1
|
Liu D, Jee J, Drilon A, Heilmann AM, Allen JM, Schrock AB, Keller-Evans RB, Li BT. Diverse ERBB2/ERBB3 Activating Alterations and Coalterations Have Implications for HER2/3-Targeted Therapies across Solid Tumors. CANCER RESEARCH COMMUNICATIONS 2025; 5:680-693. [PMID: 40178042 PMCID: PMC12022956 DOI: 10.1158/2767-9764.crc-24-0620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/06/2025] [Accepted: 04/01/2025] [Indexed: 04/05/2025]
Abstract
SIGNIFICANCE CGP provides genomic context for HER2 status beyond the information provided by IHC and FISH, including detection of ERBB2 mutations and co-alterations that may suggest sensitivity/resistance to HER2-directed therapies, and is therefore crucial for guiding treatment choice and understanding individual patient response.
Collapse
Affiliation(s)
- Dazhi Liu
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Justin Jee
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alexander Drilon
- Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medical College, New York, New York
| | | | | | | | | | - Bob T. Li
- Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
2
|
Manrai PA, McHenry A, Sun T, Santin AD, Ratner E, Lin DI, Elvin JA, Hui P, Buza N. Targetable ERBB2/HER2 Mutations in Gynecologic Malignancies: Clinicopathological, Immunohistochemical, and Molecular Correlations. Int J Gynecol Pathol 2025; 44:144-154. [PMID: 38914011 DOI: 10.1097/pgp.0000000000001050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Targeted anti-HER2 therapy has been recently added to the standard treatment recommendations in endometrial serous carcinoma. Current eligibility requires testing for HER2 overexpression and/or gene amplification by immunohistochemistry and by fluorescence in situ hybridization. However, clinical trials have also demonstrated the efficacy of anti-HER2 drugs against activating ERBB2/HER2 mutations in a variety of solid tumor types, and fam-trastuzumab deruxtecan is now approved by the US Food and Drug Administration for HER2 -mutant non-small cell lung cancer. This study aimed at evaluating the detailed clinical, histomorphological, immunohistochemical, and molecular characteristics of gynecologic malignancies with ERBB2/HER2 mutations. We identified 16 tumors with 19 ERBB2/HER2 mutations in our departmental archives: 11 endometrial primaries, 2 endocervical adenocarcinomas, 1 ovarian mucinous adenocarcinoma, 1 tubo-ovarian undifferentiated carcinoma, and 1 high-grade endometrioid adenocarcinoma of Mullerian origin. ERBB2/HER2 mutations most often involved the tyrosine kinase domain (52.6%), and the most frequent specific mutation was R678Q (31.6%), involving the juxtamembrane domain. More than half (54.5%) of endometrial carcinomas and half of all tumors were MMR-deficient, resulting from MSH6 loss in all but 2 tumors. None of the tumors (0%) were POLE- mutated, while 18.8% were TP53 -mutated. HER2 IHC was negative (score 0 or 1+) in 12 tumors (67%) and equivocal (score 2+) in 4 tumors (33%), whereas none of the tumors were scored as HER2 3+. Score 2+ was associated with R678Q, L755S, I767M mutations, and ERBB2/HER2 rearrangement with a breakpoint in exon 23. Concurrent ERBB2/HER2 amplification was identified in 2 endometrial carcinomas, with HER2/CEP17 ratios of 3.1 and 3.5. We also queried the cBioportal database, which revealed 70 ERBB2/HER2 -mutant gynecologic tumors with a total of 77 ERBB2/HER2 mutations, most often involving the active site of the tyrosine kinase domain (n=36; 46.8%), and the most common specific mutation was S310F (n=20; 26%), located in the extracellular domain. Our results provide important details regarding the clinicopathological and molecular associations of potentially actionable ERBB2/HER2 mutations in endometrial carcinoma and other gynecological cancer types and contribute to addressing clinical treatment needs and improving pathology testing recommendations in the future.
Collapse
Affiliation(s)
| | - Austin McHenry
- Department of Pathology, Yale School of Medicine, New Haven, CT
| | - Tong Sun
- Department of Pathology, Yale School of Medicine, New Haven, CT
| | - Alessandro D Santin
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale School of Medicine, New Haven, CT
| | - Elena Ratner
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale School of Medicine, New Haven, CT
| | | | | | - Pei Hui
- Department of Pathology, Yale School of Medicine, New Haven, CT
| | - Natalia Buza
- Department of Pathology, Yale School of Medicine, New Haven, CT
| |
Collapse
|
3
|
Yang M, Yao Y, Wang K, Qi L, Yang B, Khudadad M, Guo Y, Wang Y, Liu Y, Li L, Cao L, Huang Q, Guo Q, Li Q, Yao X, Wang C, Cao W. Clinicopathological characteristics and prognostic significance of HER2 status evaluation in patients with urothelial carcinoma: a retrospective single-center experience in China. Virchows Arch 2025:10.1007/s00428-025-04057-x. [PMID: 40011272 DOI: 10.1007/s00428-025-04057-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/11/2025] [Accepted: 02/16/2025] [Indexed: 02/28/2025]
Abstract
Due to the lack of an in-depth evaluation of urothelial carcinoma (UC) HER2 expression in both primary and recurrent UC, we examined possible clinical and pathological variables associated with altered UC HER2 expression. The HER2 status and clinicopathological features of primary UCs and matched recurrences from 2015 to 2023 (n = 1352) were compared by evolution category (stable or altered). In 1352 UC patients, HER2 protein expression was linked with age, histological grade, and recurrence. HER2 was expressed in 794 UC patients (58.7%) (1 + , 2 + , and 3 +). High-level (HER2 scored 2 + and 3 +) HER2 expression does not correlate with gene amplification. Significant differences in HER2 expression exist between initial and recurrent tumors (p < 0.05), including a shift from positive to negative expression. Recurrent UC patients showed associations between HER2 expression, histological grade (p < 0.05), and time to surgical recurrence (p = 0.008). HER2 expression in high-grade UC correlated positively with PD-L1 expression in tumor cells (TC) (rs = 0.222, p = 0.047) and tumor-associated immune cells (IC) (rs = 0.238, p = 0.032), especially in recurrent patients (rs = 0.464, p = 0.022). HER2 expression was associated with the biological behavior and prognosis of UC patients. Increased HER2 expression in recurrent UC indicates a poorer prognosis. Consequently, retesting for HER2 is essential in cases of recurrent UC. The association between HER2 and PD-L1 expression in high-grade, recurrent UC suggests the potential for utilizing ADC alongside immunotherapy.
Collapse
Affiliation(s)
- Manlin Yang
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Ye Yao
- Department of Biostatistics, University of Michigan, Ann Arbor, USA
| | - Kun Wang
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
- Department of Medical Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Lisha Qi
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Bo Yang
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Mahtab Khudadad
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, Dalhousie University, Room 726, 5788 University Avenue, Halifax, NS, B3H 1V8, Canada
| | - Yuhong Guo
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Yalei Wang
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Yanxue Liu
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Lingmei Li
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Lu Cao
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Qiujuan Huang
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Qianru Guo
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Qing Li
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Xin Yao
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
- Department of Medical Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Cheng Wang
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, Dalhousie University, Room 726, 5788 University Avenue, Halifax, NS, B3H 1V8, Canada.
| | - Wenfeng Cao
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China.
| |
Collapse
|
4
|
Yang L, Liu Y, Guo R, Du J, Liu L, Liu X, Zhao J, Shi F, Zhang X, Su J. CDK4 gene copy number increase and concurrent genetic changes in acral melanoma of a Chinese cohort. Pathology 2025; 57:34-39. [PMID: 39472269 DOI: 10.1016/j.pathol.2024.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 05/14/2024] [Accepted: 06/21/2024] [Indexed: 01/11/2025]
Abstract
Acral melanoma (AM) is the most common subtype of melanoma in the Asian population. Abnormalities in the p16-cyclin D1-CDK4 signalling pathway play a crucial role in the development and progression of AM. However, the CDK4 copy number variations (CNVs) in AM are under-reported. In this study, we investigated CDK4 gene copy number and concurrent molecular changes in a Chinese cohort with AM, to explore CDK4 CNVs and their significance in AM. We examined CDK4 CNVs with fluorescence in situ hybridisation (FISH) in 31 patients with AM. Six patients with CDK4 high-level copy number increase were examined by next-generation sequencing to detect concurrent molecular changes. Using FISH, 12 (12/31, 38.7%) cases showed CDK4 copy number increase, with six (6/31, 19.4%) low-level copy number increase and six (6/31, 19.4%) high-level copy number increase. Five of six CDK4 low-level copy number increase cases were accompanied by polysomy of chromosome 12, while one case was not. Two of six CDK4 high-level copy number increase cases were accompanied by polysomy of chromosome 12, while four cases were not. CDK4 copy number increase was significantly correlated with younger patient age. In six CDK4 high-level copy number increase cases, one case was found to be accompanied by NRAS mutation, one case was accompanied by HER2 mutation, one case was accompanied by BCL2L11 mutation and one case was accompanied by BRAF, HER2 and BCL2L11 mutations. Our study confirmed the presence of CDK4 copy number increase in AM cases. Detecting CDK4 copy number increase by FISH can be reliable in the diagnosis of AM. Some CDK4 copy number increases are the results of polysomy of chromosome 12. CDK4 high-level copy number increase coexists with other pathogenic mutations in AM. CDK4 appears to be a promising target for AM treatment and is expected to be combined with other targeted therapies.
Collapse
Affiliation(s)
- Leyuan Yang
- Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, China
| | - Yan Liu
- Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, China
| | - Ruiping Guo
- Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, China
| | - Juan Du
- Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, China
| | - Lingchao Liu
- Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, China
| | - Xiaolong Liu
- Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, China
| | - Jianfang Zhao
- Department of Pathology, Yan'an Branch of Peking University Third Hospital, Yan'an Traditional Chinese Medicine Hospital, Yan'an, China
| | - Fang Shi
- Department of Pathology, Yan'an Branch of Peking University Third Hospital, Yan'an Traditional Chinese Medicine Hospital, Yan'an, China
| | - Xin Zhang
- Department of Pathology, Yan'an Branch of Peking University Third Hospital, Yan'an Traditional Chinese Medicine Hospital, Yan'an, China
| | - Jing Su
- Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, China.
| |
Collapse
|
5
|
Wilding B, Woelflingseder L, Baum A, Chylinski K, Vainorius G, Gibson N, Waizenegger IC, Gerlach D, Augsten M, Spreitzer F, Shirai Y, Ikegami M, Tilandyová S, Scharn D, Pearson MA, Popow J, Obenauf AC, Yamamoto N, Kondo S, Opdam FL, Bruining A, Kohsaka S, Kraut N, Heymach JV, Solca F, Neumüller RA. Zongertinib (BI 1810631), an Irreversible HER2 TKI, Spares EGFR Signaling and Improves Therapeutic Response in Preclinical Models and Patients with HER2-Driven Cancers. Cancer Discov 2025; 15:119-138. [PMID: 39248702 PMCID: PMC11726021 DOI: 10.1158/2159-8290.cd-24-0306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/12/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
Mutations in ERBB2 (encoding HER2) occur in 2% to 4% of non-small cell lung cancer (NSCLC) and confer poor prognosis. ERBB-targeting tyrosine kinase inhibitors, approved for treating other HER2-dependent cancers, are ineffective in HER2-mutant NSCLC due to dose-limiting toxicities or suboptimal potency. We report the discovery of zongertinib (BI 1810631), a covalent HER2 inhibitor. Zongertinib potently and selectively blocks HER2, while sparing EGFR, and inhibits the growth of cells dependent on HER2 oncogenic driver events, including HER2-dependent human cancer cells resistant to trastuzumab deruxtecan. Zongertinib displays potent antitumor activity in HER2-dependent human NSCLC xenograft models and enhances the activities of antibody-drug conjugates and KRASG12C inhibitors without causing obvious toxicities. The preclinical efficacy of zongertinib translates in objective responses in patients with HER2-dependent tumors, including cholangiocarcinoma (SDC4-NRG1 fusion) and breast cancer (V777L HER2 mutation), thus supporting the ongoing clinical development of zongertinib. Significance: HER2-mutant NSCLC poses a challenge in the clinic due to limited options for targeted therapies. Pan-ERBB blockers are limited by wild-type EGFR-mediated toxicity. Zongertinib is a highly potent and wild-type EGFR-sparing HER2 inhibitor that is active in HER2-driven tumors in the preclinical and clinical settings.
Collapse
Affiliation(s)
| | | | - Anke Baum
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | | | | | - Neil Gibson
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | | | | | | | - Yukina Shirai
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Masachika Ikegami
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | | | - Dirk Scharn
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | | | | | - Anna C. Obenauf
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria
| | - Noboru Yamamoto
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Shunsuke Kondo
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Frans L. Opdam
- Division of Medical Oncology, Department of Clinical Pharmacology, Antoni van Leeuwenhoek/The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Annemarie Bruining
- Department of Radiology, Antoni van Leeuwenhoek/The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Shinji Kohsaka
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Norbert Kraut
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - John V. Heymach
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Flavio Solca
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | | |
Collapse
|
6
|
Li J, Zhang H, Wang J, Deng M, Li Z, Jiang W, Xu K, Wu L, Dong Z, Liu J, Ding Q, Yu H. Development and Validation of an AI-Driven System for Automatic Literature Analysis and Molecular Regulatory Network Construction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405395. [PMID: 39373342 PMCID: PMC11600262 DOI: 10.1002/advs.202405395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/06/2024] [Indexed: 10/08/2024]
Abstract
Decoding gene regulatory networks is essential for understanding the mechanisms underlying many complex diseases. GENET is developed, an automated system designed to extract and visualize extensive molecular relationships from published biomedical literature. Using natural language processing, entities and relations are identified from a randomly selected set of 1788 scientific articles, and visualized in a filterable knowledge graph. The performance of GENET is evaluated and compared with existing methods. The named entity recognition model has achieved an overall precision of 94.23% (4835/5131; 93.56-94.84%), recall of 97.72% (4835/4948; 97.27-98.10%), and an F1 score of 95.94%. The relation extraction model has demonstrated an overall precision of 91.63% (2593/2830; 90.55-92.59%), recall of 89.17% (2593/2908; 87.99-90.25%), and an F1 score of 90.38%. GENET significantly outperforms existing methods in extracting molecular relationships (P < 0.001). Additionally, GENET has successfully predicted WNT family member 4 regulates insulin-like growth factor 2 via signal transducer and activator of transcription 3 in colon cancer. With RNA sequencing data and multiple immunofluorescence, the authenticity of this prediction is validated, supporting the promising feasibility of GENET.
Collapse
Affiliation(s)
- Jia Li
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Key Laboratory of Digestive SystemRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive IncisionRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Engineering Research Center for Artificial Intelligence Endoscopy Interventional Treatment of Hubei ProvinceWuhanHubei430060P. R. China
| | - Hailin Zhang
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Key Laboratory of Digestive SystemRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive IncisionRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Engineering Research Center for Artificial Intelligence Endoscopy Interventional Treatment of Hubei ProvinceWuhanHubei430060P. R. China
| | - Jiamin Wang
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Key Laboratory of Digestive SystemRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive IncisionRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Engineering Research Center for Artificial Intelligence Endoscopy Interventional Treatment of Hubei ProvinceWuhanHubei430060P. R. China
| | - Mei Deng
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Key Laboratory of Digestive SystemRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive IncisionRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Engineering Research Center for Artificial Intelligence Endoscopy Interventional Treatment of Hubei ProvinceWuhanHubei430060P. R. China
| | - Zhiyong Li
- Engineering Research Center for Artificial Intelligence Endoscopy Interventional Treatment of Hubei ProvinceWuhanHubei430060P. R. China
| | - Wei Jiang
- Engineering Research Center for Artificial Intelligence Endoscopy Interventional Treatment of Hubei ProvinceWuhanHubei430060P. R. China
| | - Kejin Xu
- Engineering Research Center for Artificial Intelligence Endoscopy Interventional Treatment of Hubei ProvinceWuhanHubei430060P. R. China
| | - Lianlian Wu
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Key Laboratory of Digestive SystemRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive IncisionRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Engineering Research Center for Artificial Intelligence Endoscopy Interventional Treatment of Hubei ProvinceWuhanHubei430060P. R. China
| | - Zehua Dong
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Key Laboratory of Digestive SystemRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive IncisionRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Engineering Research Center for Artificial Intelligence Endoscopy Interventional Treatment of Hubei ProvinceWuhanHubei430060P. R. China
| | - Jun Liu
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive IncisionRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Nursing Department of Renmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
| | - Qianshan Ding
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Key Laboratory of Digestive SystemRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive IncisionRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Engineering Research Center for Artificial Intelligence Endoscopy Interventional Treatment of Hubei ProvinceWuhanHubei430060P. R. China
| | - Honggang Yu
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Key Laboratory of Digestive SystemRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive IncisionRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Engineering Research Center for Artificial Intelligence Endoscopy Interventional Treatment of Hubei ProvinceWuhanHubei430060P. R. China
| |
Collapse
|
7
|
Cen S, Yuan M, Sun Q, Hou G, Ying J, Xu Q, Zheng Y, Dong Y, Pan H, Han W. Efficacy and safety of dual blockade of HER2 and PD-1 in patients with HER2-positive gastric cancer: a retrospective, multicentre study. Sci Rep 2024; 14:25030. [PMID: 39443515 PMCID: PMC11500361 DOI: 10.1038/s41598-024-76296-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) expression is one of the most important pathological characteristics of gastric cancer. The positive rate of HER2 expression in patients with gastric cancer is approximately 20%. The phase III Keynote-811 study revealed that anti-HER2 and anti-PD-1 therapy combined with chemotherapy could significantly improve the objective response rate as first-line treatment in patients with HER2-positive advanced gastric cancer. In the present study, we aimed to evaluate the efficacy of combination therapy with trastuzumab and PD-1 inhibitors in patients with advanced HER2-positive gastric cancer in a real-world setting. Seventy-two HER2-positive gastric cancer patients from three hospitals in China were retrospectively reviewed. These patients were treated with trastuzumab plus one anti-PD-1 agent with or without chemotherapy. The overall response rate, progression-free survival and overall survival were assessed according to the Response Evaluation Criteria in Solid Tumours (RECIST 1.1). From January 2018 to October 2021, 72 patients with HER2-positive gastric cancer received trastuzumab and a PD-1 inhibitor with or without chemotherapy as neoadjuvant chemotherapy, first-line therapy, second-line therapy or salvage therapy. The ORR was 54.2% for all patients and 79.4% for previously untreated patients. The median PFS and median OS were 10 months (95% CI: 8-13 months) and 26.1 months (95% CI: 18.5-NA months), respectively, for all patients. Grade 3 adverse effects occurred in approximately 25% of patients. Immune-related adverse effects occurred in approximately 12.5% of patients. Trastuzumab and PD-1 inhibitor combination therapy with or without chemotherapy achieved satisfactory survival outcomes in patients with HER2-positive gastric cancer with acceptable safety.
Collapse
Affiliation(s)
- Shuyi Cen
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine(HIM), Chinese Academy of Sciences, 38# Guangji Road, Hangzhou, 310022, Zhejiang, PRC, China
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, 3# East Qinchun Road, Hangzhou, 310016, Zhejiang, PRC, China
| | - Meiqin Yuan
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine(HIM), Chinese Academy of Sciences, 38# Guangji Road, Hangzhou, 310022, Zhejiang, PRC, China
| | - Qunan Sun
- Department of Medical Oncology, the Second Affiliated Hospital, College of Medicine, Zhejiang University, 88# Jiefang Road, Hangzhou, 310009, Zhejiang, PRC, China
| | - Guilan Hou
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine(HIM), Chinese Academy of Sciences, 38# Guangji Road, Hangzhou, 310022, Zhejiang, PRC, China
| | - Jieer Ying
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine(HIM), Chinese Academy of Sciences, 38# Guangji Road, Hangzhou, 310022, Zhejiang, PRC, China
| | - Qi Xu
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine(HIM), Chinese Academy of Sciences, 38# Guangji Road, Hangzhou, 310022, Zhejiang, PRC, China
| | - Yu Zheng
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, 3# East Qinchun Road, Hangzhou, 310016, Zhejiang, PRC, China
| | - Ying Dong
- Department of Medical Oncology, the Second Affiliated Hospital, College of Medicine, Zhejiang University, 88# Jiefang Road, Hangzhou, 310009, Zhejiang, PRC, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, 3# East Qinchun Road, Hangzhou, 310016, Zhejiang, PRC, China.
| | - Weidong Han
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine(HIM), Chinese Academy of Sciences, 38# Guangji Road, Hangzhou, 310022, Zhejiang, PRC, China.
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, 3# East Qinchun Road, Hangzhou, 310016, Zhejiang, PRC, China.
| |
Collapse
|
8
|
Greenman M, Chang YE, McNamara B, Mutlu L, Santin AD. Unmet needs in cervical cancer - can biological therapies plug the gap? Expert Opin Biol Ther 2024; 24:995-1003. [PMID: 39311611 DOI: 10.1080/14712598.2024.2408754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 09/22/2024] [Indexed: 09/26/2024]
Abstract
INTRODUCTION Cervical cancer remains one of the most common gynecologic malignancies worldwide. A disproportionate burden of cases occurs in developing countries due to inadequate screening and treatment. Even among patients adequately treated, in the presence of locally advanced or recurrent disease, outcomes tend to be poor. The introduction of biologic therapy into treatment has increased overall survival; however, a considerable opportunity still exists to improve current standards in treatment. Biologics have shown antitumor activity in multiple tumor types and are actively being pursued for the management of cervical cancer. AREAS COVERED In this article, we will discuss the historical evolution of biologic therapy in cervical cancer including use of angiogenesis inhibitors, immune checkpoint inhibitors, antibody-drug conjugates, and vaccines. We will review how these therapies have been integrated into current treatment recommendations and discuss ongoing investigations intended to improve clinical outcomes. We also postulate on persistent gaps in care. EXPERT OPINION Biologic therapies have had a tremendous impact on our current approach to managing cervical cancer. We anticipate that significant more research and development will be committed to the continued investigation of biologics in cervical cancer in an effort to improve a historically difficult to treat malignancy.
Collapse
Affiliation(s)
- Michelle Greenman
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Yifan Emily Chang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Blair McNamara
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Levent Mutlu
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Alessandro D Santin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
9
|
Mutlu L, McNamara B, Bellone S, Manavella DD, Demirkiran C, Greenman M, Verzosa MSZ, Buza N, Hui P, Hartwich TMP, Harold J, Yang-Hartwich Y, Zipponi M, Altwerger G, Ratner E, Huang GS, Clark M, Andikyan V, Azodi M, Schwartz PE, Santin AD. Trastuzumab deruxtecan (DS-8201a), a HER2-targeting antibody-drug conjugate, demonstrates in vitro and in vivo antitumor activity against primary and metastatic ovarian tumors overexpressing HER2. Clin Exp Metastasis 2024; 41:765-775. [PMID: 38909139 DOI: 10.1007/s10585-024-10297-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 06/05/2024] [Indexed: 06/24/2024]
Abstract
High-grade serous ovarian cancer (HGSOC) and ovarian clear cell carcinoma (CC), are biologically aggressive tumors endowed with the ability to rapidly metastasize to the abdominal cavity and distant organs. About 10% of HGSOC and 30% of CC demonstrate HER2 IHC 3 + receptor over-expression. We evaluated the efficacy of trastuzumab deruxtecan (T-DXd; DS-8201a), a novel HER2-targeting antibody-drug conjugate (ADC) to an ADC isotype control (CTL ADC) against multiple HGSOC and CC tumor models. Eleven ovarian cancer cell lines including a matched primary and metastatic cell line established from the same patient, were evaluated for HER2 expression by immunohistochemistry and flow cytometry, and gene amplification by fluorescence in situ hybridization assays. In vitro experiments demonstrated T-DXd to be significantly more effective against HER2 3 + HGSOC and CC cell lines when compared to CTL ADC (p < 0.0001). T-DXd induced efficient bystander killing of HER2 non-expressing tumor cells when admixed with HER2 3 + cells. In vivo activity of T-DXd was studied in HER2 IHC 3 + HGSOC and CC mouse xenograft models. We found T-DXd to be significantly more effective than CTL ADC against HER2 3 + HGSOC (KR(CH)31) and CC (OVA10) xenografts with a significant difference in tumor growth starting at day 8 (p = 0.0003 for KR(CH)31, p < 0.0001 for OVA10). T-DXd also conferred a survival advantage in both xenograft models. T-DXd may represent an effective ADC against primary and metastatic HER2-overexpressing HGSOC and CC.
Collapse
MESH Headings
- Female
- Humans
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/pathology
- Trastuzumab/therapeutic use
- Trastuzumab/pharmacology
- Immunoconjugates/pharmacology
- Immunoconjugates/therapeutic use
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-2/antagonists & inhibitors
- Animals
- Mice
- Xenograft Model Antitumor Assays
- Camptothecin/analogs & derivatives
- Camptothecin/pharmacology
- Camptothecin/therapeutic use
- Cell Line, Tumor
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacology
- Mice, Nude
Collapse
Affiliation(s)
- Levent Mutlu
- Department of Obstetrics, Gynecology, and Reproductive Sciences, 333 Cedar Street, LSOG 305, PO Box 208063, New Haven, CT, 06520, USA
| | - Blair McNamara
- Department of Obstetrics, Gynecology, and Reproductive Sciences, 333 Cedar Street, LSOG 305, PO Box 208063, New Haven, CT, 06520, USA
| | - Stefania Bellone
- Department of Obstetrics, Gynecology, and Reproductive Sciences, 333 Cedar Street, LSOG 305, PO Box 208063, New Haven, CT, 06520, USA
| | - Diego D Manavella
- Department of Obstetrics, Gynecology, and Reproductive Sciences, 333 Cedar Street, LSOG 305, PO Box 208063, New Haven, CT, 06520, USA
| | - Cem Demirkiran
- Department of Obstetrics, Gynecology, and Reproductive Sciences, 333 Cedar Street, LSOG 305, PO Box 208063, New Haven, CT, 06520, USA
| | - Michelle Greenman
- Department of Obstetrics, Gynecology, and Reproductive Sciences, 333 Cedar Street, LSOG 305, PO Box 208063, New Haven, CT, 06520, USA
| | - Miguel Skyler Z Verzosa
- Department of Obstetrics, Gynecology, and Reproductive Sciences, 333 Cedar Street, LSOG 305, PO Box 208063, New Haven, CT, 06520, USA
| | - Natalia Buza
- Department of Pathology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Pei Hui
- Department of Pathology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Tobias Max Philipp Hartwich
- Department of Obstetrics, Gynecology, and Reproductive Sciences, 333 Cedar Street, LSOG 305, PO Box 208063, New Haven, CT, 06520, USA
| | - Justin Harold
- Department of Obstetrics, Gynecology, and Reproductive Sciences, 333 Cedar Street, LSOG 305, PO Box 208063, New Haven, CT, 06520, USA
| | - Yang Yang-Hartwich
- Department of Obstetrics, Gynecology, and Reproductive Sciences, 333 Cedar Street, LSOG 305, PO Box 208063, New Haven, CT, 06520, USA
| | - Margherita Zipponi
- Department of Obstetrics, Gynecology, and Reproductive Sciences, 333 Cedar Street, LSOG 305, PO Box 208063, New Haven, CT, 06520, USA
| | - Gary Altwerger
- Department of Obstetrics, Gynecology, and Reproductive Sciences, 333 Cedar Street, LSOG 305, PO Box 208063, New Haven, CT, 06520, USA
| | - Elena Ratner
- Department of Obstetrics, Gynecology, and Reproductive Sciences, 333 Cedar Street, LSOG 305, PO Box 208063, New Haven, CT, 06520, USA
| | - Gloria S Huang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, 333 Cedar Street, LSOG 305, PO Box 208063, New Haven, CT, 06520, USA
| | - Mitchell Clark
- Department of Obstetrics, Gynecology, and Reproductive Sciences, 333 Cedar Street, LSOG 305, PO Box 208063, New Haven, CT, 06520, USA
| | - Vaagn Andikyan
- Department of Obstetrics, Gynecology, and Reproductive Sciences, 333 Cedar Street, LSOG 305, PO Box 208063, New Haven, CT, 06520, USA
| | - Masoud Azodi
- Department of Obstetrics, Gynecology, and Reproductive Sciences, 333 Cedar Street, LSOG 305, PO Box 208063, New Haven, CT, 06520, USA
| | - Peter E Schwartz
- Department of Obstetrics, Gynecology, and Reproductive Sciences, 333 Cedar Street, LSOG 305, PO Box 208063, New Haven, CT, 06520, USA
| | - Alessandro D Santin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, 333 Cedar Street, LSOG 305, PO Box 208063, New Haven, CT, 06520, USA.
| |
Collapse
|
10
|
Dinić J, Jovanović Stojanov S, Dragoj M, Grozdanić M, Podolski-Renić A, Pešić M. Cancer Patient-Derived Cell-Based Models: Applications and Challenges in Functional Precision Medicine. Life (Basel) 2024; 14:1142. [PMID: 39337925 PMCID: PMC11433531 DOI: 10.3390/life14091142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/22/2024] [Accepted: 09/07/2024] [Indexed: 09/30/2024] Open
Abstract
The field of oncology has witnessed remarkable progress in personalized cancer therapy. Functional precision medicine has emerged as a promising avenue for achieving superior treatment outcomes by integrating omics profiling and sensitivity testing of patient-derived cancer cells. This review paper provides an in-depth analysis of the evolution of cancer-directed drugs, resistance mechanisms, and the role of functional precision medicine platforms in revolutionizing individualized treatment strategies. Using two-dimensional (2D) and three-dimensional (3D) cell cultures, patient-derived xenograft (PDX) models, and advanced functional assays has significantly improved our understanding of tumor behavior and drug response. This progress will lead to identifying more effective treatments for more patients. Considering the limited eligibility of patients based on a genome-targeted approach for receiving targeted therapy, functional precision medicine provides unprecedented opportunities for customizing medical interventions according to individual patient traits and individual drug responses. This review delineates the current landscape, explores limitations, and presents future perspectives to inspire ongoing advancements in functional precision medicine for personalized cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Milica Pešić
- Department of Neurobiology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11108 Belgrade, Serbia; (J.D.); (S.J.S.); (M.D.); (M.G.); (A.P.-R.)
| |
Collapse
|
11
|
Yoon J, Oh DY. HER2-targeted therapies beyond breast cancer - an update. Nat Rev Clin Oncol 2024; 21:675-700. [PMID: 39039196 DOI: 10.1038/s41571-024-00924-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2024] [Indexed: 07/24/2024]
Abstract
The receptor tyrosine-kinase HER2 (also known as ErbB2) is a well-established therapeutic target in patients with breast or gastric cancer selected on the basis of HER2 overexpression on immunohistochemistry and/or ERBB2 amplification on in situ hybridization. With advances in cancer molecular profiling and increased implementation of precision medicine approaches into oncology practice, actionable HER2 alterations in solid tumours have expanded to include ERBB2 mutations in addition to traditional HER2 overexpression and ERBB2 amplification. These various HER2 alterations can be found in solid tumour types beyond breast and gastric cancer, although few HER2-targeted therapeutic options have been established for the other tumour types. Nevertheless, during the 5 years since our previous Review on this topic was published in this journal, obvious and fruitful progress in the development of HER2-targeted therapies has been made, including new disease indications, innovative drugs with diverse mechanisms of action and novel frameworks for approval by regulatory authorities. These advances have culminated in the recent histology-agnostic approval of the anti-HER2 antibody-drug conjugate trastuzumab deruxtecan for patients with HER2-overexpressing solid tumours. In this new Review, we provide an update on the current development landscape of HER2-targeted therapies beyond breast cancer, as well as anticipated future HER2-directed treatment strategies to overcome resistance and thereby improve efficacy and patient outcomes.
Collapse
Affiliation(s)
- Jeesun Yoon
- Division of Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Do-Youn Oh
- Division of Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, Republic of Korea.
| |
Collapse
|
12
|
Nami B, Wang Z. A Non-Canonical p75HER2 Signaling Pathway Underlying Trastuzumab Action and Resistance in Breast Cancer. Cells 2024; 13:1452. [PMID: 39273024 PMCID: PMC11394428 DOI: 10.3390/cells13171452] [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: 07/14/2024] [Revised: 08/19/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Overexpression of HER2 occurs in 25% of breast cancer. Targeting HER2 has proven to be an effective therapeutic strategy for HER2-positive breast cancer. While trastuzumab is the most commonly used HER2 targeting agent, which has significantly improved outcomes, the overall response rate is low. To develop novel therapies to boost trastuzumab efficacy, it is critical to identify the mechanisms underlying trastuzumab action and resistance. We recently showed that the inhibition of breast cancer cell growth by trastuzumab is not through the inhibition of HER2 canonical signaling. Here we report the identification of a novel non-canonical HER2 signaling pathway and its interference by trastuzumab. We showed that HER2 signaled through a non-canonical pathway, regulated intramembrane proteolysis (RIP). In this pathway, HER2 is first cleaved by metalloprotease ADAM10 to produce an extracellular domain (ECD) that is released and the p95HER2 that contains the transmembrane domain (TM) and intracellular domain (ICD). p95HER2, if further cleaved by an intramembrane protease, γ-secretase, produced a soluble ICD p75HER2 with nuclear localization signal (NLS). p75HER2 is phosphorylated and translocated to the nucleus. Nuclear p75HER2 promotes cell proliferation. Trastuzumab targets this non-canonical HER2 pathway via inhibition of the proteolytic cleavage of HER2 by both ADAM10 and γ-secretase. However, p75HER2 pathway also confers resistance to trastuzumab once aberrantly activated. Combination of trastuzumab with ADAM10 and γ-secretase inhibitors completely blocks p75HER2 production in both BT474 and SKBR3 cells. We concluded that HER2 signals through the RIP signaling pathway that promotes cell proliferation and is targeted by trastuzumab. The aberrant HER2 RIP signaling confers resistance to trastuzumab that could be overcome by the application of inhibitors to ADAM10 and γ-secretase.
Collapse
Affiliation(s)
| | - Zhixiang Wang
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada;
| |
Collapse
|
13
|
Liu Y, Wang C, Chen G, Chen J, Chen W, Lei K, Li J, Pan Y, Li Y, Tang D, Li B, Zhao J, Zeng L. Patient derived cancer organoids model the response to HER2-CD3 bispecific antibody (BsAbHER2) generated from hydroxyapatite gene delivery system. Cancer Lett 2024; 597:217043. [PMID: 38876386 DOI: 10.1016/j.canlet.2024.217043] [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: 02/04/2024] [Revised: 05/13/2024] [Accepted: 06/04/2024] [Indexed: 06/16/2024]
Abstract
HER2-positive cancer is a prevalent subtype of malignancy with poor prognosis, yet current targeted therapies, like Trastuzumab and pyrotinib, have resulted in remission in patients with HER2-positive cancer. This study provides a novel approach for immunotherapy based on a hydroxyapatite (HA) gene delivery system producing a bispecific antibody for HER2-positive cancer treatment. An HA nanocarrier has been synthesized by the classical hydrothermal method. Particularly, the HA-nanoneedle system was able to mediate stable gene expression of minicircle DNA (MC) encoding a humanized anti-CD3/anti-HER2 bispecific antibody (BsAbHER2) in vivo. The produced BsAbs exhibited a potent killing effect not only in HER2-positive cancer cells but also in patient-derived organoids in vitro. This HA-nanoneedle gene delivery system features simple large-scale preparation and clinical applicability. Hence, the HA-nanoneedle gene delivery system combined with minicircle DNA vector encoding BsAbHER2 reported here provides a potential immunotherapy strategy for HER2-positive tumors.
Collapse
Affiliation(s)
- Yuhong Liu
- General Surgery, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, PR China; The Biobank, Scientific Research Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, PR China
| | - Chen Wang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, PR China
| | - Guochuang Chen
- Syno Minicircle Biotechnology, Shenzhen, 518055, PR China
| | - Junzong Chen
- General Surgery, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, PR China
| | - Wei Chen
- The Biobank, Scientific Research Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, PR China
| | - Kefeng Lei
- General Surgery, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, PR China
| | - Jia Li
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, PR China
| | - Yihang Pan
- The Biobank, Scientific Research Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, PR China
| | - You Li
- General Surgery, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, PR China
| | - Di Tang
- General Surgery, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, PR China.
| | - Binbin Li
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, PR China.
| | - Jing Zhao
- General Surgery, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, PR China.
| | - Leli Zeng
- The Biobank, Scientific Research Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, PR China.
| |
Collapse
|
14
|
Bon G, Di Lisa FS, Filomeno L, Arcuri T, Krasniqi E, Pizzuti L, Barba M, Messina B, Schiavoni G, Sanguineti G, Botti C, Cappelli S, Pelle F, Cavicchi F, Puccica I, Costantini M, Perracchio L, Maugeri-Saccà M, Ciliberto G, Vici P. HER2 mutation as an emerging target in advanced breast cancer. Cancer Sci 2024; 115:2147-2158. [PMID: 38715247 PMCID: PMC11247561 DOI: 10.1111/cas.16148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/21/2024] [Accepted: 02/25/2024] [Indexed: 07/13/2024] Open
Abstract
HER2 activating mutations have emerged as oncogenic drivers and therapeutic targets in a variety of human tumors. In breast cancer, these deregulations occur at low frequency, and are mostly detected in HER2-nonamplified, metastatic disease. Preclinical evidence has clarified the role of hotspot mutations in HER2 constitutive activation, defining them as an alternative mechanism to HER2 gene amplification. Furthermore, recent clinical studies have indicated the emergence of newly acquired HER2 deregulations in significant proportions of breast cancer patients who experience disease progression following both endocrine and HER2-targeted therapies. As the involvement of HER2 mutation in therapy resistance may profoundly impact patient outcomes on successive therapies, several clinical trials are currently investigating the efficacy of various HER2-targeted drugs in HER2-mutant breast cancer. In this review, we firstly summarize the structural organization of the HER2 oncogene and its historical impact on breast cancer prognosis and therapeutic advancement. Then, we provide an overview of the frequencies and functional relevance of clinically recurrent HER2 mutations in breast cancer with a special focus on their role in therapeutic resistance. Finally, we provide a collection of the clinical trials that are currently exploring novel therapeutic approaches for this patient subset and discuss the related perspectives and challenges.
Collapse
Affiliation(s)
- Giulia Bon
- Department of Research, Cellular Network and Molecular Therapeutic Target Unit, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Lorena Filomeno
- Phase IV Clinical Studies Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Teresa Arcuri
- Phase IV Clinical Studies Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
- Department of Radiological, Medical Oncology A, Policlinico Umberto I; Oncological and Anatomo-Pathological Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Eriseld Krasniqi
- Phase IV Clinical Studies Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Laura Pizzuti
- Division of Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Maddalena Barba
- Division of Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Beatrice Messina
- Department of Research, Biostatistics and Bioinformatics Unit, Clinical Trial Center, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giulia Schiavoni
- Department of Research, Biostatistics and Bioinformatics Unit, Clinical Trial Center, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giuseppe Sanguineti
- Department of Radiation Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Claudio Botti
- Department of Surgery, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Sonia Cappelli
- Department of Surgery, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Fabio Pelle
- Department of Surgery, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Flavia Cavicchi
- Department of Surgery, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Ilaria Puccica
- Department of Surgery, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Maurizio Costantini
- Department of Plastic and Reconstructive Surgery, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Letizia Perracchio
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Marcello Maugeri-Saccà
- Department of Research, Biostatistics and Bioinformatics Unit, Clinical Trial Center, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
- Scientific Direction, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Gennaro Ciliberto
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Patrizia Vici
- Phase IV Clinical Studies Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
15
|
Pan L, Li J, Xu Q, Gao Z, Yang M, Wu X, Li X. HER2/PI3K/AKT pathway in HER2-positive breast cancer: A review. Medicine (Baltimore) 2024; 103:e38508. [PMID: 38875362 PMCID: PMC11175886 DOI: 10.1097/md.0000000000038508] [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: 03/07/2024] [Revised: 05/01/2024] [Accepted: 05/17/2024] [Indexed: 06/16/2024] Open
Abstract
Breast cancer is currently the most commonly occurring cancer globally. Among breast cancer cases, the human epidermal growth factor receptor 2 (HER2)-positive breast cancer accounts for 15% to 20% and is a crucial focus in the treatment of breast cancer. Common HER2-targeted drugs approved for treating early and/or advanced breast cancer include trastuzumab and pertuzumab, which effectively improve patient prognosis. However, despite treatment, most patients with terminal HER2-positive breast cancer ultimately suffer death from the disease due to primary or acquired drug resistance. The prevalence of aberrantly activated the protein kinase B (AKT) signaling in HER2-positive breast cancer was already observed in previous studies. It is well known that p-AKT expression is linked to an unfavorable prognosis, and the phosphatidylinositol-3-kinase (PI3K)/AKT pathway, as the most common mutated pathway in breast cancer, plays a major role in the mechanism of drug resistance. Therefore, in the current review, we summarize the molecular alterations present in HER2-positive breast cancer, elucidate the relationships between HER2 overexpression and alterations in the PI3K/AKT signaling pathway and the pathways of the alterations in breast cancer, and summarize the resistant mechanism of drugs targeting the HER2-AKT pathway, which will provide an adjunctive therapeutic rationale for subsequent resistance to directed therapy in the future.
Collapse
Affiliation(s)
- Linghui Pan
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Jinling Li
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- Department of Laboratory Medicine, Chonggang General Hospital, Chongqing, China
| | - Qi Xu
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Zili Gao
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Mao Yang
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Xiaoping Wu
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Xuesen Li
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
16
|
Li BT, Meric-Bernstam F, Bardia A, Naito Y, Siena S, Aftimos P, Anderson I, Curigliano G, de Miguel M, Kalra M, Oh DY, Park JO, Postel-Vinay S, Rha SY, Satoh T, Spanggaard I, Michelini F, Smith A, Machado KK, Saura C. Trastuzumab deruxtecan in patients with solid tumours harbouring specific activating HER2 mutations (DESTINY-PanTumor01): an international, phase 2 study. Lancet Oncol 2024; 25:707-719. [PMID: 38710187 DOI: 10.1016/s1470-2045(24)00140-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/01/2024] [Accepted: 03/05/2024] [Indexed: 05/08/2024]
Abstract
BACKGROUND Trastuzumab deruxtecan is a HER2-directed antibody-drug conjugate approved by the US Food and Drug Administration and the European Medicines Agency for HER2-mutant non-small-cell lung cancer. Few treatment options exist for patients with HER2-mutant solid tumours beyond lung cancers. We investigated trastuzumab deruxtecan in metastatic solid tumours with specific activating HER2 mutations. METHODS In this open-label, phase 2, basket study done in 29 centres in Asia, Europe, and North America, we investigated trastuzumab deruxtecan (5·4 mg/kg every 3 weeks by intravenous infusion) in patients aged 18 years or older with unresectable or metastatic solid tumours with specific activating HER2 mutations, an Eastern Cooperative Oncology Group performance status of 0 or 1, and disease progression following previous treatment (previous HER2-targeted therapy was permitted) or with no satisfactory alternative treatment options. The primary endpoint was confirmed objective response rate by independent central review. Anti-tumour activity and safety were analysed in all patients who received at least one dose of trastuzumab deruxtecan. This trial is registered with ClinicalTrials.gov, NCT04639219, and is active but no longer recruiting. FINDINGS Between Dec 30, 2020, and Jan 25, 2023, 102 patients (62 [61%] female and 40 [39%] male; median age 66·5 years [IQR 58-72]; 51 [50%] White, two [2%] Black or African American, 38 [37%] Asian, and 11 [11%] did not have race information reported) with solid tumours with activating HER2 mutations received trastuzumab deruxtecan and were included in the anti-tumour activity and safety analyses sets. Patients had a median of three (IQR 2-4) previous treatment regimens. The median duration of follow-up was 8·61 months (IQR 3·71-12·68). The objective response rate by independent central review was 29·4% (95% CI 20·8-39·3; 30 of 102 patients). 52 (51%) patients had a treatment-emergent adverse event of grade 3 or worse; the most common events (in ≥5% of patients) were anaemia (16 [16%]) and neutrophil count decreased (eight [8%]). Drug-related treatment-emergent serious adverse events occurred in ten (10%) patients. Adjudicated drug-related interstitial lung disease or pneumonitis of any grade occurred in 11 patients (11%; three grade 1, five grade 2, one grade 3, and two grade 5); there were two (2%) cases of fatal adjudicated drug-related interstitial lung disease or pneumonitis. INTERPRETATION Trastuzumab deruxtecan showed anti-tumour activity and durable responses in heavily pretreated patients across multiple tumour types with activating HER2 mutations, with no new safety signals. Prespecified HER2 mutations might be targeted by HER2-directed antibody-drug conjugates and our findings support further investigation of trastuzumab deruxtecan in the pan-tumour setting. FUNDING AstraZeneca and Daiichi Sankyo.
Collapse
Affiliation(s)
- Bob T Li
- Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medicine, Cornell University, New York, NY, USA.
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aditya Bardia
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Yoichi Naito
- Department of General Internal Medicine, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Salvatore Siena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy
| | - Philippe Aftimos
- Institut Jules Bordet-Université Libre de Bruxelles, Brussels, Belgium
| | | | - Giuseppe Curigliano
- Istituto Europeo di Oncologia, IRCCS Milan, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy
| | - Maria de Miguel
- START Madrid Centro Integral Oncológico Clara Campal (CIOCC)-HM Sanchinarro University Hospital, Madrid, Spain
| | - Maitri Kalra
- Ball Memorial Hospital (IU Health), Muncie, IN, USA
| | - Do-Youn Oh
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, Korea
| | - Joon Oh Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sophie Postel-Vinay
- Inserm Unit U981 Gustave Roussy, Villejuif, France; Cancer Institute, University College London, London, UK
| | - Sun Young Rha
- Yonsei Cancer Center, Yonsei University Health System, Seoul, Korea
| | | | - Iben Spanggaard
- Department of Oncology, Rigshospitalet-Copenhagen University Hospital, Copenhagen, Denmark
| | - Flavia Michelini
- Translational Medicine, Oncology Research and Development, AstraZeneca, Waltham, MA, USA
| | - Ann Smith
- Oncology Biometrics, Oncology Research and Development, AstraZeneca, Cambridge, UK
| | - Karime Kalil Machado
- Late Development Oncology, Oncology Research and Development, AstraZeneca, Gaithersburg, MD, USA
| | - Cristina Saura
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
17
|
Ferrari G, Del Rio B, Novello S, Passiglia F. HER2-Altered Non-Small Cell Lung Cancer: A Journey from Current Approaches to Emerging Strategies. Cancers (Basel) 2024; 16:2018. [PMID: 38893138 PMCID: PMC11171190 DOI: 10.3390/cancers16112018] [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: 04/23/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
For patients diagnosed with advanced HER2-altered non-small cell lung cancer (NSCLC), the current standard of care is represented by a platinum-pemetrexed-based chemotherapy, eventually in combination with immunotherapy. Different pan-HER tyrosine kinase inhibitors have been evaluated in limited phase II trials, yielding generally unsatisfactory outcomes, although certain genotypes demonstrated some clinical benefit. Conversely, antibody-drug conjugates (ADCs) targeting HER2, particularly trastuzumab-deruxtecan, have shown promising results against HER2-mutant disease, including a great intracranial activity in patients with brain metastasis. Based on the results obtained from DESTINY-Lung01 and DESTINY-Lung02 trials, trastuzumab deruxtecan received regulatory approval as the first targeted therapy for pre-treated, HER2-mutant, advanced NSCLC patients. More recently, the Food and Drug Administration (FDA) granted the accelerated approval of trastuzumab deruxtecan for advanced, pre-treated HER2-positive solid tumours with no other treatment options. In this scenario, emerging evidence is increasingly pointing towards the exploration of combination regimens with synergistic effects in the advanced disease. In this review, we provide a detailed summary of current approaches and emerging strategies in the management of HER2-altered NSCLC, also focusing on unmet needs, including the treatment of patients with brain metastases.
Collapse
Affiliation(s)
| | | | | | - Francesco Passiglia
- Department of Oncology, University of Turin, San Luigi Hospital, 10124 Orbassano, Italy; (G.F.); (B.D.R.); (S.N.)
| |
Collapse
|
18
|
Bui CM, Oren A, Balzer B, Maluf H, Medeiros F. Human Epidermal Growth Factor Receptor 2 Overexpression/Amplification in Primary Ovarian Endometrioid Carcinoma. Appl Immunohistochem Mol Morphol 2024; 32:222-228. [PMID: 38635473 DOI: 10.1097/pai.0000000000001194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 03/07/2024] [Indexed: 04/20/2024]
Abstract
Human epidermal growth factor receptor 2 (HER2) expression has become increasingly helpful in predicting responses to anti-HER2 agents in gynecological cancers. This study retrospectively analyzed HER2 expression in 48 primary ovarian endometrioid carcinomas. HER2 immunohistochemistry was performed using the Ventana platform (Clone 4B5 monoclonal predilute) following the manufacturer's protocol. HER2 expression was equivocal (score 2+) by image analysis in 2 cases (4.17%) based on the breast cancer criteria. Fluorescence in situ hybridization was negative for HER2 amplification in one case (International Federation of Gynecology and Obstetrics, grade 1) and positive in the other (International Federation of Gynecology and Obstetrics, grade 3). Our findings contribute to the growing evidence that HER2 is overexpressed in a small proportion of ovarian endometrioid carcinoma, and thus may serve as a potential therapeutic target in selected cases.
Collapse
Affiliation(s)
- Chau M Bui
- Department of Pathology and Laboratory Medicine, Cedars Sinai Medical Center, Los Angeles, CA
| | | | | | | | | |
Collapse
|
19
|
Nasrazadani A, Marti JLG, Lathrop K, Restrepo A, Leu SY, Bhat G, Brufsky A. Poziotinib treatment in patients with HER2-positive advanced breast cancer who have received prior anti-HER2 regimens. Breast Cancer Res Treat 2024; 205:29-37. [PMID: 38261228 DOI: 10.1007/s10549-023-07236-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 11/22/2023] [Indexed: 01/24/2024]
Abstract
PURPOSE Poziotinib is an irreversible pan-inhibitor of the human epidermal growth factor receptor (HER) that has shown acceptable tolerability and antitumor activity in phase I and II trials in patients with advanced solid tumors. In the present open-label, multicenter phase II study, we demonstrate safety, tolerability, and preliminary efficacy data from two different dosing schedules in patients with HER2-positive advanced breast cancer. PATIENTS AND METHODS Patients who had received at least two prior HER2-directed therapy lines for advanced disease, received 24 mg poziotinib on an intermittent dosing schedule (cohort 1) or 16 mg poziotinib once daily on a continuous dosing schedule (cohort 2). The primary endpoint was overall response rate (ORR). Secondary endpoints were progression-free survival (PFS), disease control rate (DCR), and time to progression (TTP). Secondary endpoints additionally included safety and pharmacokinetics. RESULTS A total of 67 patients were enrolled. The ORR was 30% in both groups (p = 0.98). DCR was 60% vs 78% (p = 0.15) and median PFS and TTP were 4.1 vs 4.9 months (both p = 0.30) for cohorts 1 and 2, respectively. The most common treatment related adverse events (AEs) of any grade included diarrhea (88% vs 85%, p = 0.76), rash (88% vs 88%, p = 0.96), and stomatitis (64% vs 56%, p = 0.52), with grade 3-4 diarrhea occurring in 33% vs 32% of patients (p = 0.93) and grade 3-4 rash in 27% vs 35% of patients (p = 0.48) in cohort 1 vs cohort 2, respectively. CONCLUSION Poziotinib demonstrated evidence of clinical activity in patients with pre-treated HER2-positive advanced breast cancer, although high levels of toxicity may preclude further studies at this time.
Collapse
Affiliation(s)
- Azadeh Nasrazadani
- Division of Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Kate Lathrop
- The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | | | | | | | - Adam Brufsky
- Magee-Womens Hospital of UPMC, 300 Halket St, Pittsbugh, PA, 15213, USA.
| |
Collapse
|
20
|
Ten Haaft BH, Pedregal M, Prato J, Klümpen HJ, Moreno V, Lamarca A. Revolutionizing anti-HER2 therapies for extrahepatic cholangiocarcinoma and gallbladder cancer: Current advancements and future perspectives. Eur J Cancer 2024; 199:113564. [PMID: 38266541 DOI: 10.1016/j.ejca.2024.113564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/10/2024] [Accepted: 01/15/2024] [Indexed: 01/26/2024]
Abstract
Biliary tract cancers (BTCs) encompass a heterogeneous group of rare tumors, including intrahepatic cholangiocarcinoma (iCCA), extrahepatic cholangiocarcinoma (eCCA), gallbladder cancer (GBC) and ampullary cancer (AC). The present first-line palliative treatment regimen comprises gemcitabine and cisplatin in combination with immunotherapy based on two randomized controlled studies. Despite the thorough investigation of these palliative treatments, long-term survival remains low. Moving beyond conventional chemotherapies and immunotherapies, the realm of precision medicine has demonstrated remarkable efficacy in malignancies such as breast and gastric cancers, characterized by notable HER2 overexpression rates. In the context of biliary tract cancer, significant HER2 alterations are observed, particularly within eCCA and GBC, heightening the interest in precision medicine. Various anti-HER2 therapies, including trastuzumab, pertuzumab, trastuzumab-deruxtecan, zanidatamab and neratinib, have undergone investigation. The objective of this review is to summarize the current evidence and outline future directions of targeted HER2 treatment therapy in patients with biliary tract tumors, specially extrahepatic cholangiocarcinoma and gallbladder cancer.
Collapse
Affiliation(s)
- Britte Hea Ten Haaft
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, the Netherlands; Cancer Center Amsterdam, the Netherlands
| | - Manuel Pedregal
- START Madrid-FJD Phase I Unit, Department of Medical Oncology, Fundación Jiménez Díaz University Hospital, Madrid Spain
| | - Javier Prato
- START Madrid-FJD Phase I Unit, Department of Medical Oncology, Fundación Jiménez Díaz University Hospital, Madrid Spain
| | - Heinz-Josef Klümpen
- Cancer Center Amsterdam, the Netherlands; Amsterdam UMC, location University of Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands
| | - Victor Moreno
- START Madrid-FJD Phase I Unit, Department of Medical Oncology, Fundación Jiménez Díaz University Hospital, Madrid Spain
| | - Angela Lamarca
- Department of Oncology, OncoHealth Institute, Fundación Jiménez Díaz University Hospital, Madrid Spain; Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom.
| |
Collapse
|
21
|
Ahluwalia MS, Khosla AA, Ozair A, Gouda MA, Subbiah V. Impact of tissue-agnostic approvals on management of primary brain tumors. Trends Cancer 2024; 10:256-274. [PMID: 38245379 DOI: 10.1016/j.trecan.2023.11.005] [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: 08/10/2023] [Revised: 11/11/2023] [Accepted: 11/17/2023] [Indexed: 01/22/2024]
Abstract
Novel tissue-agnostic therapeutics targeting driver mutations in tumor cells have been recently approved by FDA, driven by basket trials that have demonstrated their efficacy and safety across diverse tumor histology. However, the relative rarity of primary brain tumors (PBTs) has limited their representation in early trials of tissue-agnostic medications. Thus, consensus continues to evolve regarding utility of tissue-agnostic medications in routine practice for PBTs, a diverse group of neoplasms characterized by limited treatment options and unfavorable prognoses. We describe current and potential impact of tissue-agnostic approvals on management of PBTs. We discuss data from clinical trials for PBTs regarding tissue-agnostic targets, including BRAFV600E, neurotrophic tyrosine receptor kinase (NTRK) fusions, microsatellite instability-high (MSI-High), mismatch repair deficiency (dMMR), and high tumor mutational burden (TMB-H), in context of challenges in managing PBTs. Described are additional tissue-agnostic targets that hold promise for benefiting patients with PBTs, including RET fusion, fibroblast growth factor receptor (FGFR), ERBB2/HER2, and KRASG12C, and TP53Y220C.
Collapse
Affiliation(s)
- Manmeet S Ahluwalia
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA; Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Atulya A Khosla
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA; Department of Internal Medicine, William Beaumont University Hospital, Royal Oak, MI, USA
| | - Ahmad Ozair
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA; Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Mohamed A Gouda
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vivek Subbiah
- Early Phase Drug Development Program, Sarah Cannon Research Institute, Nashville, TN, USA.
| |
Collapse
|
22
|
Malik S, Pradeep SP, Kumar V, Xiao Y, Deng Y, Fan R, Vasquez JC, Singh V, Bahal R. Antitumor efficacy of a sequence-specific DNA-targeted γPNA-based c-Myc inhibitor. Cell Rep Med 2024; 5:101354. [PMID: 38183981 PMCID: PMC10829792 DOI: 10.1016/j.xcrm.2023.101354] [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: 10/13/2022] [Revised: 09/21/2023] [Accepted: 12/11/2023] [Indexed: 01/08/2024]
Abstract
Targeting oncogenes at the genomic DNA level can open new avenues for precision medicine. Significant efforts are ongoing to target oncogenes using RNA-targeted and protein-targeted platforms, but no progress has been made to target genomic DNA for cancer therapy. Here, we introduce a gamma peptide nucleic acid (γPNA)-based genomic DNA-targeted platform to silence oncogenes in vivo. γPNAs efficiently invade the mixed sequences of genomic DNA with high affinity and specificity. As a proof of concept, we establish that γPNA can inhibit c-Myc transcription in multiple cell lines. We evaluate the in vivo efficacy and safety of genomic DNA targeting in three pre-clinical models. We also establish that anti-transcription γPNA in combination with histone deacetylase inhibitors and chemotherapeutic drugs results in robust antitumor activity in cell-line- and patient-derived xenografts. Overall, this strategy offers a unique therapeutic platform to target genomic DNA to inhibit oncogenes for cancer therapy.
Collapse
Affiliation(s)
- Shipra Malik
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Sai Pallavi Pradeep
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Vikas Kumar
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Yong Xiao
- Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA; Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yanxiang Deng
- Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA; Yale Stem Cell Center and Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA; Yale Stem Cell Center and Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA; Human and Translational Immunology, Yale School of Medicine, New Haven, CT 06520, USA; Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Juan C Vasquez
- Department of Pediatrics, Yale School of Medicine, New Haven, CT 06520, USA
| | - Vijender Singh
- Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
| | - Raman Bahal
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA.
| |
Collapse
|
23
|
Sayin AZ, Abali Z, Senyuz S, Cankara F, Gursoy A, Keskin O. Conformational diversity and protein-protein interfaces in drug repurposing in Ras signaling pathway. Sci Rep 2024; 14:1239. [PMID: 38216592 PMCID: PMC10786864 DOI: 10.1038/s41598-023-50913-8] [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: 08/14/2023] [Accepted: 12/27/2023] [Indexed: 01/14/2024] Open
Abstract
We focus on drug repurposing in the Ras signaling pathway, considering structural similarities of protein-protein interfaces. The interfaces formed by physically interacting proteins are found from PDB if available and via PRISM (PRotein Interaction by Structural Matching) otherwise. The structural coverage of these interactions has been increased from 21 to 92% using PRISM. Multiple conformations of each protein are used to include protein dynamics and diversity. Next, we find FDA-approved drugs bound to structurally similar protein-protein interfaces. The results suggest that HIV protease inhibitors tipranavir, indinavir, and saquinavir may bind to EGFR and ERBB3/HER3 interface. Tipranavir and indinavir may also bind to EGFR and ERBB2/HER2 interface. Additionally, a drug used in Alzheimer's disease can bind to RAF1 and BRAF interface. Hence, we propose a methodology to find drugs to be potentially used for cancer using a dataset of structurally similar protein-protein interface clusters rather than pockets in a systematic way.
Collapse
Affiliation(s)
- Ahenk Zeynep Sayin
- Department of Chemical and Biological Engineering, College of Engineering, Koc University, Rumeli Feneri Yolu Sariyer, 34450, Istanbul, Turkey
| | - Zeynep Abali
- Graduate School of Science and Engineering, Computational Sciences and Engineering, Koc University, 34450, Istanbul, Turkey
| | - Simge Senyuz
- Graduate School of Science and Engineering, Computational Sciences and Engineering, Koc University, 34450, Istanbul, Turkey
| | - Fatma Cankara
- Graduate School of Science and Engineering, Computational Sciences and Engineering, Koc University, 34450, Istanbul, Turkey
| | - Attila Gursoy
- Department of Computer Engineering, Koc University, 34450, Istanbul, Turkey
| | - Ozlem Keskin
- Department of Chemical and Biological Engineering, College of Engineering, Koc University, Rumeli Feneri Yolu Sariyer, 34450, Istanbul, Turkey.
| |
Collapse
|
24
|
Zhang D, Shi X, Zheng W, Zhang X, Chen Y. Rare HER2 L796P missense mutation promotes the growth and oncogenic signaling in breast cancer cells. Proteomics Clin Appl 2024; 18:e2300061. [PMID: 37672800 DOI: 10.1002/prca.202300061] [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: 05/30/2023] [Accepted: 08/28/2023] [Indexed: 09/08/2023]
Abstract
PURPOSE This research aimed to find potential HER2 mutations that would have an impact on breast cancer and investigate the underlying mechanism. EXPERIMENTAL DESIGN This study first investigated 238 pairs of breast cancer and para-cancerous tissue samples from patients on the targeted next-generation sequencing (tNGS) platform. CCK-8 and clone formation assay were used to investigate whether the mutation exerts proliferative effects on breast cancer cells. In addition, mass spectrometry-based comparative proteomic and phosphoproteomic analyses of the mutation types and wild types of MCF-7 cell lines were carried out. RESULTS Among the identified mutations, a new mutation HER2 L796P promoted the proliferation of breast cancer cells and had resistance to lapatinib using CCK-8 cell proliferation assay and clone formation assay. The bioinformatic analysis showed that RAS family proteins and ERK phosphorylated proteins significantly increased in the L796P mutant cells. The Gene Ontology (GO) analysis revealed that L796P mutation affected the function of breast cancer at the level of upstream genes in the MAPK and PI3K-AKT-TOR pathways. CONCLUSIONS AND CLINICAL RELEVANCE This study demonstrated that a rare mutation HER2 L796P could be a potential therapeutic target for the clinical management of breast cancer.
Collapse
Affiliation(s)
- Dongxue Zhang
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Xiaoyu Shi
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Weimin Zheng
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Xian Zhang
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yun Chen
- School of Pharmacy, Nanjing Medical University, Nanjing, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing, China
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Nanjing, China
| |
Collapse
|
25
|
Mu Y, Meng Y, Du Y, Liu X, Zhang J. Clinical characteristics and treatment outcomes of HER2 mutation and HER2 fusion in 22 patients with advanced breast cancer. Thorac Cancer 2023; 14:3381-3388. [PMID: 37863840 PMCID: PMC10693943 DOI: 10.1111/1759-7714.15130] [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: 08/15/2023] [Accepted: 09/25/2023] [Indexed: 10/22/2023] Open
Abstract
BACKGROUND The clinical characteristics and efficacy of human epidermal growth factor receptor-2 (HER-2)-directed agents against HER2 mutations and HER2 fusions in breast cancer are obscure due to their low frequency. METHODS We conducted a retrospective study in patients with advanced breast cancer harboring HER2 mutations and/or HER2 fusions between January 1, 2017 and January 1, 2021. RESULTS Among a total of 22 patients, 17 HER2 mutations were detected, including L755S, S310F, R100=, V777L, R897W, T862A, 440-17C > G, H878Y, V842I, 73 + 9G > C, T278fs, E1069K, L755P, 226-11C > T, 574 + 12C>T, L114V and P128L. The majority of patients had ductal carcinoma, which mostly coexisted with HER2 amplification/overexpression. The median progression-free survival (PFS) of the 22 patients was 6.9 months (95% CI: 4.7, 9.1) in the first-line setting. The median PFS of patients who received first-line trastuzumab-based regimens was significantly longer than that of patients who received a first-line tyrosine kinase inhibitor (TKI) (10.8 months [95% CI: 2.9, 18.7] vs. 1.9 months [95% CI: 0.8, 3.0], p < 0.005). A total of 14 patients were treated with anti-HER2 antibody-drug conjugate (ADC), among whom the median treatment line of first-time of administration of anti-HER2 ADC was 4.5 (range, 1-10). Anti-HER2 ADC reached an objective response rate (ORR) of 42.9%, a disease control rate (DCR) of 85.7% and a median PFS of 7.3 months (95% CI: 4.4-10.1) from the first-time of administration. CONCLUSION Our data demonstrated the clinical benefit of anti-HER2 treatment in Chinese breast cancer patients harboring HER2 mutation and/or HER2 fusion. The value of immunotherapy and treatment selection among individual HER2 variants needs further study.
Collapse
Affiliation(s)
- Yuxin Mu
- Phase I Clinical Trial CenterFudan University Shanghai Cancer CenterShanghaiChina
| | - Yanchun Meng
- Phase I Clinical Trial CenterFudan University Shanghai Cancer CenterShanghaiChina
| | - Yiqun Du
- Phase I Clinical Trial CenterFudan University Shanghai Cancer CenterShanghaiChina
| | - Xiaojun Liu
- Phase I Clinical Trial CenterFudan University Shanghai Cancer CenterShanghaiChina
| | - Jian Zhang
- Phase I Clinical Trial CenterFudan University Shanghai Cancer CenterShanghaiChina
| |
Collapse
|
26
|
Zheng Y, Shen G, Zhang C, Huo X, Xin Y, Fang Q, Guan Y, Zhao F, Ren D, Liu Z, Wang M, Zhao J. Efficacy of anti-HER2 drugs in the treatment of patients with HER2-mutated cancers: a systematic review and meta-analysis. Clin Exp Med 2023; 23:3205-3216. [PMID: 37120775 DOI: 10.1007/s10238-023-01072-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 04/06/2023] [Indexed: 05/01/2023]
Abstract
Anti-human epidermal growth factor receptor-2 (anti-HER2) therapy has shown excellent efficacy in patients with HER2 overexpression and amplification. Although HER2 mutations are rarely expressed in several cancers, when they occur, they can activate the HER2 signaling pathway. In recent years, studies have shown that anti-HER2 drugs have promising efficacy in patients with HER2 mutations. Based on keywords, we searched databases, such as PubMed, Embase, and Cochrane Library, and the main conference abstracts. We extracted data on objective response rate (ORR), clinical benefit rate (CBR), duration of response (DOR), progression-free survival (PFS), and overall survival (OS) from studies on the efficacy of anti-HER2 therapies in patients with HER2-mutated cancers, and analyzed grade 3 or higher adverse events (AEs). We included 19 single-arm clinical studies and 3 randomized controlled trials (RCTs), containing a total of 1017 patients with HER2 mutations, involving seven drugs and nine cancers, and 18 studies enrolled a high proportion of heavily pretreated patients who had received multiple lines of therapy. Our results showed pooled ORR and CBR of 25.0% (range, 3.8-72.7%; 95% CI, 18-32%) and 36.0% (range, 8.3-63.0%; 95% CI, 31-42%) for anti-HER2 therapy in HER2-mutated cancers. The pooled median PFS, OS, DOR were 4.89 (95% CI, 4.16-5.62), 12.78 (95% CI, 10.24-15.32), and 8.12 (95% CI, 6.48-9.75) months, respectively. In a subgroup analysis, we analyzed the ORR for different cancers, showing 27.0, 25.0, 23.0, and 16.0% for breast, lung, cervical, and biliary tract cancers, respectively. ORR analyses were performed for different drugs as monotherapy or in combination, showing 60.0% for trastuzumab deruxtecan (T-DXd), 31.0% for pyrotinib, 26.0% for neratinib combined with trastuzumab, 25.0% for neratinib combined with fulvestrant, 19.0% for trastuzumab combined with pertuzumab, and 16.0% for neratinib. In addition, we found that diarrhoea, neutropenia, and thrombocytopenia were the most common grade ≥ 3 AEs associated with anti-HER2 therapeutic agents. In this meta-analysis of heavily pretreated patients with HER2 mutations, anti-HER2 therapies, DS-8201 and trastuzumab emtansine, showed promising efficacy and activity. Anti-HER2 therapies showed different efficacies in different or the same cancer settings and all had a tolerable safety profile.
Collapse
Affiliation(s)
- Yonghui Zheng
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital, Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Guoshuang Shen
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital, Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Chengrong Zhang
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital, Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Xingfa Huo
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital, Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Yuanfang Xin
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital, Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Qianqian Fang
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital, Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Yumei Guan
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital, Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Fuxing Zhao
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital, Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Dengfeng Ren
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital, Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Zhen Liu
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital, Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Miaozhou Wang
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital, Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Jiuda Zhao
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital, Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China.
| |
Collapse
|
27
|
Tian H, Qu M, Zhang G, Yuan L, Shi Q, Wang Y, Yang Y, Zhang Y, Qi X. Dramatic Response to Pyrotinib and T-DM1 in HER2-Negative Metastatic Breast Cancer With 2 Activating HER2 Mutations. Oncologist 2023:7147272. [PMID: 37120151 DOI: 10.1093/oncolo/oyad122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/28/2023] [Indexed: 05/01/2023] Open
Abstract
HER2 signaling is activated in response to somatic HER2 mutations, which are often found in invasive lobular breast cancer (ILC) and are associated with poor prognosis. Tyrosine kinase inhibitors (TKIs) have demonstrated considerable antitumor activity in patients with HER2-mutated advanced breast cancer (BC). Further, several clinical trials have indicated that HER2-targeted antibody-drug conjugates (ADCs) exhibit promising efficacy in lung cancer with HER2 mutations, and the efficacy of ADCs against HER2-mutated BC is currently being evaluated. Several preclinical studies have demonstrated that the therapeutic efficacy of ADCs in HER2-mutated cancer can be enhanced by the addition of irreversible TKIs, but the potential of such a combined treatment regimen for the treatment of HER2-mutated BC has not been reported. Herein, we describe a case in which a patient with estrogen receptor-positive/HER2-negative metastatic ILC with 2 activating HER2 mutations (D769H and V777L) exhibited a significant and durable response to anti-HER2 treatment with pyrotinib (an irreversible TKI) in combination with ado-trastuzumab emtansine, which was administered after multiple lines of therapy that had resulted in disease progression. Further, based on the evidence from the present case, TKI plus ADC seems to be a promising combination anti-HER2 regimen for patients with HER2-negative/HER2-mutated advanced BC, although further rigorous studies are warranted to confirm these findings.
Collapse
Affiliation(s)
- Hao Tian
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Man Qu
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Guozhi Zhang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Long Yuan
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Qiyun Shi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Yinhuan Wang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Ying Yang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Yi Zhang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Xiaowei Qi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| |
Collapse
|
28
|
Han C, McNamara B, Bellone S, Harold J, Manara P, Hartwich TMP, Mutlu L, Yang-Hartwich Y, Zipponi M, Demirkiran C, Verzosa SM, Altwerger G, Ratner E, Huang GS, Clark M, Andikyan V, Azodi M, Dottino PR, Schwartz PE, Santin AD. The Poly (ADP-ribose) polymerase inhibitor olaparib and pan-ErbB inhibitor neratinib are highly synergistic in HER2 overexpressing epithelial ovarian carcinoma in vitro and in vivo. Gynecol Oncol 2023; 170:172-178. [PMID: 36706643 PMCID: PMC10023457 DOI: 10.1016/j.ygyno.2023.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/22/2022] [Accepted: 01/16/2023] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Ovarian cancer (OC) is associated with the highest gynecologic cancer mortality. The development of novel, effective combinations of targeted therapeutics remains an unmet medical need. We evaluated the preclinical efficacy of the Poly (ADP-ribose) polymerase (PARP) inhibitor (olaparib) and the pan-ErbB inhibitor (neratinib) as single agents and in combination in ovarian cancer cell lines and xenografts with variable HER2 expression. METHODS In vitro cell viability with olaparib, neratinib, and their combination was assessed using flow-cytometry based assays against a panel of OC primary cell lines with variable HER2 expression. Immunoblotting experiments were performed to elucidate the mechanism of activity and synergism. The in vivo antitumor activity of the olaparib/neratinib combination versus single agents was tested in HER2 positive xenograft OC models. RESULTS HER2 + OC cell lines demonstrated higher sensitivity to olaparib and neratinib when compared to HER2 negative tumors (i.e., IC50: 2.06 ± 0.33 μM vs. 39.28 ± 30.51 μM, p = 0.0035 for olaparib and 19.42 ± 2.63 nM vs. 235.0 ± 165.0 nM, p = 0.0035 for neratinib). The combination of olaparib with neratinib was more potent when compared to single-agent olaparib or neratinib both in vitro and in vivo, and demonstrated synergy in all primary HER2 + OC models. Western blot experiments showed neratinib decreased pHER2/neu while increased Poly(ADP-ribose) (PAR) enzymatic activity; olaparib increased pHER2/Neu expression and blocked PAR activatio. Olaparib/neratinib in combination decreased both pHER2/Neu as well as PAR activation. CONCLUSION The combination of olaparib and neratinib is synergistic and endowed with remarkable preclinical activity against HER2+ ovarian cancers. This combination may represent a novel therapeutic option for ovarian cancer patients with HER2+, homologous recombination-proficient tumors resistant to chemotherapy.
Collapse
Affiliation(s)
- Chanhee Han
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, CT 06520, USA
- Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA 30322
| | - Blair McNamara
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, CT 06520, USA
| | - Stefania Bellone
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, CT 06520, USA
| | - Justin Harold
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, CT 06520, USA
| | - Paola Manara
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, CT 06520, USA
| | - Tobias Max Philipp Hartwich
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, CT 06520, USA
| | - Levent Mutlu
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, CT 06520, USA
| | - Yang Yang-Hartwich
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, CT 06520, USA
| | - Margherita Zipponi
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, CT 06520, USA
| | - Cem Demirkiran
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, CT 06520, USA
| | - Skylar Miguel Verzosa
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, CT 06520, USA
| | - Gary Altwerger
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, CT 06520, USA
| | - Elena Ratner
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, CT 06520, USA
| | - Gloria S. Huang
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, CT 06520, USA
| | - Mitchell Clark
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, CT 06520, USA
| | - Vaagn Andikyan
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, CT 06520, USA
| | - Masoud Azodi
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, CT 06520, USA
| | - Peter R. Dottino
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, CT 06520, USA
| | - Peter E. Schwartz
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, CT 06520, USA
| | - Alessandro D. Santin
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, CT 06520, USA
| |
Collapse
|
29
|
King DA, Weiel JJ, Reyes R, Mills M, Itchon A, Fisher GA, Ford JM, Suarez CJ. Therapeutic Implications of Oncogenic Missense HER2 ( ERBB2) Mutations in Gastric Adenocarcinoma. JCO Precis Oncol 2023; 7:e2200093. [PMID: 36787506 DOI: 10.1200/po.22.00093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Affiliation(s)
- Daniel A King
- Northwell Health Cancer Institute and Feinstein Institute of Research, Lake Success, NY
| | - Julianna J Weiel
- Department of Pathology, Stanford University School of Medicine, Stanford, CA.,Department of Pathology, Billings Clinic, Billings, MT
| | - Rochelle Reyes
- Early Drug Development Group, Stanford Cancer Institute, Stanford, CA
| | - Meredith Mills
- Division of Oncology, Department of Internal Medicine, Stanford University, Stanford, CA
| | - Antonita Itchon
- Division of Oncology, Department of Internal Medicine, Stanford University, Stanford, CA
| | - George A Fisher
- Division of Oncology, Department of Internal Medicine, Stanford University, Stanford, CA
| | - James M Ford
- Division of Oncology, Department of Internal Medicine, Stanford University, Stanford, CA
| | - Carlos J Suarez
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
30
|
Sun Q, Li Q, Gao F, Wu H, Fu Y, Yang J, Fan X, Cui X, Pu X. HER2 overexpression/amplification status in colorectal cancer: a comparison between immunohistochemistry and fluorescence in situ hybridization using five different immunohistochemical scoring criteria. J Cancer Res Clin Oncol 2023; 149:579-592. [PMID: 36018511 PMCID: PMC9931822 DOI: 10.1007/s00432-022-04230-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/19/2022] [Indexed: 10/15/2022]
Abstract
OBJECTIVE Although HER2 has gradually become an important therapeutic target for colorectal cancer (CRC), a unified and standard HER2 scoring system was still not established in CRC, and the debatable results of immunohistochemistry and fluorescence in situ hybridization (FISH) in CRC requires further exploration. METHODS In this study, we use five immunohistochemical (IHC) scoring criteria (i.e., IRS-p, IRS-m, GEA-s, GEA-b and HERACLES) and two FISH criteria to evaluate HER2 status, and further evaluate the correlation between HER2 status and clinicopathological features, survival in a large, unselected Chinese cohort of 664 CRCs. RESULTS Finally, we set HER2/CEP17 ratio ≥ 2.0, or an average HER2 copy number ≥ 6.0 as FISH-positive threshold and the amplification rate of HER2 gene was 7.08% (47/664).The HER2 positivity (IHC 3+) was 2.71%, 3.16%, 2.56%, 2.71% and 3.16%, according to the IHC scoring criteria of IRS-p, IRS-m, GEA-s, GEA-b and HERACLES, respectively. Set FISH results as the golden standard; receiver-operating characteristic analysis showed that IRS-p had both high sensitivity and specificity than other IHC scoring systems to evaluate HER2 status. Based on IRS-p criterion, There were significant differences in tumor differentiation (p = 0.038), lymphatic vascular invasion (p = 0.001), pN stage (p value = 0.043), and overall survival (p < 0.001) among IHC score 0-3 + groups. Meanwhile, there were significant differences in pT stage (p = 0.031), pN stage (p = 0.009) and overall survival (p < 0.001) among FISH subgroups. CONCLUSION The IRS-p criterion was more suitable for assessing the HER2 status in CRC patients than other IHC criteria. Whereas for FISH scoring system, only HER2/CEP17 < 2.0, meanwhile HER2cn < 4.0 and HER2cn ≥ 6.0 were subgroups with unique clinicopathological characteristics.
Collapse
Affiliation(s)
- Qi Sun
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhong-Shan Road, Nanjing, 210008, Jiangsu, China
- Center for Digestive Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Qi Li
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhong-Shan Road, Nanjing, 210008, Jiangsu, China
| | - Fuping Gao
- Department of Pathology, Gaochun People's Hospital, Nanjing, 210008, Jiangsu, China
| | - Hongyan Wu
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhong-Shan Road, Nanjing, 210008, Jiangsu, China
| | - Yao Fu
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhong-Shan Road, Nanjing, 210008, Jiangsu, China
| | - Jun Yang
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhong-Shan Road, Nanjing, 210008, Jiangsu, China
| | - Xiangshan Fan
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhong-Shan Road, Nanjing, 210008, Jiangsu, China.
| | - Xiaobin Cui
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhong-Shan Road, Nanjing, 210008, Jiangsu, China.
| | - Xiaohong Pu
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhong-Shan Road, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
31
|
An anti-EGFR antibody-drug conjugate overcomes resistance to HER2-targeted drugs. Cancer Lett 2023; 554:216024. [PMID: 36455759 DOI: 10.1016/j.canlet.2022.216024] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/02/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022]
Abstract
Trastuzumab-emtansine (T-DM1) is an antibody-drug conjugate (ADC) that was approved in 2013 to treat HER2+ breast cancer. Despite its efficacy in the clinic, some patients exhibit intrinsic or acquired resistance to such ADC. To characterize mechanisms of resistance to T-DM1, we isolated several HER2+ resistant clones derived from the HCC1954 HER2+ cell line. The isolated clones were different as per their transcriptomic profiles. However, all the T-DM1-resistant clones showed decreased HER2 levels. Yet, the clones were still oncogenically dependent on HER2, as indicated by knock down experiments. The decrease in HER2 expression caused acquired resistance to T-DM1 and to other anti-HER2 therapies. Antibody array analyses showed that the epidermal growth factor receptor (EGFR) was expressed in these T-DM1-resistant HCC1954 clones. Indeed, therapies targeting EGFR, particularly cetuximab-DM1, demonstrated a strong anti-proliferative action on cells with acquired resistance to T-DM1 and HER2 loss. The expression of EGFR in cells resistant to T-DM1 offers the possibility of using therapies directed to this receptor to combat resistance to anti-HER2 drugs and loss of HER2 overexpression.
Collapse
|
32
|
Shang Y, Mo J, Huo R, Li X, Fang G, Wei Z, Gu G, Zhu X, Zhang C, Liu C, Yan D. Investigation of the prevalence and clinical implications of ERBB2 exon 16 skipping mutations in Chinese pan-cancer patients. Front Oncol 2023; 12:1064598. [PMID: 36686783 PMCID: PMC9859631 DOI: 10.3389/fonc.2022.1064598] [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: 10/08/2022] [Accepted: 12/13/2022] [Indexed: 01/08/2023] Open
Abstract
Background Although rare, ERBB2 exon 16 skipping mutations (ERBB2ΔEx16) have been implicated in resistance to anti-HER2 and anti-EGFR targeted agents. Our study investigated the prevalence and clinical significance of ERBB2ΔEx16 in Chinese pan-cancer patients. Methods We retrospectively screened 40996 patients, spanning 19 cancer types, who had available genomic profiles acquired with DNA-based next-generation sequencing (NGS). We characterized the clinical and molecular features of the ERBB2ΔEx16-positive patients. Furthermore, we also analyzed a pan-cancer dataset from the Cancer Genome Atlas (TCGA; n=8705). Results A total of 22 patients were detected with ERBB2ΔEx16, resulting in an overall prevalence rate of 0.054% (22/40996). Of them, 16 patients had lung cancer (LC; 0.05%, 16/30890), five patients had gastric cancer (GC; 0.35%, 5/1448), and one patient had ovarian cancer (0.12%, 1/826). Among the 16 LC patients, ERBB2ΔEx16 was detected in four treatment-naïve EGFR/ALK-negative patients and 12 EGFR-positive patients after the onset of resistance to EGFR tyrosine kinase inhibitors (TKIs). The treatment-naïve patients harbored no LC-associated oncogenic drivers except ERBB2 amplification, suggesting a potential oncogenic role for ERBB2ΔEx16. Consistently, ERBB2ΔEx16+ patients from TCGA data also carried no known drivers despite various concurrent alterations. In the 12 EGFR TKI-resistant LC patients, relative variant frequencies for ERBB2ΔEx16 were lower than in untreated patients, suggesting ERBB2ΔEx16 as secondary alterations following TKI treatment and thereby implicating ERBB2ΔEx16 in mediating therapeutic resistance. Conclusions Our study identified an overall ERBB2ΔEx16 prevalence rate of 0.054% and provided insights into the clinical implications of ERBB2ΔEx16 in Chinese pan-cancer patients.
Collapse
Affiliation(s)
- Yanhong Shang
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, China
| | - Jianming Mo
- Department of Pulmonary and Critical Care Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Ran Huo
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, China
| | - Xiaofang Li
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, China
| | - Guotao Fang
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, China
| | - Zichun Wei
- Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Guomin Gu
- Second Department of Pulmonary Medicine, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, China
| | - Xiaodan Zhu
- Second Department of Pulmonary Medicine, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, China
| | - Chan Zhang
- Department of Oncology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Chunling Liu
- Department of Pulmonary Medicine, Affiliated Cancer Hospital of Xinjiang Medical, Urumqi, China,*Correspondence: Dong Yan, ; Chunling Liu,
| | - Dong Yan
- Department of Oncology, Beijing Luhe Hospital, Capital Medical University, Beijing, China,*Correspondence: Dong Yan, ; Chunling Liu,
| |
Collapse
|
33
|
Zagami P, Nicolò E, Corti C, Valenza C, Curigliano G. New Concepts in Cardio-Oncology. Cancer Treat Res 2023; 188:303-341. [PMID: 38175351 DOI: 10.1007/978-3-031-33602-7_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Cancer and cardiovascular disease are the two major causes of morbidity and mortality in worldwide. Discovering new therapeutic agents for the management of breast cancer (BC) has increased the numbers of cancer survivors but with the risk of cardiovascular adverse events (CV-AEs). All drugs can potentially damage the cardiovascular system, with different types of clinical manifestations from ischemic myocardial disease to vasculitis, thrombosis or pericarditis. An early detection of CV-AEs guarantees an earlier treatment, which is associated with better outcomes. Cardio-oncology field enlarged its studies to improve prevention, monitoring and treatment of all cardiotoxic manifestations related to old or modern oncological agents. A multidisciplinary approach with a close partnership between oncologists and cardiologists is essential for an optimal management and therapeutic decision-making. The aim of this chapter is to review all types of cardiotoxic manifestations related to novel and old agents approved for treatment of BC patients including chemotherapy, anti-HER2 agents, cyclin-dependent kinase 4/6 inhibitors, PolyADP-ribose polymerase (PARP) inhibitors, antiangiogenic drugs and immunotherapy. We also focused our discussion on prevention, monitoring, treatment, and management of CV-AEs.
Collapse
Affiliation(s)
- Paola Zagami
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Milan, Italy.
- Department of Oncology and Hematology, University of Milano, Milan, Italy.
| | - Eleonora Nicolò
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology, University of Milano, Milan, Italy
| | - Chiara Corti
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology, University of Milano, Milan, Italy
| | - Carmine Valenza
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology, University of Milano, Milan, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology, University of Milano, Milan, Italy
| |
Collapse
|
34
|
Effects of Human Papilloma Virus E6/E7 Oncoproteins on Genomic Structure in Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2022; 14:cancers14246190. [PMID: 36551675 PMCID: PMC9777059 DOI: 10.3390/cancers14246190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Human Papilloma Virus (HPV) is highly prevalent within the U.S., with studies estimating that over 80% of individuals will contract the virus in their lifetime. HPV is considered a primary risk factor for the development and progression of oropharyngeal cancers. The impact of the HPV virus's E6 and E7 oncoproteins on cellular signaling pathways and genomic integration has been extensively characterized. Indirect genomic effects; however, remain relatively unidentified. In this study, we analyzed 83 HPV+ Head and Neck Squamous Cell Carcinoma (HNSCC) patients of varying HPV types. Expression counts of the HPV E6 and E7 oncogenes were estimated across samples and correlated with genomic mutational classes. High expression of E6 and E7 oncoproteins was associated with a greater number of total point mutations, especially on chromosomes 1, 11, and 17, which have been implicated in HPV-mediated cancers in previous studies. Samples with high E6 and E7 expression also exhibited more frequent non-clustered structural variation and a lack of clustered variation altogether. Copy number segments were present with fewer number of repeats in high E6 and E7 expression samples, which is known to correlate with decreased expression of affected genes. E6 and E7 expression was associated with increased activity of several cellular pathways associated in oncogenesis and telomere maintenance. In comprehensively characterizing the effects of the HPV oncoproteins on the human genome, potential mechanisms of HNSCC pathogenesis may be further elucidated.
Collapse
|
35
|
Deciphering the Impact of HER2 Alterations on Non-Small-Cell Lung Cancer: From Biological Mechanisms to Therapeutic Approaches. J Pers Med 2022; 12:jpm12101651. [PMID: 36294789 PMCID: PMC9605102 DOI: 10.3390/jpm12101651] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
Despite the recent increase in the number of types of treatments, non-small-cell lung cancer (NSCLC) remains the major cause of death from cancer worldwide. So, there is an urgent need to develop new therapeutic strategies. The HER2 gene codes for tyrosine kinase receptor whose alterations are known to drive carcinogenesis. HER2 alterations, including amplification, mutations, and overexpression, have been mainly described in breast and gastric cancers, but up to 4% of NSCLC harbor actionable HER2 mutations. HER2-targeted therapy for NSCLC with trastuzumab, pertuzumab, and trastuzumab emtansine has failed to demonstrate an improvement in survival. Nevertheless, recent data from phase II trials have shed light on promising specific therapies for HER2-mutant NSCLC such as trastuzumab deruxtecan. Herein, we aimed to provide an updated review on the biology, epidemiology, molecular testing, and therapeutic strategies for NSCLC with HER2 molecular alterations.
Collapse
|
36
|
Sun S, Ren L, Miao Z, Hua L, Wang D, Deng J, Chen J, Liu N, Gong Y. Application of MRI-Based Radiomics in Preoperative Prediction of NF2 Alteration in Intracranial Meningiomas. Front Oncol 2022; 12:879528. [PMID: 36267986 PMCID: PMC9578175 DOI: 10.3389/fonc.2022.879528] [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: 02/19/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
PurposeThis study aimed to investigate the feasibility of predicting NF2 mutation status based on the MR radiomic analysis in patients with intracranial meningioma.MethodsThis retrospective study included 105 patients with meningiomas, including 60 NF2-mutant samples and 45 wild-type samples. Radiomic features were extracted from magnetic resonance imaging scans, including T1-weighted, T2-weighted, and contrast T1-weighted images. Student’s t-test and LASSO regression were performed to select the radiomic features. All patients were randomly divided into training and validation cohorts in a 7:3 ratio. Five linear models (RF, SVM, LR, KNN, and xgboost) were trained to predict the NF2 mutational status. Receiver operating characteristic curve and precision-recall analyses were used to evaluate the model performance. Student’s t-tests were then used to compare the posterior probabilities of NF2 mut/loss prediction for patients with different NF2 statuses.ResultsNine features had nonzero coefficients in the LASSO regression model. No significant differences was observed in the clinical features. Nine features showed significant differences in patients with different NF2 statuses. Among all machine learning algorithms, SVM showed the best performance. The area under curve and accuracy of the predictive model were 0.85; the F1-score of the precision-recall curve was 0.80. The model risk was assessed by plotting calibration curves. The p-value for the H-L goodness of fit test was 0.411 (p> 0.05), which indicated that the difference between the obtained model and the perfect model was statistically insignificant. The AUC of our model in external validation was 0.83.ConclusionA combination of radiomic analysis and machine learning showed potential clinical utility in the prediction of preoperative NF2 status. These findings could aid in developing customized neurosurgery plans and meningioma management strategies before postoperative pathology.
Collapse
Affiliation(s)
- Shuchen Sun
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Neurosurgery, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
| | - Leihao Ren
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Neurosurgery, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
| | - Zong Miao
- Department of Neurosurgery, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Lingyang Hua
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Neurosurgery, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
| | - Daijun Wang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Neurosurgery, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
| | - Jiaojiao Deng
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Neurosurgery, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
| | - Jiawei Chen
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Neurosurgery, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
| | - Ning Liu
- Department of Neurosurgery, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Ye Gong
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Neurosurgery, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai, China
- *Correspondence: Ye Gong,
| |
Collapse
|
37
|
Gámez-Chiachio M, Sarrió D, Moreno-Bueno G. Novel Therapies and Strategies to Overcome Resistance to Anti-HER2-Targeted Drugs. Cancers (Basel) 2022; 14:4543. [PMID: 36139701 PMCID: PMC9496705 DOI: 10.3390/cancers14184543] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/09/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
The prognosis and quality of life of HER2 breast cancer patients have significantly improved due to the crucial clinical benefit of various anti-HER2 targeted therapies. However, HER2 tumors can possess or develop several resistance mechanisms to these treatments, thus leaving patients with a limited set of additional therapeutic options. Fortunately, to overcome this problem, in recent years, multiple different and complementary approaches have been developed (such as antibody-drug conjugates (ADCs)) that are in clinical or preclinical stages. In this review, we focus on emerging strategies other than on ADCs that are either aimed at directly target the HER2 receptor (i.e., novel tyrosine kinase inhibitors) or subsequent intracellular signaling (e.g., PI3K/AKT/mTOR, CDK4/6 inhibitors, etc.), as well as on innovative approaches designed to attack other potential tumor weaknesses (such as immunotherapy, autophagy blockade, or targeting of other genes within the HER2 amplicon). Moreover, relevant technical advances such as anti-HER2 nanotherapies and immunotoxins are also discussed. In brief, this review summarizes the impact of novel therapeutic approaches on current and future clinical management of aggressive HER2 breast tumors.
Collapse
Affiliation(s)
- Manuel Gámez-Chiachio
- Biochemistry Department, Medicine Faculty, Universidad Autónoma Madrid-CSIC, IdiPaz, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red-Oncología (CIBERONC), 28029 Madrid, Spain
| | - David Sarrió
- Biochemistry Department, Medicine Faculty, Universidad Autónoma Madrid-CSIC, IdiPaz, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red-Oncología (CIBERONC), 28029 Madrid, Spain
| | - Gema Moreno-Bueno
- Biochemistry Department, Medicine Faculty, Universidad Autónoma Madrid-CSIC, IdiPaz, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red-Oncología (CIBERONC), 28029 Madrid, Spain
- MD Anderson International Foundation, 28033 Madrid, Spain
| |
Collapse
|
38
|
Li S, Liu Q, Liu M, Liu T. Response to Pyrotinib in a Patient with Metastatic Bladder Urothelial Carcinoma Harboring HER2 V842I Mutation: A Case Report. Cancer Manag Res 2022; 14:2927-2932. [PMID: 36200096 PMCID: PMC9528799 DOI: 10.2147/cmar.s365951] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/10/2022] [Indexed: 12/19/2022] Open
Abstract
Background The highest incidence of human epidermal growth factor receptor 2 (HER2) mutations has been observed in bladder cancer (BC). However, the function of HER2 mutation in tumor progression and metastasis remains unclear. Currently, no responses to the pan-HER kinase inhibitor were observed in HER2-mutant BC. Case Presentation We described a patient with metastatic bladder urothelial carcinoma (BUC) carrying a HER2 V842I mutation both in circulating tumor DNA (ctDNA) and biopsy sample. The patient was then treated with a HER2 tyrosine kinase inhibitor, pyrotinib, and responded well. However, the targeting treatment was terminated due to G3 diarrhea. Reduced dose of pyrotinib was later added to late-line treatment, the patient’s tumor again responded with a significant decrease in CA199. Conclusion This is the first reported case of HER2 V842I mutation successfully treated with pyrotinib in BUC, suggesting pyrotinib therapy might serve as a therapeutic option for BUC patients harboring HER2 activating mutation.
Collapse
Affiliation(s)
- Suyao Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Qing Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Mengling Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
- Correspondence: Tianshu Liu, Department of Medical Oncology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 20032, People’s Republic of China, Email
| |
Collapse
|
39
|
Yu H, Tai Q, Yang C, Weng L, Gao M, Zhang X. Counting Protein Number in a Single Cell by a Picoliter Liquid Operating Technology. Anal Chem 2022; 94:11925-11933. [PMID: 35980697 DOI: 10.1021/acs.analchem.2c02701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ultra-low-copy number proteins play a crucial role in exploring cellular heterogeneity and the insight of protein biomarkers in a single cell. However, counting ultra-low-copy number target proteins in a single cell remains a grand challenge. Herein, we developed a so-called single-cell picoliter liquid operating technology for counting target proteins in a single cell. An ingenious volume-controllable sampling technique was employed to capture a single cell for subsequent analysis. Remarkably, 50 pL of sample volume was employed for sample preparation, single-cell capture, in-droplet lysis, and target protein immobilization on a functionalized coverslip in a monolayer. Then, target protein antibodies coupled with quantum dots were added and incubated to label those immobilized proteins. After clean-up, a single-view image under 100× objective was taken, and the 80 × 80 μm2 view image was then applied to count the precise copy number of the target proteins in the single cell. Furthermore, good linearity and repeatability were achieved for ultra-low-copy number proteins, ranging from 1 to 1500. Finally, the expression level of human epidermal growth factor receptor 2 in single cells from both MCF-7 and MDA-MB-231 cell lines was also analyzed. In a word, this work stimulated the development of capillary-based single-cell analysis and updated the connotation of counting ultra-low-copy number proteins.
Collapse
Affiliation(s)
- Hailong Yu
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - Qunfei Tai
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - Chenjie Yang
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - Lingxiao Weng
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - Mingxia Gao
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - Xiangmin Zhang
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| |
Collapse
|
40
|
Wilding B, Scharn D, Böse D, Baum A, Santoro V, Chetta P, Schnitzer R, Botesteanu DA, Reiser C, Kornigg S, Knesl P, Hörmann A, Köferle A, Corcokovic M, Lieb S, Scholz G, Bruchhaus J, Spina M, Balla J, Peric-Simov B, Zimmer J, Mitzner S, Fett TN, Beran A, Lamarre L, Gerstberger T, Gerlach D, Bauer M, Bergner A, Schlattl A, Bader G, Treu M, Engelhardt H, Zahn S, Fuchs JE, Zuber J, Ettmayer P, Pearson M, Petronczki M, Kraut N, McConnell DB, Solca F, Neumüller RA. Discovery of potent and selective HER2 inhibitors with efficacy against HER2 exon 20 insertion-driven tumors, which preserve wild-type EGFR signaling. NATURE CANCER 2022; 3:821-836. [PMID: 35883003 DOI: 10.1038/s43018-022-00412-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 06/16/2022] [Indexed: 06/15/2023]
Abstract
Oncogenic alterations in human epidermal growth factor receptor 2 (HER2) occur in approximately 2% of patients with non-small cell lung cancer and predominantly affect the tyrosine kinase domain and cluster in exon 20 of the ERBB2 gene. Most clinical-grade tyrosine kinase inhibitors are limited by either insufficient selectivity against wild-type (WT) epidermal growth factor receptor (EGFR), which is a major cause of dose-limiting toxicity or by potency against HER2 exon 20 mutant variants. Here we report the discovery of covalent tyrosine kinase inhibitors that potently inhibit HER2 exon 20 mutants while sparing WT EGFR, which reduce tumor cell survival and proliferation in vitro and result in regressions in preclinical xenograft models of HER2 exon 20 mutant non-small cell lung cancer, concomitant with inhibition of downstream HER2 signaling. Our results suggest that HER2 exon 20 insertion-driven tumors can be effectively treated by a potent and highly selective HER2 inhibitor while sparing WT EGFR, paving the way for clinical translation.
Collapse
Affiliation(s)
| | | | | | - Anke Baum
- Boehringer Ingelheim RCV, Vienna, Austria
| | | | | | | | | | | | | | - Petr Knesl
- Boehringer Ingelheim RCV, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Gerd Bader
- Boehringer Ingelheim RCV, Vienna, Austria
| | | | | | | | | | - Johannes Zuber
- Institute of Molecular Pathology (IMP), Vienna, Austria
- Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Yan F, Rinn KJ, Kullnat JA, Wu AY, Ennett MD, Scott EL, Kaplan HG. Response of Leptomeningeal Metastasis of Breast Cancer With a HER2/neu Activating Variant to Tucatinib: A Case Report. J Natl Compr Canc Netw 2022; 20:745-752. [PMID: 35405660 DOI: 10.6004/jnccn.2022.7006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 01/24/2022] [Indexed: 11/17/2022]
Abstract
Metastatic breast cancer demonstrates HER2/neu amplification approximately 15% of the time. However, HER2 mutations, which often stimulate tumor growth, occur in only 3% to 5% of patients, and are seen more frequently in metastatic versus primary tumors. They are more frequent in lobular carcinoma, including triple-negative lobular cancer. Many of these variants are resistant to trastuzumab and lapatinib. However, neratinib can be efficacious, and recent data suggest that antibody-drug conjugates (ADCs) such as ado-trastuzumab emtansine (T-DM1) and trastuzumab deruxtecan may also be helpful. Laboratory and clinical data raise the possibility that simultaneous treatment with ADCs plus neratinib may be even more efficacious. Tucatinib, which has demonstrated significant activity in the central nervous system, has also been shown in vitro to be active against a number of these HER2 variants. This report describes a patient with metastatic estrogen receptor-positive, HER2-nonamplified breast cancer with an activating HER2 mutation whose tumor became resistant to neratinib as well as capecitabine, but whose subsequent leptomeningeal disease had a dramatically successful response to tucatinib plus capecitabine. As the frequency of HER2 mutations increases during the evolution of metastatic breast cancer, it is important to obtain genomic evaluation on these tumors with either repeat tissue or liquid biopsy as they progress over time.
Collapse
Affiliation(s)
| | | | | | - Aimee Y Wu
- 3University of California Los Angeles, Los Angeles, California
| | | | | | | |
Collapse
|
42
|
Yin Y, Song L, Shi D, Liu B, Li X, Yang M, Liu B, Wang D, Qin J. Identification of Recurrent Insertions and Deletions in Exon 18 and 19 of Human Epidermal Growth Factor Receptor 2 as Potential Drivers in Non-Small-Cell Lung Cancer and Other Cancer Types. JCO Precis Oncol 2022; 6:e2100325. [PMID: 35171661 PMCID: PMC8865527 DOI: 10.1200/po.21.00325] [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] [Indexed: 12/02/2022] Open
Abstract
PURPOSE Human epidermal growth factor receptor 2 (HER2) belongs to the same family as epidermal growth factor receptor (EGFR) and is known as an important cancer driver gene. Insertions and deletions (indels) are frequent driver mutations in both EGFR and HER2. The most common HER2 indels are the exon 20 insertions within the kinase domain, while others are rarely reported. Our study aimed to investigate other indels of HER2 that may act as driver mutations in Chinese patients with different cancer types. METHODS In this retrospective study, patient samples were subjected to targeted sequencing covering HER2 and other cancer-related genes. Mutation profiles of patients harboring HER2 exon 18/19 indels were described. Identified HER2 exon 18/19 indels in our study were compared with external data from COSMIC. In silico and in vitro analyses were performed on selected indels of HER2 exon 18 and 19, respectively. RESULTS A total of 25 indels in HER2 exon 18/19, 17 of which being recurrent, were identified in 20 of 53,591 patients with lung cancer (0.037%), two of 5,888 patients with colorectal cancer (0.034%), two of 3,774 patients with breast cancer (0.053%), and one of 14 patients with urothelial carcinoma of the renal pelvis (7.1%). Most patients harboring HER2 exon 18/19 indels were absent of known driver mutations. In lung cancer, mutation profiles were comparable between patients carrying HER2 exon 18/19 indels and the two established HER2 drivers (exon 20 insertions and S310 mutations). The in silico and in vitro analyses suggested an activated state conferred by HER2 exon 18/19 indels, which could be targeted by different tyrosine kinase inhibitors. CONCLUSION Our study revealed a class of rare but unique indels in HER2 exon 18/19, which may act as driver mutations in several cancer types. HER2 exon 18/19 indels identified in a small subset of patients may act as a novel class of cancer drivers. ![]() ![]()
Collapse
Affiliation(s)
- Yan Yin
- Department of Respiratory and Critical Medicine, Tianjin Chest Hospital, Tianjin, China
| | - Lijie Song
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dongsheng Shi
- Department of Respiratory and Critical Medicine, Tianjin Chest Hospital, Tianjin, China
| | - Bin Liu
- Department of Respiratory and Critical Medicine, Tianjin Chest Hospital, Tianjin, China
| | - Xiangke Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Minjie Yang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bihao Liu
- Department of Urology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dejuan Wang
- Department of Urology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jianwen Qin
- Department of Respiratory and Critical Medicine, Tianjin Chest Hospital, Tianjin, China
| |
Collapse
|
43
|
Cen S, Xu H, Liu Z, Zhao R, Pan H, Han W. Immune microenvironment characteristics and their implications for immune checkpoint inhibitor efficacy in HER2-overexpressing gastric cancer. Clin Exp Immunol 2022; 207:318-328. [PMID: 35553632 PMCID: PMC9113110 DOI: 10.1093/cei/uxac007] [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: 09/28/2021] [Revised: 12/14/2021] [Accepted: 02/02/2022] [Indexed: 02/06/2023] Open
Abstract
HER2-positive gastric cancer is a distinct tumor subtype, accounting for ~10% of gastric cancer cases. It is characterized by HER2 overexpression and responds well to HER2-targeting therapies. Recently, the addition of immune checkpoint inhibitors to HER2-targeting therapies produced satisfactory outcomes in these patients. In the present study, we used gene expression profiles and patient surgical sections to analyze the tumor immune microenvironment characteristics of gastric tumors with high HER2 expression. Several differentially enriched pathways were identified between the HER2 high-expression group and the low-expression group, such as pathways related to cytokine-cytokine receptor interactions, calcium signaling, and cell adhesion molecules. Tumors with high HER2 expression comprised fewer stromal cells and fewer immune cells, and had higher tumor purity. They also presented with lower expression of PD-1, PD-L1, CTLA-4, TIGIT, and LAG-3. In conclusion, our study provides a comprehensive blueprint of the immune microenvironment of HER2-positive gastric tumors. This analysis highlights the importance of considering the tumor microenvironment when assessing response to immune checkpoint inhibitors.
Collapse
Affiliation(s)
| | | | | | - Rongjie Zhao
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People’s Republic of China
| | - Hongming Pan
- Hongming Pan, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3# East Qinchun Road, Hangzhou, Zhejiang 310016, China.
| | - Weidong Han
- Correspondence: Weidong Han, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3# East Qinchun Road, Hangzhou, Zhejiang 310016, China.
| |
Collapse
|
44
|
Corti C, Giachetti PPMB, Eggermont AMM, Delaloge S, Curigliano G. Therapeutic vaccines for breast cancer: Has the time finally come? Eur J Cancer 2022; 160:150-174. [PMID: 34823982 PMCID: PMC8608270 DOI: 10.1016/j.ejca.2021.10.027] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 12/15/2022]
Abstract
The ability to exploit the immune system as a weapon against cancer has revolutionised the treatment of cancer patients, especially through immune checkpoint inhibitors (ICIs). However, ICIs demonstrated a modest benefit in treating breast cancer (BC), with the exception of certain subsets of triple-negative BCs. An immune-suppressive tumour microenvironment (TME), typically present in BC, is an important factor in the poor response to immunotherapy. After almost two decades of poor clinical trial results, cancer vaccines (CVs), an active immunotherapy, have come back in the spotlight because of some technological advancements, ultimately boosted by coronavirus disease 2019 pandemic. In particular, neoantigens are emerging as the preferred targets for CVs, with gene-based and viral vector-based platforms in development. Moreover, lipid nanoparticles proved to be immunogenic and efficient delivery vehicles. Past clinical trials investigating CVs focused especially on the metastatic disease, where the TME is more likely compromised by inhibitory mechanisms. In this sense, favouring the use of CVs as monotherapy in premalignant or in the adjuvant setting and establishing combination treatments (i.e. CV plus ICI) in late-stage disease are promising strategies. This review provides a full overview of the past and current breast cancer vaccine landscape.
Collapse
Affiliation(s)
- Chiara Corti
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - Pier P M B Giachetti
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - Alexander M M Eggermont
- Princess Máxima Center, Utrecht, the Netherlands; Department of Cancer Medicine, Institut Gustave Roussy, Villejuif, France
| | - Suzette Delaloge
- Department of Cancer Medicine, Institut Gustave Roussy, Villejuif, France
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy.
| |
Collapse
|
45
|
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
|
46
|
Díaz-Rodríguez E, Gandullo-Sánchez L, Ocaña A, Pandiella A. Novel ADCs and Strategies to Overcome Resistance to Anti-HER2 ADCs. Cancers (Basel) 2021; 14:154. [PMID: 35008318 PMCID: PMC8750930 DOI: 10.3390/cancers14010154] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/27/2021] [Accepted: 12/27/2021] [Indexed: 12/23/2022] Open
Abstract
During recent years, a number of new compounds against HER2 have reached clinics, improving the prognosis and quality of life of HER2-positive breast cancer patients. Nonetheless, resistance to standard-of-care drugs has motivated the development of novel agents, such as new antibody-drug conjugates (ADCs). The latter are a group of drugs that benefit from the potency of cytotoxic agents whose action is specifically guided to the tumor by the target-specific antibody. Two anti-HER2 ADCs have reached the clinic: trastuzumab-emtansine and, more recently, trastuzumab-deruxtecan. In addition, several other HER2-targeted ADCs are in preclinical or clinical development, some of them with promising signs of activity. In the present review, the structure, mechanism of action, and potential resistance to all these ADCs will be described. Specific attention will be given to discussing novel strategies to circumvent resistance to ADCs.
Collapse
Affiliation(s)
- Elena Díaz-Rodríguez
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-IBSAL and CIBERONC, 37007 Salamanca, Spain; (E.D.-R.); (L.G.-S.)
- Departamento de Bioquímica y Biología Molecular, University of Salamanca, 37007 Salamanca, Spain
| | - Lucía Gandullo-Sánchez
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-IBSAL and CIBERONC, 37007 Salamanca, Spain; (E.D.-R.); (L.G.-S.)
| | - Alberto Ocaña
- Hospital Clínico San Carlos, Centro de Investigación Biomédica en Red de Oncología (CIBERONC), 28040 Madrid, Spain;
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-IBSAL and CIBERONC, 37007 Salamanca, Spain; (E.D.-R.); (L.G.-S.)
| |
Collapse
|
47
|
Gambardella V, Alfaro-Cervelló C, Cejalvo JM, Tapia M, Cervantes A. In the literature: August 2021. ESMO Open 2021; 6:100247. [PMID: 34411970 PMCID: PMC8377552 DOI: 10.1016/j.esmoop.2021.100247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 07/27/2021] [Indexed: 11/20/2022] Open
Affiliation(s)
- V Gambardella
- Department of Medical Oncology, Hospital Clínico Universitario, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - C Alfaro-Cervelló
- Department of Pathology, Hospital Clínico Universitario, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - J M Cejalvo
- Department of Medical Oncology, Hospital Clínico Universitario, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - M Tapia
- Department of Medical Oncology, Hospital Clínico Universitario, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - A Cervantes
- Department of Medical Oncology, Hospital Clínico Universitario, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
48
|
Giannoudis A, Sartori A, Eastoe L, Zakaria R, Charlton C, Hickson N, Platt-Higgins A, Rudland PS, Irwin D, Jenkinson MD, Palmieri C. Genomic profiling using the UltraSEEK panel identifies discordancy between paired primary and breast cancer brain metastases and an association with brain metastasis-free survival. Breast Cancer Res Treat 2021; 190:241-253. [PMID: 34499316 PMCID: PMC8558178 DOI: 10.1007/s10549-021-06364-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 08/08/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE Brain metastases (BM) are an increasing clinical problem. This study aimed to assess paired primary breast cancers (BC) and BM for aberrations within TP53, PIK3CA, ESR1, ERBB2 and AKT utilising the MassARRAY® UltraSEEK® technology (Agena Bioscience, San Diego, USA). METHODS DNA isolated from 32 paired primary BCs and BMs was screened using the custom UltraSEEK® Breast Cancer Panel. Data acquisition and analysis was performed by the Agena Bioscience Typer software v4.0.26.74. RESULTS Mutations were identified in 91% primary BCs and 88% BM cases. TP53, AKT1, ESR1, PIK3CA and ERBB2 genes were mutated in 68.8%, 37.5%, 31.3%, 28.1% and 3.1% respectively of primary BCs and in 59.4%, 37.5%, 28.1%, 28.1% and 3.1% respectively of BMs. Differences in the mutations within the 5 genes between BC and paired BM were identified in 62.5% of paired cases. In primary BCs, ER-positive/HER2-negative cases harboured the most mutations (70%), followed by ER-positive/HER2-positive (15%) and triple-negatives (13.4%), whereas in BMs, the highest number of mutations was observed in triple-negative (52.5%), followed by ER-positive/HER2-negative (35.6%) and ER-negative/HER2-positive (12%). There was a significant association between the number of mutations in the primary BC and breast-to-brain metastasis-free survival (p = 0.0001) but not with overall survival (p = 0.056). CONCLUSION These data demonstrate the discordancy between primary BC and BM, as well as the presence of clinically important, actionable mutations in BCBM. The UltraSEEK® Breast Cancer Panel provides a tool for BCBM that can be utilised to direct more tailored treatment decisions and for clinical studies investigating targeted agents.
Collapse
Affiliation(s)
- Athina Giannoudis
- Institute of Systems, Molecular and Integrative Biology, Molecular and Clinical Cancer Medicine, University of Liverpool, Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK
| | | | | | - Rasheed Zakaria
- Institute of Systems, Molecular and Integrative Biology, Molecular and Clinical Cancer Medicine, University of Liverpool, Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK
- Department of Neurosurgery, The Walton Centre NHS Foundation Trust, Liverpool, UK
| | | | - Nicholas Hickson
- Manchester University Hospital NHS Foundation Trust, Manchester, UK
| | - Angela Platt-Higgins
- Institute of Systems, Molecular and Integrative Biology, Molecular and Clinical Cancer Medicine, University of Liverpool, Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK
| | - Philip S Rudland
- Institute of Systems, Molecular and Integrative Biology, Molecular and Clinical Cancer Medicine, University of Liverpool, Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK
| | | | - Michael D Jenkinson
- Department of Neurosurgery, The Walton Centre NHS Foundation Trust, Liverpool, UK
- Institute of Systems, Molecular and Integrative Biology, Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
| | - Carlo Palmieri
- Institute of Systems, Molecular and Integrative Biology, Molecular and Clinical Cancer Medicine, University of Liverpool, Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK.
- The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, UK.
| |
Collapse
|
49
|
Hanker AB, Brown BP, Meiler J, Marín A, Jayanthan HS, Ye D, Lin CC, Akamatsu H, Lee KM, Chatterjee S, Sudhan DR, Servetto A, Brewer MR, Koch JP, Sheehan JH, He J, Lalani AS, Arteaga CL. Co-occurring gain-of-function mutations in HER2 and HER3 modulate HER2/HER3 activation, oncogenesis, and HER2 inhibitor sensitivity. Cancer Cell 2021; 39:1099-1114.e8. [PMID: 34171264 PMCID: PMC8355076 DOI: 10.1016/j.ccell.2021.06.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 02/28/2021] [Accepted: 06/02/2021] [Indexed: 12/24/2022]
Abstract
Activating mutations in HER2 (ERBB2) drive the growth of a subset of breast and other cancers and tend to co-occur with HER3 (ERBB3) missense mutations. The HER2 tyrosine kinase inhibitor neratinib has shown clinical activity against HER2-mutant tumors. To characterize the role of HER3 mutations in HER2-mutant tumors, we integrate computational structural modeling with biochemical and cell biological analyses. Computational modeling predicts that the frequent HER3E928G kinase domain mutation enhances the affinity of HER2/HER3 and reduces binding of HER2 to its inhibitor neratinib. Co-expression of mutant HER2/HER3 enhances HER2/HER3 co-immunoprecipitation and ligand-independent activation of HER2/HER3 and PI3K/AKT, resulting in enhanced growth, invasiveness, and resistance to HER2-targeted therapies, which can be reversed by combined treatment with PI3Kα inhibitors. Our results provide a mechanistic rationale for the evolutionary selection of co-occurring HER2/HER3 mutations and the recent clinical observations that HER3 mutations are associated with a poor response to neratinib in HER2-mutant cancers.
Collapse
MESH Headings
- Aminopyridines/administration & dosage
- Animals
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Cell Line, Tumor
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Female
- Gain of Function Mutation
- Humans
- Mice, Nude
- Molecular Docking Simulation
- Molecular Dynamics Simulation
- Morpholines/administration & dosage
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphoinositide-3 Kinase Inhibitors/administration & dosage
- Protein Multimerization
- Quinolines/administration & dosage
- Quinolines/chemistry
- Quinolines/metabolism
- Quinolines/pharmacology
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/chemistry
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-3/chemistry
- Receptor, ErbB-3/genetics
- Receptor, ErbB-3/metabolism
- Trastuzumab/pharmacology
- Xenograft Model Antitumor Assays
- Mice
Collapse
Affiliation(s)
- Ariella B Hanker
- UTSW Simmons Comprehensive Cancer Center, Dallas, 5323 Harry Hines Boulevard, TX 75390, USA; Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Benjamin P Brown
- Chemical and Physical Biology Program, Center for Structural Biology, and Medical Scientist Training Program, Vanderbilt University, Nashville, TN 37240, USA
| | - Jens Meiler
- Department of Chemistry and Center for Structural Biology, Vanderbilt University, Nashville, TN 37240, USA; Institute for Drug Discovery, Leipzig University Medical School, Leipzig, SAC 04103, Germany
| | - Arnaldo Marín
- UTSW Simmons Comprehensive Cancer Center, Dallas, 5323 Harry Hines Boulevard, TX 75390, USA; Doctoral Program in Medical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Harikrishna S Jayanthan
- Department of Chemistry and Center for Structural Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - Dan Ye
- UTSW Simmons Comprehensive Cancer Center, Dallas, 5323 Harry Hines Boulevard, TX 75390, USA
| | - Chang-Ching Lin
- UTSW Simmons Comprehensive Cancer Center, Dallas, 5323 Harry Hines Boulevard, TX 75390, USA
| | - Hiroaki Akamatsu
- UTSW Simmons Comprehensive Cancer Center, Dallas, 5323 Harry Hines Boulevard, TX 75390, USA
| | - Kyung-Min Lee
- UTSW Simmons Comprehensive Cancer Center, Dallas, 5323 Harry Hines Boulevard, TX 75390, USA; Department of Life Sciences, College of Natural Science, Hanyang University, Seoul 04736, Republic of Korea
| | - Sumanta Chatterjee
- UTSW Simmons Comprehensive Cancer Center, Dallas, 5323 Harry Hines Boulevard, TX 75390, USA
| | - Dhivya R Sudhan
- UTSW Simmons Comprehensive Cancer Center, Dallas, 5323 Harry Hines Boulevard, TX 75390, USA
| | - Alberto Servetto
- UTSW Simmons Comprehensive Cancer Center, Dallas, 5323 Harry Hines Boulevard, TX 75390, USA
| | - Monica Red Brewer
- Department of Chemistry and Center for Structural Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - James P Koch
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jonathan H Sheehan
- Division of Infectious Diseases, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jie He
- Foundation Medicine, Cambridge, MA 02141, USA
| | | | - Carlos L Arteaga
- UTSW Simmons Comprehensive Cancer Center, Dallas, 5323 Harry Hines Boulevard, TX 75390, USA; Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
50
|
Clinicopathologic features and treatment advances in cancers with HER2 alterations. Biochim Biophys Acta Rev Cancer 2021; 1876:188605. [PMID: 34358635 DOI: 10.1016/j.bbcan.2021.188605] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 12/14/2022]
Abstract
HER2 is one of the most important proteins of the epidermal growth factor receptor (EGFR) family, whose alterations include amplification, overexpression and gene mutation. Growing attention has been given to HER2 as a biomarker for prognosis, an indicator for treatment response and a target for new drugs. Tumors with HER2 alterations have been well studied in multiple locations as distinct entities for treatment, especially breast cancer, gastric cancer, lung cancer and colorectal cancer. These four cancers are the leading causes of cancer incidence and cancer-related death worldwide. The present study details the landscape of HER2 amplification/overexpression and mutations and gives an up-to-date analysis of current clinical trials in the four cancers mentioned above. Different HER2-altered cancers not only respond differently to HER2-targeting therapies but also display diverse survival outcomes. Even in the same type of cancer, HER2 amplification/overexpression differs from HER2 mutation in terms of clinicopathologic features and treatment strategies. As an emerging strategy in cancer treatment, immune checkpoint inhibitors demonstrate distinct outcomes in HER2-altered breast cancer, gastric cancer and lung cancer.
Collapse
|