
Collapse
Affiliation(s)
- Javier Torres-Jiménez
- Medical Oncology Department, MD Anderson Cancer Center Madrid, 28033 Madrid, Spain
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (J.E.-V.); (R.F.-M.)
| | - Jorge Esteban-Villarrubia
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (J.E.-V.); (R.F.-M.)
- Medical Oncology Department, University Hospital 12 de Octubre, 28041 Madrid, Spain
| | - Reyes Ferreiro-Monteagudo
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (J.E.-V.); (R.F.-M.)
| |
Collapse
|
24
|
Ferguson DC, Momeni Boroujeni A, Zheng T, Mohanty AS, Ho AL, Arcila ME, Ross DS, Dogan S. ERBB2 amplification status in 67 salivary duct carcinomas assessed by immunohistochemistry, fluorescence in situ hybridization, and targeted exome sequencing. Mod Pathol 2022; 35:895-902. [PMID: 34963694 PMCID: PMC10363285 DOI: 10.1038/s41379-021-00999-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 11/09/2022]
Abstract
Salivary duct carcinoma (SDC) is an aggressive salivary gland malignancy with poor survival. Approximately 30% SDC harbor HER2 amplification and response to trastuzumab has been reported. However, a systematic approach for HER2 status assessment in this tumor type has not been established. A total of 67 tumor samples were evaluated for HER2 protein overexpression or ERBB2 gene amplification using at least 2 methods: immunohistochemistry (IHC), fluorescence in situ hybridization (FISH), and/or targeted exome next-generation sequencing (NGS). NGS assessed ERBB2 copy number fold change (FC) and total copy number (TCN). HER2 status was first determined by IHC/FISH according to the 2018 ASCO/CAP breast cancer guidelines. FISH results, the "gold standard", were compared with the NGS results. All (15/15) IHC positive, 35% (6/17) equivocal, and no (0/19) IHC negative SDC were HER2 amplified by FISH. HER2 FISH signal/cell showed a good correlation with FC (Spearman correlation: 0.708, R2: 0.501, p < 0.0001) and TCN (Spearman correlation: 0.763, R2: 0.582, p < 0.0001). Receiver operating characteristics curve estimation showed an area under curve (AUC) of 0.975 for ERBB2 FC. FC cutoff of ≥1.8 corresponded to an accuracy of 95.2% for ERBB2 amplification (Youden's index: 0.84, sensitivity: 89.47%, specificity: 100%). FC < 1.3 could be reliably classified as ERBB2 not amplified and FC ≥ 1.3 and <1.8 as equivocal. TCN estimation showed AUC of 0.981. TCN cutoff of >6.0 corresponded to an accuracy of 92% for HER2 amplification (Youden's index: 0.81, sensitivity: 81.2%, specificity: 100%). TCN < 4 could be reliably classified as ERBB2 not amplified and TCN ≥ 4.0 and ≤6.0 as equivocal. FC and TCN were binarized with respective cutoffs of ≥1.8 and ≥6.0 and the proportion of agreement with FISH were 95% and 92%, respectively. The assessment of ERBB2 copy number by NGS is accurate and reliable with FC or TCN nearly equivalent to FISH in identifying HER2 amplified SDC.
Collapse
Affiliation(s)
- Donna C Ferguson
- Departments of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Amir Momeni Boroujeni
- Departments of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Tao Zheng
- Departments of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Abhinita S Mohanty
- Departments of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Alan L Ho
- Departments of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Maria E Arcila
- Departments of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Dara S Ross
- Departments of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Snjezana Dogan
- Departments of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| |
Collapse
|
25
|
Vokes NI, Chambers E, Nguyen T, Coolidge A, Lydon CA, Le X, Sholl L, Heymach JV, Nishino M, Van Allen EM, Jänne PA. Concurrent TP53 Mutations Facilitate Resistance Evolution in EGFR-Mutant Lung Adenocarcinoma. J Thorac Oncol 2022; 17:779-792. [PMID: 35331964 PMCID: PMC10478031 DOI: 10.1016/j.jtho.2022.02.011] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 02/08/2022] [Accepted: 02/15/2022] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Patients with EGFR-mutant NSCLC experience variable duration of benefit on EGFR tyrosine kinase inhibitors. The effect of concurrent genomic alterations on outcome has been incompletely described. METHODS In this retrospective study, targeted next-generation sequencing data were collected from patients with EGFR-mutant lung cancer treated at the Dana-Farber Cancer Institute. Clinical data were collected and correlated with somatic mutation data. Associations between TP53 mutation status, genomic features, and mutational processes were analyzed. RESULTS A total of 269 patients were identified for inclusion in the cohort. Among 185 response-assessable patients with pretreatment specimens, TP53 alterations were the most common event associated with decreased first-line progression-free survival and decreased overall survival, along with DNMT3A, KEAP1, and ASXL1 alterations. Reduced progression-free survival on later-line osimertinib in 33 patients was associated with MET, APC, and ERBB4 alterations. Further investigation of the effect of TP53 alterations revealed an association with worse outcomes even in patients with good initial radiographic response, and faster acquisition of T790M and other resistance mechanisms. TP53-mutated tumors had higher mutational burdens and increased mutagenesis with exposure to therapy and tobacco. Cell cycle alterations were not independently predictive, but portended worse OS in conjunction with TP53 alterations. CONCLUSIONS TP53 alterations associate with faster resistance evolution independent of mechanism in EGFR-mutant NSCLC and may cooperate with other genomic events to mediate acquisition of resistance mutations to EGFR tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Natalie I Vokes
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Emily Chambers
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Tom Nguyen
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Alexis Coolidge
- Department of Internal Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Christine A Lydon
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Xiuning Le
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lynette Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - John V Heymach
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mizuki Nishino
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts; Department of Imaging, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts; Broad Institute of Harvard and Massachusetts Institute of Technology, Boston, Massachusetts
| | - Pasi A Jänne
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| |
Collapse
|
26
|
Karan C, Tan E, Sarfraz H, Knepper TC, Walko CM, Felder S, Kim R, Sahin IH. Human Epidermal Growth Factor Receptor 2-Targeting Approaches for Colorectal Cancer: Clinical Implications of Novel Treatments and Future Therapeutic Avenues. JCO Oncol Pract 2022; 18:545-554. [PMID: 35613416 DOI: 10.1200/op.21.00904] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The treatment paradigm for colorectal cancer (CRC) has changed significantly over the past decade with targeted therapeutics. Human epidermal growth factor receptor 2 (HER2) amplification is seen among 3%-4% of patients with metastatic CRC (mCRC). The biological discovery of HER2 amplification in cancer cells has led to practice-changing drug development for several solid tumors, including breast, gastric, and esophageal cancers. HER2 amplification is now highly actionable in CRC with distinct therapeutic combinations, including the combination of monoclonal antibodies and HER2 receptor-specific tyrosine kinase inhibitors, as well as antibody-drug conjugates, that delivers targeted cytotoxic agents. However, it is essential to define the therapeutic role and sequence of these different combinations, some of which are already part of standard clinical practice. In this review article, we discuss recent clinical studies demonstrating the clinical benefits of each distinct therapeutic approach and their impacts on the current management of HER2-amplified mCRC. We also review ongoing clinical trials targeting the HER2 pathway in mCRC and elaborate on novel therapeutic opportunities in this space that may further define the changing paradigm of HER2-targeted therapy for CRC.
Collapse
Affiliation(s)
- Canan Karan
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Elaine Tan
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Humaira Sarfraz
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Todd C Knepper
- Department of Individualized Cancer Management, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Christine M Walko
- Department of Individualized Cancer Management, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Seth Felder
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Richard Kim
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Ibrahim Halil Sahin
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| |
Collapse
|
27
|
Ahcene Djaballah S, Daniel F, Milani A, Ricagno G, Lonardi S. HER2 in Colorectal Cancer: The Long and Winding Road From Negative Predictive Factor to Positive Actionable Target. Am Soc Clin Oncol Educ Book 2022; 42:1-14. [PMID: 35580290 DOI: 10.1200/edbk_351354] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Human epidermal growth factor receptor 2 (HER2) is a well-known oncogenic driver in different tumors and an approved therapeutic target in breast and gastroesophageal cancer. In metastatic colorectal cancer, only 3% to 5% of patients present with HER2 alterations: somatic mutations and amplifications. HER2 was first assessed as a biomarker of resistance to anti-EGFR therapy; however, in more recent years, its role as a potential actionable target has emerged. In this article, we discuss the predictive and prognostic value of HER2 in metastatic colorectal cancer, its emerging role as an actionable therapeutic target, and its possible future developments.
Collapse
Affiliation(s)
| | - Francesca Daniel
- Medical Oncology Unit 1, Veneto Institute of Oncology IRCCS, Padua, Italy
| | - Anna Milani
- Medical Oncology Unit 3, Veneto Institute of Oncology IRCCS, Padua, Italy.,Department of Surgery, Oncology, and Gastroenterology, University of Padua, Padua, Italy
| | - Gianmarco Ricagno
- Medical Oncology Unit 3, Veneto Institute of Oncology IRCCS, Padua, Italy.,Department of Surgery, Oncology, and Gastroenterology, University of Padua, Padua, Italy
| | - Sara Lonardi
- Medical Oncology Unit 3, Veneto Institute of Oncology IRCCS, Padua, Italy
| |
Collapse
|
28
|
Strickler JH, Yoshino T, Graham RP, Siena S, Bekaii-Saab T. Diagnosis and Treatment of ERBB2-Positive Metastatic Colorectal Cancer: A Review. JAMA Oncol 2022; 8:760-769. [PMID: 35238866 DOI: 10.1001/jamaoncol.2021.8196] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Importance Amplification of ERBB2 (formerly referred to as HER2) is present in nearly 3% of patients with metastatic colorectal cancer overall and 5% of patients with KRAS and NRAS wild-type tumors. Despite the availability of several ERBB2-targeted therapeutic options for patients with ERBB2-positive breast and gastric/gastroesophageal tumors, to date, there are currently no approved therapies for patients with ERBB2-positive metastatic colorectal cancer, although ERBB2-targeted therapies are recommended in National Comprehensive Cancer Network guidelines. Recent evidence indicates that anti-ERBB2 therapeutic strategies are active in patients with ERBB2-positive metastatic colorectal cancer and could potentially represent a new standard-of-care. Observations The protein ERBB2 is a member of a family of epidermal growth factor receptors that also includes epidermal growth factor receptor (ERBB1), ERBB3, and ERBB4. Amplification of ERBB2 leads to overexpression of the ERBB2 tyrosine kinase receptor, resulting in aberrant signaling and cell migration, growth, adhesion, and differentiation. Colorectal tumors that harbor ERBB2 amplification are more likely to originate on the left side of the colon, are associated with primary and acquired resistance to anti-epidermal growth factor receptor therapies, and have increased incidence of central nervous system metastases. Using immunohistochemistry, fluorescence in situ hybridization, next-generation sequencing, and liquid biopsy techniques, several randomized clinical trials have evaluated the efficacy of ERBB2-targeted therapies in patients with ERBB2-positive metastatic colorectal cancer. These therapies include monoclonal antibodies, antibody-drug conjugates, and tyrosine kinase inhibitors, many of which were associated with favorable efficacy and safety profiles when treating patients with ERBB2-positive metastatic colorectal cancer. Conclusions and Relevance The results of this review suggest the ERBB2 receptor is a promising target for patients with metastatic colorectal cancer; however, to date, no therapies are approved for use in this patient population. Therefore, it is imperative to continue to work to address this unmet need so that patients with ERBB2-positive metastatic colorectal cancer have therapeutic options should they become refractory to treatment with standard therapies.
Collapse
Affiliation(s)
| | - Takayuki Yoshino
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Rondell P Graham
- Division of Laboratory Genetics and Genomics, Division of Anatomic Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Salvatore Siena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda and Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | | |
Collapse
|
29
|
Mohamed AA, Lau DK, Chau I. HER2 targeted therapy in colorectal cancer: New horizons. Cancer Treat Rev 2022; 105:102363. [DOI: 10.1016/j.ctrv.2022.102363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/18/2022] [Accepted: 02/19/2022] [Indexed: 12/18/2022]
|
30
|
Wang QW, Wang XY, Zhang QW, Chen JN, Zhou YJ, Tang ZR, Wang RL, Chen H, Chen H, Li XB. ERBB2 Mutations as Potential Predictors for Recurrence in Colorectal Serrated Polyps by Targeted Next-Generation Sequencing. Front Oncol 2022; 12:769709. [PMID: 35402217 PMCID: PMC8984468 DOI: 10.3389/fonc.2022.769709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 02/22/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundFollow-up guidelines for serrated polyps (SPs) are mainly based on factors such as histology and size with limited evidence. The underlying genomic mechanism of SPs in relation to recurrence risks is utterly unknown.MethodsWe applied targeted next-generation sequencing (NGS) approach on two groups of SPs [polyp-relapsed SPs (PRSPs) vs. polyp-free SPs (PFSPs)] based on the surveillance outcomes to compare differences of DNA variants in 71 colorectal cancer-associated genes. A multicenter validation cohort was established longitudinally from 2016 to 2019 to confirm the relevant results.ResultsAmong the 96 NGS samples, at least one mutant after filtration was detected in 90 samples (94%). Molecular profiling presented BRAF, KRAS, and APC as top 3 mutated genes. FBXW7, MSH2, and ERBB2 might be recurrence-relevant, while DMD, BRCA1, and BRCA2 might be negatively correlated with recurrence. Notably, ERBB2 mutants (R678Q and V842I) (n = 5) had higher risks of polyp recurrence than the wild types (n = 85), with a median polyp-free interval of 15 months compared to 26 months [P < 0.001; hazard ratio (HR) = 4.9; 95% confidence interval (CI) = 1.9–12.8]. Furthermore, a multicenter cohort composed by 321 SPs verified that ERBB2-mutated SPs had increased risks of polyp recurrence (P < 0.001; HR = 3.7; 95% CI = 2.3–6.0) and advanced neoplastic lesion (ANL) recurrence (P < 0.001; HR = 10.0; 95% CI = 2.7–36.9) compared with wild-type SPs, respectively.ConclusionsOur results are emphasizing that SP individuals with ERBB2 mutants are at higher risks of subsequent colorectal neoplasms. ERBB2 mutants might work as facilitated markers for prediction of high-risk SPs and might implicate a potential mechanism in the serrated pathway to colorectal carcinoma (CRC).
Collapse
Affiliation(s)
- Qi-Wen Wang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Xin-Yuan Wang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Qing-Wei Zhang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Jin-Nan Chen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Yu-Jie Zhou
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Zhao-Rong Tang
- Department of Gastroenterology and Hepatology, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Rui-Lan Wang
- Department of Gastroenterology and Hepatology, Sichuan Provincial Corps Hospital of Chinese People’s Armed Forces, Leshan, China
| | - Haoyan Chen
- State Key Laboratory for Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huimin Chen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
- *Correspondence: Xiao-Bo Li, ; Huimin Chen,
| | - Xiao-Bo Li
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
- *Correspondence: Xiao-Bo Li, ; Huimin Chen,
| |
Collapse
|
31
|
Kim H, Kim R, Kim HR, Jo H, Kim H, Ha SY, Park JO, Park YS, Kim ST. HER2 Aberrations as a Novel Marker in Advanced Biliary Tract Cancer. Front Oncol 2022; 12:834104. [PMID: 35252005 PMCID: PMC8896348 DOI: 10.3389/fonc.2022.834104] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/18/2022] [Indexed: 11/13/2022] Open
Abstract
HER2 aberrations have been reported as a novel biomarker in HER2-directed therapy or as a prognostic marker in various tumor types. However, in advanced biliary tract cancer (BTC), there have been few studies regarding HER2 aberrations as a biomarker. We analyzed 121 advanced BTC patients who had been treated with Gemcitabine/Cisplatin (GP) as a 1st line therapy between November 2019 and April 2021. Next-generation sequencing (NGS), namely, HER2 aberrations was performed in all patients. The TruSight™ Oncology 500 assay from Illumina was used for the NGS panel. Among 121 patients with advanced BTC, HER2 aberrations were observed in 18 patients (14.9%). For subtypes of HER2 aberrations, point mutation was observed in 5 patients (27.8%), gene amplification in 11 patients (61.1%), and both point mutation and gene amplification in 2 patients (11.1%). The frequency of HER2 aberrations was significantly different according to the primary tumor (p = 0.009). In gallbladder cancer, HER2 aberrations were observed at a relatively high frequency (36.4%). The tumor response to GP did not differ between patients with and without HER2 aberrations (33.3%, vs. 26.2%, respectively, p = 0.571). The median progression-free survival (PFS) to GP was 4.7 months (95% CI, 4.0 to 5.5 months) in patients with HER2 aberrations and 7.0 months (95% CI, 5.2 to 8.8 months) without HER2 aberrations (p = 0.776). The median overall survival (OS) was not reached and not reached in patients with and without HER2 aberrations (p = 0.739), respectively. The univariate analysis for PFS to GP and OS showed that HER2 aberrations were not an independent factor for survival. This study showed that the HER2 aberrations were observed in 14.9% of advanced BTC and were not an independent biomarker for survival.
Collapse
Affiliation(s)
- Hongsik Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.,Division of Hematology-Oncology, Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, South Korea
| | - Ryul Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hye Ryeon Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hyunji Jo
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hana Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Sang Yun Ha
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Joon Oh Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Young Suk Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Seung Tae Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| |
Collapse
|
32
|
Shah AN, Sunderraj A, Finkelman B, See SH, Davis AA, Gerratana L, Wehbe F, Katam N, Mahalingam D, Gradishar WJ, Behdad A, Blanco L, Cristofanilli M. Positive predictive value of ERBB2 copy number gain by tissue or circulating tumor DNA next-generation sequencing across advanced cancers. Oncotarget 2022; 13:273-280. [PMID: 35126865 PMCID: PMC8809397 DOI: 10.18632/oncotarget.28188] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/10/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND The correlation of ERBB2 copy number gain (CNG) from tissue or circulating tumor DNA (ctDNA) by next-generation sequencing (NGS) with standard HER2 tissue evaluation is not well understood. MATERIALS AND METHODS We retrospectively identified patients with ERBB2 CNG on commercial NGS. We described their clinical-pathologic features and calculated the positive predictive value (PPV) of ERBB2 CNG by NGS for HER2-positivity by IHC and FISH testing. RESULTS 176 patients had NGS revealing an ERBB2 CNG (112 by tumor tissue and 91 by ctDNA). The cancer subtypes with the most cases with ERBB2 CNG by NGS were breast (n = 67), non-small cell lung (n = 25), colorectal (n = 18), gastroesophageal (n = 17), pancreatic (n = 11), and uterine (n = 11). The PPV of ERBB2 CNG in determining HER2 positivity by standard IHC/FISH definitions was 88% for tissue NGS (n = 57) and 80% for ctDNA (n = 47). The PPV among breast cancer patients for tissue NGS was 97% (n = 35) and ctDNA was 93% (n = 39). However, for non-breast cancer cases, the PPV of ERBB2 amplification by tissue NGS dropped to 76% (n = 22) and by ctDNA to 44% (n = 7). CONCLUSIONS ERBB2 CNG by NGS is detected in numerous malignancies for which HER2 testing is not standard. Detection of ERBB2 CNG by tissue NGS and ctDNA has a high PPV for true HER2-positivity by standard IHC and/or FISH testing in breast cancer.
Collapse
Affiliation(s)
- Ami N. Shah
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Ashwin Sunderraj
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Brian Finkelman
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Sharlene H. See
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Andrew A. Davis
- Siteman Cancer Center of Washington University, St. Louis, MO, USA
| | - Lorenzo Gerratana
- Department of Medical Oncology, Centro di Riferimento Oncologico (CRO), IRCCS, Aviano, Italy
| | - Firas Wehbe
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Neelima Katam
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Deva Mahalingam
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - William J. Gradishar
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Amir Behdad
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Luis Blanco
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Massimo Cristofanilli
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| |
Collapse
|
33
|
Cho D, Lord SJ, Simes J, Cooper W, Friedlander M, Bae S, Lee CK. Next-generation sequencing, should I use anti-HER2 therapy for HER2-amplified tumors off-label? Illustrating an extrapolation framework. Ther Adv Med Oncol 2022; 14:17588359221112822. [PMID: 35923921 PMCID: PMC9340898 DOI: 10.1177/17588359221112822] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/23/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Next-generation sequencing is used to increase targeted treatment opportunities, particularly for patients who have exhausted standard options. Where randomized controlled trial evidence for a targeted therapy is available for molecular alterations in one tumor type, the dilemma for the clinician is whether ‘matching’ targeted agents should be recommended off-label for the same molecular alterations detected in other tumor types, for which no trial data are available to guide practice. To judge the likely benefits, it may be possible to extrapolate evidence from cancers where treatment benefits have been established. Methods: We present a framework for assessing the appropriateness of extrapolation using trastuzumab, an anti-HER2 antibody, for HER2-amplified tumors where trastuzumab use would be off-label as an illustrative example. Results: The following should be considered for the tumor type where trastuzumab would be off-label: (a) reliability of the NGS assay for detecting HER2 amplification; (b) criteria for defining HER2 positivity; (c) strength of evidence supporting the actionability of HER2 amplification and trastuzumab; (d) whether better clinical outcomes with trastuzumab are due to a more favorable natural history rather than trastuzumab effect; (e) signals of trastuzumab activity and whether it translates to clinically meaningful benefit; (f) whether the safety profile of trastuzumab differs from established indications; and (g) discussion points for shared decision making (SDM) to facilitate informed consent. Conclusion: We present a systematic approach for appraising evidence to support extrapolating trastuzumab benefits from established indications to off-label applications. Extrapolation criteria and areas of uncertainty to inform SDM are outlined. This framework is potentially generalizable to other tumor-agnostic biomarker-targeted therapy scenarios. It is a practical approach for clinicians to apply in routine practice and should be considered by molecular tumor boards who make off-label recommendations.
Collapse
Affiliation(s)
- Doah Cho
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Locked Bag 77, Camperdown, NSW 1450, Australia
| | - Sarah J. Lord
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
- School of Medicine, University of Notre Dame, Sydney, NSW, Australia
| | - John Simes
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Wendy Cooper
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
- Sydney Medical School, University of Sydney, Camperdown, NSW, Australia
- Discipline of Pathology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Michael Friedlander
- Prince of Wales Clinical School, University of New South Wales, Sydney, NSW, Australia
- Department of Medical Oncology, Prince of Wales Hospital, Randwick, NSW, Australia
| | - Susie Bae
- Department of Oncology, Eastern Health, Box Hill, VIC, Australia
- Department of Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Chee Khoon Lee
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
- Cancer Care Centre, St George Hospital, Kogarah, NSW, Australia
| |
Collapse
|
34
|
Precision Medicine for the Treatment of Colorectal Cancer: the Evolution and Status of Molecular Profiling and Biomarkers. CURRENT COLORECTAL CANCER REPORTS 2021. [DOI: 10.1007/s11888-021-00466-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
35
|
Zhao Y, Wang S, Yang Z, Dong Y, Wang Y, Zhang L, Hu H, Han B. Co-Occurring Potentially Actionable Oncogenic Drivers in Non-Small Cell Lung Cancer. Front Oncol 2021; 11:665484. [PMID: 34221980 PMCID: PMC8242190 DOI: 10.3389/fonc.2021.665484] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/16/2021] [Indexed: 12/25/2022] Open
Abstract
Background Several oncogenic drivers in non-small cell lung cancer (NSCLC) are considered actionable with available or promising targeted therapies. Although targetable drivers rarely overlap with each other, there were a minority of patients harboring co-occurring actionable oncogenic targets, whose clinical characteristics and prognosis are not yet clear. Methods A total of 3,077 patients with NSCLC who underwent molecular analysis by NGS were included, and their demographic and clinical data were retrospectively collected. Results Our study found that the frequency of NSCLC patients harboring co-occurring potentially actionable alterations was approximately 1.5% (46/3077); after excluding patients with EGFR-undetermined mutations, the incidence was 1.3% (40/3077); 80% (37/46) harbored both EGFR mutations and other potentially actionable drivers such as MET amplification (21.6%; 8/37) and alterations in ERBB2 including mutations (27%; 10/37) and amplification (21.6%; 8/37); other combinations of potentially actionable drivers including alterations in ERBB2, KRAS, MET, ALK, and RET were also identified. Additionally, de novo MET/ERBB2 amplification in patients harboring EGFR-mutant NSCLC treated with first-generation EGFR tyrosine kinase inhibitors (TKIs) was associated with shorter PFS (p < 0.05). The efficacy of TKIs in NSCLC patients harboring other co-occurring potentially actionable drivers varied across different molecular subtypes. Conclusions Approximately 1.5% of NSCLCs harbored co-occurring potentially actionable oncogenic drivers, commonly involving EGFR mutations. Co-occurring actionable targets may impact the efficacy of TKIs; therefore, future clinical trials in these patients should be anticipated to tailor the combination or sequential treatment strategies.
Collapse
Affiliation(s)
- Yiming Zhao
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Shuyuan Wang
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhengyu Yang
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Dong
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yanan Wang
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Lele Zhang
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hai Hu
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Baohui Han
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
36
|
Iwaya M, Hayashi H, Nakajima T, Matsuda K, Kinugawa Y, Tobe Y, Tateishi Y, Iwaya Y, Uehara T, Ota H. Colitis-associated colorectal adenocarcinomas frequently express claudin 18 isoform 2: implications for claudin 18.2 monoclonal antibody therapy. Histopathology 2021; 79:227-237. [PMID: 33590909 DOI: 10.1111/his.14358] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/02/2021] [Accepted: 02/15/2021] [Indexed: 01/09/2023]
Abstract
AIMS Claudin 18 (CLDN18) is a member of the claudin family of cell surface proteins, which are widely expressed in epithelial cells and play a role in cell-cell adhesion. CLDN18 isoform 2 (CLDN18.2) is specifically expressed in gastric epithelial cells, and is frequently expressed at high levels in gastric adenocarcinoma. On the basis of this, zolbetuximab, a targeted monoclonal antibody, has been developed for patients with CLDN18.2-positive gastro-oesophageal adenocarcinoma. Colitis-associated colorectal adenocarcinomas (CACs) tend to lose intestinal markers and show aberrant gastric mucin expression. Furthermore, clinical trials of human epidermal growth factor receptor 2 (HER2) inhibitor therapy for colorectal carcinoma are ongoing. However, the expression profile of CLDN18.2 and HER2 has not been described in a series of human CACs. METHODS AND RESULTS We performed immunohistochemistry for CLDN18 and HER2 on 56 consecutive CACs from 55 inflammatory bowel disease patients, and compared the expression profile with that of a control group of 56 sporadic colorectal adenocarcinomas (CRCs). CLDN18.1 expression and CLDN18.2 expression were validated by reverse transcription polymerase chain reaction (PCR) in paraffin-embedded CRC tissues. CLDN18 was positive in 27% (15/56) of CACs and in 5% (3/56) of sporadic CRCs (P = 0.004), and CLDN18-positive CACs were more likely to have lymph node metastasis than CLDN18-negative CACs (67% versus 36%; P = 0.017). CLDN18 expression was significantly associated with MUC5AC expression (P < 0.001) and loss of special AT-rich sequence-binding protein 2 expression (P = 0.005) in CACs. CLDN18.2 was expressed in CRCs that were immunoreactive for CLDN18. Only 4% of CACs were immunoreactive for HER2, and no differences were identified in sporadic CRCs. CONCLUSIONS These findings suggest that certain CAC cases may be candidates for targeted zolbetuximab therapy.
Collapse
Affiliation(s)
- Mai Iwaya
- Department of Laboratory Medicine, Shinshu University Hospital, Matsumoto, Japan
| | - Hiroyuki Hayashi
- Department of Pathology, Yokohama Municipal Citizen's Hospital, Yokohama, Japan
| | - Tomoyuki Nakajima
- Department of Laboratory Medicine, Shinshu University Hospital, Matsumoto, Japan
| | - Kazuyuki Matsuda
- Department of Clinical Laboratory Sciences, School of Health Sciences, Shinshu University, Matsumoto, Japan
| | - Yasuhiro Kinugawa
- Department of Laboratory Medicine, Shinshu University Hospital, Matsumoto, Japan
| | - Yosuke Tobe
- Department of Laboratory Medicine, Shinshu University Hospital, Matsumoto, Japan
| | - Yoko Tateishi
- Department of Pathology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Yugo Iwaya
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takeshi Uehara
- Department of Laboratory Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hiroyoshi Ota
- Department of Clinical Laboratory Sciences, School of Health Sciences, Shinshu University, Matsumoto, Japan
| |
Collapse
|
37
|
Robinson CL, Harrison BT, Ligon AH, Dong F, Maffeis V, Matulonis U, Nucci MR, Kolin DL. Detection of ERBB2 amplification in uterine serous carcinoma by next-generation sequencing: an approach highly concordant with standard assays. Mod Pathol 2021; 34:603-612. [PMID: 33077919 DOI: 10.1038/s41379-020-00695-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 09/23/2020] [Accepted: 09/23/2020] [Indexed: 01/29/2023]
Abstract
Uterine serous carcinoma is an aggressive subtype of endometrial cancer that accounts for fewer than 10% of endometrial carcinomas but is responsible for about half of deaths. A subset of cases has HER2 overexpression secondary to ERBB2 gene amplification, and these patients may benefit from anti-HER2 therapies, such as trastuzumab. HER2 protein overexpression is currently assessed by immunohistochemistry (IHC) and ERBB2 gene amplification by fluorescence in situ hybridization (FISH). Targeted next-generation sequencing (NGS) is increasingly used to routinely identify predictive and prognostic molecular abnormalities in endometrial carcinoma. To investigate the ability of a targeted NGS panel to detect ERBB2 amplification, we identified cases of uterine serous carcinoma (n = 93) and compared HER2 expression by IHC and copy number assessed by FISH with copy number status assessed by NGS. ERBB2 copy number status using a combination of IHC and FISH was interpreted using the 2018 ASCO/CAP guidelines for breast carcinoma. ERBB2 amplification by NGS was determined by the relative number of reads mapping to ERBB2 in tumor DNA compared to control nonneoplastic DNA. Cases with copy number ≥6 were considered amplified and copy number <6 were non-amplified. By IHC, 70 specimens were classified as negative (0 or 1+), 19 were classified as equivocal (2+), and 4 were classified as positive (3+). Using combined IHC/FISH, ERBB2 amplification was observed in 8 of 93 cases (9%). NGS identified the same 8 cases with copy number ≥6; all 85 others had copy number <6. In this series, NGS had 100% concordance with combined IHC/FISH in identifying ERBB2 amplification. NGS is highly accurate in detecting ERBB2 amplification in uterine serous carcinoma and provides an alternative to measurement by IHC and FISH.
Collapse
Affiliation(s)
| | - Beth T Harrison
- Department of Pathology, Division of Women's and Perinatal Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Azra H Ligon
- Department of Pathology, Division of Clinical Cytogenetics, Brigham and Women's Hospital, Boston, MA, USA
| | - Fei Dong
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Valeria Maffeis
- Department of Medicine (DIMED), Surgical Pathology and Cytopathology Unit, University of Padova, Padova, Italy
| | | | - Marisa R Nucci
- Department of Pathology, Division of Women's and Perinatal Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - David L Kolin
- Department of Pathology, Division of Women's and Perinatal Pathology, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
38
|
Liu Z, Shi M, Li X, Song S, Liu N, Du H, Ye J, Li H, Zhang Z, Zhang L. HER2 copy number as predictor of disease-free survival in HER2-positive resectable gastric adenocarcinoma. J Cancer Res Clin Oncol 2021; 147:1315-1324. [PMID: 33543328 PMCID: PMC8021510 DOI: 10.1007/s00432-021-03522-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 01/10/2021] [Indexed: 01/28/2023]
Abstract
Purpose The identification of HER2 overexpression in a subset of gastric adenocarcinoma (GA) patients represents a significant step forward in unveiling the molecular complexity of this disease. The predictive and prognostic value of HER2 amplification in advanced HER2 inhibitor-treated GA patients has been investigated. However, its predictive value in resectable patients remains elusive. Methods We enrolled 98 treatment-naïve resectable Chinese GA patients with HER2 overexpression assessed using IHC. Capture-based targeted sequencing using a panel consisting of 41 gastrointestinal cancer-related genes was performed on tumor tissues. Furthermore, we also investigated the correlation between HER2 copy number (CN) and survival outcomes. Results Of the 98 HER2-overexpressed patients, 90 had HER2 CN amplification assessed using next-generation sequencing, achieving 92% concordance. The most commonly seen concurrent mutations were occurring in TP53, EGFR and PIK3CA. We found HER2 CN as a continuous variable was an independent predictor associated with DFS (p = 0.029). Our study revealed HER2 CN-high patients showed a trend of intestinal-type GA predominant (p = 0.075) and older age (p = 0.07). The median HER2 CN was 15.34, which was used to divide the cohort into CN-high and CN-low groups. Patients with high HER2 CN had a significantly shorter DFS than patients with low HER2 CN (p = 0.002). Furthermore, HER2 CN as a categorical variable was also an independent predictor associated with DFS in patients. Conclusion We elucidated the mutation spectrum of HER2-positive resectable Chinese GA patients and the association between HER2 CN and DFS. Our work revealed HER2 CN as an independent risk factor predicted unfavorable prognosis in HER2-positive GA patients and allowed us to further stratify HER2-positive resectable GA patients for disease management.
Collapse
Affiliation(s)
- Zimin Liu
- Oncology Department, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| | - Mingpeng Shi
- Operating Room of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Xiaoxiao Li
- Oncology Department, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Shanai Song
- Oncology Department, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Ning Liu
- Oncology Department, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Haiwei Du
- Burning Rock Biotech, Guangzhou, China
| | - Junyi Ye
- Burning Rock Biotech, Guangzhou, China
| | - Haiyan Li
- Burning Rock Biotech, Guangzhou, China
| | | | - Lu Zhang
- Burning Rock Biotech, Guangzhou, China
| |
Collapse
|
39
|
Jeong H, Jeong JH, Kim KP, Lee SS, Oh DW, Park DH, Song TJ, Park Y, Hong SM, Ryoo BY, Yoo C. Feasibility of HER2-Targeted Therapy in Advanced Biliary Tract Cancer: A Prospective Pilot Study of Trastuzumab Biosimilar in Combination with Gemcitabine Plus Cisplatin. Cancers (Basel) 2021; 13:cancers13020161. [PMID: 33418871 PMCID: PMC7825072 DOI: 10.3390/cancers13020161] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/24/2020] [Accepted: 01/02/2021] [Indexed: 12/22/2022] Open
Abstract
The prognosis of advanced biliary tract cancer (BTC) is poor with the standard gemcitabine and cisplatin (GemCis) regimen. Given that the rates of human epidermal growth factor receptor 2 (HER2) positivity in BTC reaches around 15%, HER2-targeted therapy needs further investigation. This study aims to evaluate the preliminary efficacy/safety of first-line trastuzumab-pkrb plus GemCis in patients with advanced BTC. Patients with unresectable/metastatic HER2-positive BTC received trastuzumab-pkrb (on day 1 of each cycle, 8 mg/kg for the first cycle and 6 mg/kg for subsequent cycles), gemcitabine (1000 mg/m2 on day 1 and 8) and cisplatin (25 mg/m2 on day 1 and 8) every 3 weeks. Of the 41 patients screened, 7 had HER2-positive tumours and 4 were enrolled. The median age was 72.5 years (one male). Primary tumour locations included extrahepatic (N = 2) and intrahepatic (N = 1) bile ducts, and gallbladder (N = 1). Best overall response was a partial response in two patients and stable disease in two patients. Median progression-free survival (PFS) was 6.1 months and median overall survival (OS) was not reached. The most common grade 3 adverse event was neutropenia (75%), but febrile neutropenia did not occur. No patient discontinued treatment due to adverse events. Trastuzumab-pkrb with GemCis showed promising preliminary feasibility in patients with HER2-positive advanced BTC.
Collapse
Affiliation(s)
- Hyehyun Jeong
- Asan Medical Center, Department of Oncology, University of Ulsan College of Medicine, Seoul 05505, Korea; (H.J.); (J.H.J.); (K.-P.K.); (B.-Y.R.)
| | - Jae Ho Jeong
- Asan Medical Center, Department of Oncology, University of Ulsan College of Medicine, Seoul 05505, Korea; (H.J.); (J.H.J.); (K.-P.K.); (B.-Y.R.)
| | - Kyu-Pyo Kim
- Asan Medical Center, Department of Oncology, University of Ulsan College of Medicine, Seoul 05505, Korea; (H.J.); (J.H.J.); (K.-P.K.); (B.-Y.R.)
| | - Sang Soo Lee
- Asan Medical Center, Department of Gastroenterology, University of Ulsan College of Medicine, Seoul 05505, Korea; (S.S.L.); (D.W.O.); (D.H.P.); (T.J.S.)
| | - Dong Wook Oh
- Asan Medical Center, Department of Gastroenterology, University of Ulsan College of Medicine, Seoul 05505, Korea; (S.S.L.); (D.W.O.); (D.H.P.); (T.J.S.)
| | - Do Hyun Park
- Asan Medical Center, Department of Gastroenterology, University of Ulsan College of Medicine, Seoul 05505, Korea; (S.S.L.); (D.W.O.); (D.H.P.); (T.J.S.)
| | - Tae Jun Song
- Asan Medical Center, Department of Gastroenterology, University of Ulsan College of Medicine, Seoul 05505, Korea; (S.S.L.); (D.W.O.); (D.H.P.); (T.J.S.)
| | - Yangsoon Park
- Asan Medical Center, Department of Pathology, University of Ulsan College of Medicine, Seoul 05505, Korea; (Y.P.); (S.-M.H.)
| | - Seung-Mo Hong
- Asan Medical Center, Department of Pathology, University of Ulsan College of Medicine, Seoul 05505, Korea; (Y.P.); (S.-M.H.)
| | - Baek-Yeol Ryoo
- Asan Medical Center, Department of Oncology, University of Ulsan College of Medicine, Seoul 05505, Korea; (H.J.); (J.H.J.); (K.-P.K.); (B.-Y.R.)
| | - Changhoon Yoo
- Asan Medical Center, Department of Oncology, University of Ulsan College of Medicine, Seoul 05505, Korea; (H.J.); (J.H.J.); (K.-P.K.); (B.-Y.R.)
- Correspondence: ; Tel.: +82-2-3010-1727
| |
Collapse
|
40
|
Navarro S, Cuatrecasas M, Hernández-Losa J, Landolfi S, Musulén E, Ramón Y Cajal S, García-Carbonero R, García-Foncillas J, Pérez-Segura P, Salazar R, Vera R, García-Alfonso P. [Update of the recommendations for the determination of biomarkers in colorectal carcinoma. National Consensus of the Spanish Society of Medical Oncology and the Spanish Society of Pathology]. REVISTA ESPAÑOLA DE PATOLOGÍA : PUBLICACIÓN OFICIAL DE LA SOCIEDAD ESPAÑOLA DE ANATOMÍA PATOLÓGICA Y DE LA SOCIEDAD ESPAÑOLA DE CITOLOGÍA 2020; 54:41-54. [PMID: 33455693 DOI: 10.1016/j.patol.2020.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/02/2020] [Accepted: 07/26/2020] [Indexed: 11/25/2022]
Abstract
This update of the consensus of the Spanish Society of Medical Oncology (Sociedad Española de Oncología Médica - SEOM) and the Spanish Society of Pathology (Sociedad Española de Anatomía Patológica - SEAP), reviews the advances in the analysis of biomarkers in advanced colorectal cancer (CRC) as well as susceptibility markers of hereditary CRC and molecular biomarkers of localized CRC. Recently published information on the essential determination of KRAS, NRAS and BRAF mutations and the possible benefits of determining the amplification of human epidermal growth factor receptor 2 (HER2), the expression of proteins in the DNA repair pathway and the study of NTRK fusions are also evaluated. From a pathological point of view, the importance of analysing the tumour budding and poorly differentiated clusters and its prognostic value in CRC is reviewed, as well as the impact of molecular lymph node analysis on lymph node staging in CRC. The incorporation of pan-genomic technologies, such as next-generation sequencing (NGS) and liquid biopsy in the clinical management of patients with CRC is also outlined. All these aspects are developed in this guide which, like the previous one, will be revised when necessary in the future.
Collapse
Affiliation(s)
- Samuel Navarro
- Departamento de Patología, Universidad de Valencia, Hospital Clínico Universitario de Valencia, CIBERONC, Valencia, España.
| | | | - Javier Hernández-Losa
- Departamento de Patología, Hospital Universitario Vall d'Hebron, CIBERONC, Barcelona, España
| | - Stefania Landolfi
- Departamento de Patología, Hospital Universitario Vall d'Hebron, CIBERONC, Barcelona, España
| | - Eva Musulén
- Departamento de Patología, Hospital Universitari General de Catalunya, Grupo Quirónsalud, Sant Cugat del Vallès, España; Grupo de Epigenética del Cáncer, Institut de Recerca contra la Leucèmia Josep Carreras, Badalona, España
| | - Santiago Ramón Y Cajal
- Departamento de Patología, Hospital Universitario Vall d'Hebron, CIBERONC, Barcelona, España
| | - Rocío García-Carbonero
- Departamento de Oncología Médica, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), UCM, CNIO, CIBERONC, Madrid, España
| | - Jesús García-Foncillas
- Departamento de Oncología, Hospital Universitario Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, España
| | - Pedro Pérez-Segura
- Departamento de Oncología Médica, Hospital Clínico Universitario San Carlos, CIBERONC, Madrid, España
| | - Ramón Salazar
- Departamento de Oncología Médica, ICO ĹHospitalet, Oncobell Program (IDIBELL), CIBERONC, Hospitalet de Llobregat, España
| | - Ruth Vera
- Departamento de Oncología Médica, Complejo Hospitalario de Navarra, Navarrabiomed, IDISNA, Pamplona, España
| | - Pilar García-Alfonso
- Departamento de Oncología Médica, Hospital General Universitario Gregorio Marañón, Madrid, España
| |
Collapse
|
41
|
Mukherji R, Marshall JL, Seeber A. Genomic Alterations and Their Implications on Survival in Nonmetastatic Colorectal Cancer: Status Quo and Future Perspectives. Cancers (Basel) 2020; 12:E2001. [PMID: 32707813 PMCID: PMC7465976 DOI: 10.3390/cancers12082001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/17/2020] [Accepted: 07/18/2020] [Indexed: 02/08/2023] Open
Abstract
The selection of treatment according to genomic alterations is a standard approach in metastatic colorectal cancer but is only starting to have an impact in the earlier stages of the disease. The status if genes like KRAS, BRAF, and MMR has substantial survival implications, and concerted research efforts have revolutionized treatment towards precision oncology. In contrast, a genomic-based approach has not changed the adjuvant setting after curative tumor-resection in the daily routine so far. This review focuses on the current knowledge regarding prognostic and predictive genomic biomarkers in patients with locally advanced nonmetastasized colorectal cancer. Furthermore, we provide an outlook on future challenges for a personalized adjuvant treatment approach in patients with colorectal cancer.
Collapse
Affiliation(s)
- Reetu Mukherji
- Ruesch Center for The Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA;
| | - John L. Marshall
- Ruesch Center for The Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA;
| | - Andreas Seeber
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck A-6020, Austria;
| |
Collapse
|
42
|
Abstract
HER2 (ERBB2) is a member of the ERBB family of receptor tyrosine kinases and functions to drive signaling in the RAS/RAF/MEK/ERK and PI3K/AKT/mTOR pathways. Overall, approximately 2-3% of CRCs exhibit ERBB2 amplification. Multiple phase II clinical trials have now shown that ERBB2 amplification can be predictive of response to anti-ERBB2 targeted therapy. Consequently, recently released guidelines from the National Comprehensive Cancer Network recommend treatment with anti-ERBB2 targeted therapy for RAS wild-type, ERBB2-amplified metastatic CRC. While circumspection is still needed, ERBB2 amplification has now emerged as the next standard-of-care biomarker for metastatic CRC, expanding targeted therapy options for these patients.
Collapse
Affiliation(s)
- Jonathan A Nowak
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.
| |
Collapse
|
43
|
García-Alfonso P, García-Carbonero R, García-Foncillas J, Pérez-Segura P, Salazar R, Vera R, Ramón Y Cajal S, Hernández-Losa J, Landolfi S, Musulén E, Cuatrecasas M, Navarro S. Update of the recommendations for the determination of biomarkers in colorectal carcinoma: National Consensus of the Spanish Society of Medical Oncology and the Spanish Society of Pathology. Clin Transl Oncol 2020; 22:1976-1991. [PMID: 32418154 PMCID: PMC7505870 DOI: 10.1007/s12094-020-02357-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/28/2020] [Indexed: 12/16/2022]
Abstract
In this update of the consensus of the Spanish Society of Medical Oncology (Sociedad Española de Oncología Médica—SEOM) and the Spanish Society of Pathology (Sociedad Española de Anatomía Patológica—SEAP), advances in the analysis of biomarkers in advanced colorectal cancer (CRC) as well as susceptibility markers of hereditary CRC and molecular biomarkers of localized CRC are reviewed. Recently published information on the essential determination of KRAS, NRAS and BRAF mutations and the convenience of determining the amplification of human epidermal growth factor receptor 2 (HER2), the expression of proteins in the DNA repair pathway and the study of NTRK fusions are also evaluated. From the pathological point of view, the importance of analysing the tumour budding and poorly differentiated clusters, and its prognostic value in CRC is reviewed, as well as the impact of molecular lymph node analysis on lymph node staging in CRC. The incorporation of pan-genomic technologies, such as next-generation sequencing (NGS) and liquid biopsy in the clinical management of patients with CRC is also outlined. All these aspects are developed in this guide, which, like the previous one, will remain open to any necessary revision in the future.
Collapse
Affiliation(s)
- P García-Alfonso
- Departament of Medical Oncology, Hospital General Universitario Gregorio Marañón, Madrid, Spain.
| | - R García-Carbonero
- Departament of Medical Oncology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), UCM, CNIO, CIBERONC, Madrid, Spain
| | - J García-Foncillas
- Departament of Medical Oncology, Hospital Universitario Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - P Pérez-Segura
- Departament of Medical Oncology, Hospital Clínico Universitario San Carlos, CIBERONC, Madrid, Spain
| | - R Salazar
- Departament of Medical Oncology, ICO L'Hospitalet, Oncobell Program (IDIBELL), CIBERONC, Hospitalet de Llobregat, Spain
| | - R Vera
- Departament of Medical Oncology, Complejo Hospitalario de Navarra; Navarrabiomed, IDISNA, Pamplona, Spain
| | - S Ramón Y Cajal
- Department of Pathology, Hospital Universitario Vall D'Hebron, CIBERONC, Barcelona, Spain
| | - J Hernández-Losa
- Department of Pathology, Hospital Universitario Vall D'Hebron, CIBERONC, Barcelona, Spain
| | - S Landolfi
- Department of Pathology, Hospital Universitario Vall D'Hebron, CIBERONC, Barcelona, Spain
| | - E Musulén
- Department of Pathology, Hospital Universitari General de Catalunya, Grupo Quirónsalud, Sant Cugat del Vallès, Spain.,Cancer Epigenetics Group, Institut de Recerca Contra La Leucèmia Josep Carreras, Badalona, Spain
| | - M Cuatrecasas
- Department of Pathology, Hospital Clinic, CIBERehd, Barcelona, Spain
| | - S Navarro
- Department of Pathology, University of Valencia, Hospital Clínico Universitario de Valencia, CIBERONC, Valencia, Spain
| |
Collapse
|