1
|
Shen K, Yuan S, Su N, Tang F, Rehim S, Wang H, Guo H, Zhang Y, Wu Y, Wang H. Monotherapy and combination therapy using antibody‑drug conjugates for platinum‑resistant ovarian cancer. Oncol Rep 2025; 53:68. [PMID: 40242965 PMCID: PMC12046379 DOI: 10.3892/or.2025.8901] [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: 12/04/2024] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Platinum‑resistant ovarian cancer (PROC) is a significant clinical challenge due to the limited number of treatment options and poor outcomes. Moreover, cytotoxic drugs have an unsatisfactory therapeutic efficacy, high toxicity and side effects. An antibody‑drug conjugate (ADC) is a novel cancer therapeutic strategy that combines an antibody, a linker and a payload. ADCs precisely target the tumor cells by binding to the antigen on the surface of tumor cells, thus accurately delivering the cytotoxic drugs and minimizing systemic toxicity. The approval of mirvetuximab soravtansine by the US Food and Drug Administration for treating folate receptor alpha‑positive, platinum‑resistant epithelial ovarian cancer has promoted studies on the use of ADCs in ovarian cancer. A phase III clinical trial showed that mirvetuximab soravtansine achieved an objective remission rate of 42.3% in platinum‑resistant, FRα‑positive ovarian cancer, compared with 15.9% using chemotherapy, demonstrating its immense potential for ADC development. The present review summarizes the research progress on the use of ADCs in PROC as a monotherapy and combination therapy and considers the future development direction of ADCs in PROC.
Collapse
Affiliation(s)
- Ke Shen
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Shuang Yuan
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Ning Su
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
- Department of Gynecologic Oncology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Furong Tang
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Shamsnur Rehim
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Han Wang
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Huihui Guo
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Yu Zhang
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Yufeng Wu
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Hongjing Wang
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
2
|
Rieke DT, Bitzer M, Bleckmann A, Desuki A, Ernst T, Esposito I, Gerger A, Glimm H, Horak P, Hübschmann D, Illert AL, Kunzmann V, Loges S, Pretzell I, Schmitz K, Seeber A, Söhlke B, Wicki A, Wolf J, Maschmeyer G. Precision oncology - Guideline of the Austrian, German and Swiss Societies for hematology and medical oncology. Eur J Cancer 2025; 220:115331. [PMID: 40058270 DOI: 10.1016/j.ejca.2025.115331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 04/26/2025]
Abstract
Precision oncology is a multi-step process including patient selection, tumor profiling, molecular tumor board discussion and personalized cancer management. So far, it remains largely unstandardized. The implementation of precision oncology can be beneficial for patients but implementation differs widely between tumor types and local practices. A working group was established by the Austrian, German and Swiss societies for Hematology and Medical Oncology to establish an expert consensus on evidence-based standards and implementation of precision oncology. Herein, we present a summary of this guideline. The full documents are available at www.onkopedia-guidelines.info.
Collapse
Affiliation(s)
- Damian T Rieke
- Comprehensive Cancer Center and Department for Hematology, Oncology and Cancer Immunology Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Germany.
| | - Michael Bitzer
- Department of Internal Medicine I, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Annalen Bleckmann
- Department of Medicine A, Hematology, Oncology and Pneumology, West German Cancer Center, University Hospital Muenster, Muenster, Germany
| | - Alexander Desuki
- University Cancer Center Mainz, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Thomas Ernst
- Department of Hematology/Oncology, Clinic of Internal Medicine II, Jena University Hospital, Jena, Germany
| | - Irene Esposito
- Institute of Pathology, Heinrich Heine University and University Hospital of Duesseldorf, Dusseldorf, Germany
| | - Armin Gerger
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Hanno Glimm
- Department for Translational Medical Oncology, National Center for Tumor Diseases Dresden (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Peter Horak
- Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniel Hübschmann
- Computational Oncology, Molecular Precision Oncology Program, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anna Lena Illert
- Center for Personalized Medicine (ZPM), Technical University of Munich (TUM), School of Medicine and Health, Klinikum rechts der Isar, TUM University Hospital, Munich, Germany
| | - Volker Kunzmann
- Department of Internal Medicine II, Medical Oncology, Comprehensive Cancer Center Mainfranken Würzburg, University Hospital Würzburg, Germany
| | - Sonja Loges
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Ina Pretzell
- West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Katja Schmitz
- Tyrolpath Obrist Brunhuber GmbH and Krankenhaus St. Vinzenz, Zams, Austria
| | - Andreas Seeber
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Bärbel Söhlke
- Zielgenau e.V. Patient network for personalized lung cancer therapy, Kerpener Str. 6250937 Cologne, Germany
| | - Andreas Wicki
- Department of Medical Oncology and Hematology, University Hospital of Zurich, Switzerland; University of Zurich, Switzerland
| | - Jürgen Wolf
- Department of Internal Medicine, Center for Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | - Georg Maschmeyer
- Deutsche Gesellschaft für Hämatologie und medizinische Onkologie e.V., Bauhofstr. 12, Berlin 10117, Germany
| |
Collapse
|
3
|
Chang Y, Zhao D, Wang Z, Zhu K, Guo A, Cao J, Wu C, Ding S. Expression of HER2 in urothelial carcinoma and its significance. Curr Urol 2025; 19:201-207. [PMID: 40376477 PMCID: PMC12076403 DOI: 10.1097/cu9.0000000000000249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 12/06/2023] [Indexed: 05/18/2025] Open
Abstract
Objectives We explored the expression levels and clinical significance of human epidermal growth factor receptor 2 (HER2) in urothelial carcinoma (UC) tissues. Materials and methods Patient data were reviewed, and 111 paraffin specimens of UC obtained from the Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, from 2020 to 2021 were collected. Immunohistochemistry was used to detect HER2 protein expression in all UC tumor tissues. The χ 2 and Fisher exact tests were used to analyze the relationship between HER2 protein expression and clinicopathological data (sex, age, histopathological diagnosis, invasiveness, histopathological grade, maximum tumor diameter, muscle invasion, regional lymph node metastasis, and clinical stage). Results In this study, 92 cases (82.88%) showed HER2 protein expression, and there was a statistically significant difference in the distribution of HER2 positivity (immunohistochemistry 2+ and 3+) according to the pathological grades of UC (p = 0.021). Human epidermal growth factor receptor 2 positivity was not associated with sex, age, histopathological diagnosis, invasiveness, maximum tumor diameter, muscle invasion, regional lymph node metastasis, or clinical stage (all p < 0.05). Conclusions Human epidermal growth factor receptor 2 protein is highly expressed in UC, and its expression may be closely related to the high pathological grade of UC.
Collapse
Affiliation(s)
- Yao Chang
- Department of Urology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Delong Zhao
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zicheng Wang
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Urology, Southern theater General Hospital, Guangzhou, China
| | - Kejia Zhu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Andong Guo
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jishuang Cao
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chenrui Wu
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Sentai Ding
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
4
|
Boonsriroj H, Tangtrongsup S, Arunorat J, Mamom T, Chantawong P. Unveiling HER2 immunoexpression in canine hepatoid gland neoplasms: clinicopathological and morphological associations. Int J Vet Sci Med 2025; 13:1-12. [PMID: 40290666 PMCID: PMC12024500 DOI: 10.1080/23144599.2025.2495522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/18/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025] Open
Abstract
Canine hepatoid gland neoplasms (HGNs) are significant clinical concerns due to their high prevalence and diverse biological behaviour. Human epidermal growth factor receptor 2 (HER2), a tyrosine kinase receptor implicated in various aspects of tumorigenesis, has been extensively studied in human and animal neoplasms but remains unexplored in HGNs. This study aimed to assess HER2 immunoexpression in canine HGNs and its association with clinicopathological and morphological features. A total of 61 formalin-fixed paraffin-embedded samples, including normal hepatoid glands (n = 10), hepatoid gland adenomas (HGAs, n = 20), hepatoid gland epitheliomas (HGEs, n = 16), and hepatoid gland carcinomas (HGCs, n = 15), were analysed using immunohistochemistry. HER2 expression was scored based on percentage positivity and staining intensity. HER2-positive expression was detected in 50% of HGEs (score 2 + ) and 73.3% of HGCs, with 36.4% of cases scoring 3 + . In contrast, all HGAs and normal hepatoid tissues were HER2-immunonegative. Statistical analysis revealed significant differences in HER2 expression among normal and neoplastic hepatoid glands (p < 0.001). Only in HGCs, HER2 expression was significantly associated with tissue invasion (p = 0.007), mitotic count (p = 0.033), and nuclear pleomorphism (p = 0.007). These findings suggest that HER2 may play a role in the progression of malignant HGNs, particularly HGCs. This preliminary study highlights the potential of HER2 as a diagnostic marker and emphasizes the need for further investigation into its prognostic value and role in HER2-targeted therapy for canine HGCs.
Collapse
Affiliation(s)
- Hassadin Boonsriroj
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Mahanakorn University of Technology, Bangkok, Thailand
- Animal Diagnostic Laboratory Center, Faculty of Veterinary Medicine, Mahanakorn University of Technology, Bangkok, Thailand
| | - Sahatchai Tangtrongsup
- Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
- Research Center of Producing and Development of Products and Innovations for Animal Health and Production, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Jirapat Arunorat
- Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
- Research Center for Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Thanongsak Mamom
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Mahanakorn University of Technology, Bangkok, Thailand
- Animal Diagnostic Laboratory Center, Faculty of Veterinary Medicine, Mahanakorn University of Technology, Bangkok, Thailand
| | - Pinkarn Chantawong
- Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
- Research Center of Producing and Development of Products and Innovations for Animal Health and Production, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
- Research Center for Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
5
|
Yu R, Ji X, Zhang P, Zhang H, Qu H, Dong W. The potential of chimeric antigen receptor -T cell therapy for endocrine cancer. World J Surg Oncol 2025; 23:153. [PMID: 40264184 PMCID: PMC12012980 DOI: 10.1186/s12957-025-03745-x] [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: 12/20/2024] [Accepted: 03/07/2025] [Indexed: 04/24/2025] Open
Abstract
Endocrine cancer, a relatively rare and heterogeneous tumor with diverse clinical features. The facile synthesis of hormones further complicates endocrine cancer treatment. Thus, the development of safe and effective systemic treatment approaches, such as chimeric antigen receptor (CAR) T cell therapy, is imperative to enhance the prognosis of patients with endocrine cancer. Although this therapy has achieved good results in the treatment of hematological malignancies, it encounters diverse complications and challenges in the context of endocrine cancer. This review delineates the generation of CAR-T cells, examines the potential of CAR-T cell therapy for endocrine cancer, enumerates pivotal antigens linked to endocrine cancer, encapsulates the challenges confronted with CAR-T cell therapy for endocrine cancer, and expounds upon strategies to overcome these limitations. The primary objective is to provide insightful perspectives that can contribute to the advancement of CAR-T cell therapy in the field of endocrine cancer.
Collapse
Affiliation(s)
- Ruonan Yu
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Xiaoyu Ji
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Ping Zhang
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Hao Zhang
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Huiling Qu
- Department of Neurology, The General Hospital of Northern Theater Command, 83 Wen Hua Road, Shenyang, Liaoning, 110840, China.
| | - Wenwu Dong
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China.
| |
Collapse
|
6
|
Tao W, Zhang J, Meng X, Han X, Wang Q, Lin Y, Cheng L, Liu M, Da D, Zhang H, Fan J, Zhang L, Liu S, Li S, Gao F, Ren Y. Development and clinical evaluation of [ 68Ga]Ga-NODAGA-ADAPT6 as a novel HER2-targeted PET radiotracer for breast cancer imaging and treatment monitoring. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07286-z. [PMID: 40257612 DOI: 10.1007/s00259-025-07286-z] [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: 02/08/2025] [Accepted: 04/10/2025] [Indexed: 04/22/2025]
Abstract
PURPOSE Accurate assessment of human epidermal growth factor receptor type 2 (HER2) expression is crucial for diagnosis, treatment planning, and monitoring of breast cancer patients. A 68Ga-labeled tracer based on the albumin-binding domain-derived affinity protein 6 (ADAPT6) was developed to evaluate HER2 expression in breast cancer. METHODS The gene encoding ADAPT6 was modified with N-terminal (GHEHEHEDANS) and C-terminal (GSSC) extensions to enhance its functionality. The precursor was synthesized, purified, and characterized, followed by radiolabeling with 68Ga to produce [68Ga]Ga-NODAGA-ADAPT6. In vivo metabolism and biodistribution studies were performed in HCC1954 (HER2-positive) and MDA-MB-468 (HER2-negative) tumor-bearing mice. Additionally, with ethical approval and informed consent, 22 breast cancer patients underwent [68Ga]Ga-NODAGA-ADAPT6 PET imaging to assess HER2 expression in primary and metastatic lesions. RESULTS The tracer was prepared with a radiochemical purity exceeding 99% and demonstrated high stability in vivo. Micro-PET/CT imaging revealed significant accumulation of the radiotracer in HCC1954 tumors, which was markedly reduced after HER2 blockade with trastuzumab. In contrast, MDA-MB-468 tumors showed minimal uptake. In the clinical study, [68Ga]Ga-NODAGA-ADAPT6 PET images displayed varying levels of radiotracer uptake in primary and metastatic lesions, which correlated well with the HER2 expression status determined by pathological analysis. CONCLUSION [68Ga]Ga-NODAGA-ADAPT6 exhibited excellent pharmacokinetic properties and high specificity for HER2-expressing lesions in PET imaging. These findings highlight its potential as a promising tool for distinguishing different levels of HER2 expression in breast cancer, aiding in personalized treatment strategies.
Collapse
Affiliation(s)
- Weijing Tao
- Department of Nuclear Medicine, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huai'an, 223300, Jiangsu, China.
| | - Jinglin Zhang
- Key Laboratory for Experimental Teratology of the Ministry of Education and Research Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Xin Meng
- Department of Breast and Thyroid Surgery, The Huai'an Clinical College of Xuzhou Medical University, Huai'an, 223300, Jiangsu, China
| | - Xuedong Han
- Department of Breast and Thyroid Surgery, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huai'an, Jiangsu, 223300, China
| | - Qiuhu Wang
- Department of Nuclear Medicine, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huai'an, 223300, Jiangsu, China
| | - Yixiang Lin
- Key Laboratory for Experimental Teratology of the Ministry of Education and Research Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Luyi Cheng
- Department of Nuclear Medicine, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huai'an, 223300, Jiangsu, China
| | - Minmin Liu
- Department of Breast and Thyroid Surgery, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huai'an, Jiangsu, 223300, China
| | - Dongzhu Da
- Department of Breast and Thyroid Surgery, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huai'an, Jiangsu, 223300, China
| | - Huai Zhang
- Department of Nuclear Medicine, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huai'an, 223300, Jiangsu, China
| | - Junfu Fan
- Department of Nuclear Medicine, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huai'an, 223300, Jiangsu, China
| | - Lianmei Zhang
- Department of Pathology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huai'an, 223300, Jiangsu, China
| | - Shuangyue Liu
- Department of Breast and Thyroid Surgery, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huai'an, Jiangsu, 223300, China
| | - Shuo Li
- Department of Breast and Thyroid Surgery, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huai'an, Jiangsu, 223300, China
| | - Feng Gao
- Key Laboratory for Experimental Teratology of the Ministry of Education and Research Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, Shandong, China.
| | - Yi Ren
- Department of Breast and Thyroid Surgery, The Huai'an Clinical College of Xuzhou Medical University, Huai'an, 223300, Jiangsu, China.
- Department of Breast and Thyroid Surgery, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huai'an, Jiangsu, 223300, China.
| |
Collapse
|
7
|
Hu J, Yan L, Liu J, Chen M, Liu P, Deng D, Zhang C, He Y, Fan B, Li H, Gong G, Xiao J, Wang R, Guan X, Tong S, Li Y, Li N, Tang Z, Zhang T, Li H, Huang B, Gao N, He W, Cai Z, Liu Y, Liu Z, Gan Y, Cui Y, Dai Y, Cai Y, Nie Z, Ou Z, Chen J, Zu X. Efficacy and biomarker analysis of neoadjuvant disitamab vedotin (RC48‐ADC) combined immunotherapy in patients with muscle‐invasive bladder cancer: A multi‐center real‐world study. IMETA 2025. [DOI: 10.1002/imt2.70033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 04/01/2025] [Indexed: 05/04/2025]
Affiliation(s)
- Jiao Hu
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Luzhe Yan
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Jinhui Liu
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Minfeng Chen
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Peihua Liu
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Dingshan Deng
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Chaobin Zhang
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Yunbo He
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Benyi Fan
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Huihuang Li
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Guanghui Gong
- Department of Pathology, Xiangya Hospital Central South University Changsha China
| | - Jiatong Xiao
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Ruizhe Wang
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Xiao Guan
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Shiyu Tong
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Yangle Li
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Nannan Li
- Department of Urology The First Hospital of Changsha Changsha China
| | - Zhiwang Tang
- Department of Urology The First Hospital of Changsha Changsha China
| | - Teng Zhang
- Department of Urology First People's Hospital of Guiyang Guiyang China
| | - Hao Li
- Department of Urology First People's Hospital of Guiyang Guiyang China
| | - Bin Huang
- Department of Urology Xiangya Boai Rehabilitation Hospital Changsha China
| | - Ning Gao
- Department of Urology Xiangya Boai Rehabilitation Hospital Changsha China
| | - Wei He
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Zhiyong Cai
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Yifan Liu
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Zefu Liu
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Yu Gan
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Yu Cui
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Yuanqing Dai
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Yi Cai
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Zhenyu Nie
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Zhenyu Ou
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Jinbo Chen
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
| | - Xiongbing Zu
- Department of Urology, Xiangya Hospital Central South University Changsha China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University Changsha China
- Furong Laboratory Changsha China
- Department of Urology, Hunan Provincial People's Hospital the First Affiliated Hospital of Hunan Normal University Changsha China
| |
Collapse
|
8
|
Westerberg C, Mestre Borras A, Ståhl S, Löfblom J. Affibody-based HER2 prodrug shows conditional cytotoxic effect on HER2-positive cancer cells. Biochem Biophys Res Commun 2025; 758:151660. [PMID: 40117970 DOI: 10.1016/j.bbrc.2025.151660] [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: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025]
Abstract
Therapeutic affinity proteins offer a targeted mode of action due to their typically high affinity and specificity for disease-associated molecules. In cancer therapy, such target molecules are often overexpressed receptors on tumor cells. However, their presence in healthy tissues can lead to on-target, off-tumor toxicity, necessitating strategies to enhance tumor selectivity. Here, we present an affibody-based prodrug concept that exploits tumor-associated proteases for selective activation. As proof of concept, we designed, produced, and characterized HER2-specific prodrug candidates, each incorporating a distinct protease substrate for selective activation by tumor-associated proteases. Their activation by corresponding proteases and subsequent HER2 binding were assessed. The most promising prodrug candidate was conjugated to the cytotoxic agent DM1 and evaluated for cytotoxicity in HER2-positive cancer cells. The results demonstrated potent, HER2-dependent cell killing, with markedly reduced cytotoxicity in the absence of prodrug activation. These findings support the feasibility of affibody-based prodrugs as a strategy to enhance tumor selectivity and minimize off-tumor toxicity in targeted cancer therapy.
Collapse
Affiliation(s)
- Cornelia Westerberg
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Anna Mestre Borras
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Stefan Ståhl
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - John Löfblom
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden.
| |
Collapse
|
9
|
Jin G, Wang J. Both complete response and long-term survival after combination therapy with toripalimab in a patient with meta-oligometastases cervical cancer: a case report. Front Immunol 2025; 16:1542795. [PMID: 40260238 PMCID: PMC12009880 DOI: 10.3389/fimmu.2025.1542795] [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: 12/10/2024] [Accepted: 03/21/2025] [Indexed: 04/23/2025] Open
Abstract
Background The therapeutic landscape for recurrent or metastatic cervical cancer remains limited, with few options available. According to National Comprehensive Cancer Network (NCCN) guidelines, pembrolizumab combined with chemotherapy, with or without bevacizumab, is recommended for affected patients. Despite these guidelines, recurrence rates remain elevated, and survival outcomes following standard interventions are unsatisfactory. Furthermore, real-world management of recurrent or metastatic cervical cancer presents inherent complexities, often requiring an integrative, multidimensional treatment approach to enhance long-term survival. The pressing need to refine and adopt multimodal therapeutic strategies is evident in addressing the persistent challenges associated with disease recurrence and progression. Case description The case involved a 40-year-old female diagnosed with advanced cervical cancer who underwent radical hysterectomy. Postoperative pathology identified high-risk features, including lymph node involvement, necessitating adjuvant chemoradiotherapy. However, disease progression occurred during treatment, manifesting as metastases in the left supraclavicular and axillary lymph nodes. Subsequent local radiotherapy and systemic therapy led to a favorable response. By November 2024, overall survival (OS) had surpassed 72 months, with toripalimab administered for 65 months, during which no immunotherapy-related adverse events occurred. Conclusion This case offers clinical insight into the efficacy and safety of integrating chemotherapy, immunotherapy, and radiotherapy in recurrent or metastatic cervical cancer. The multimodal approach contributes to prolonged survival in this patient. Further clinical trials are essential to substantiate the therapeutic benefits of this regimen in broader patient cohorts.
Collapse
Affiliation(s)
- Ge Jin
- 1 Department of Gynecologic Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jun Wang
- Department of Radiation Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
10
|
Huang H, Zhou Y, Shang C, Zhang Y, Shen Y. A novel anti-HER2/EGFR bispecific antibody-drug conjugate demonstrates promising antitumor efficacy and overcomes resistance to HER2- or EGFR-targeted ADCs. Invest New Drugs 2025; 43:262-275. [PMID: 39982632 DOI: 10.1007/s10637-025-01507-w] [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: 11/23/2024] [Accepted: 01/17/2025] [Indexed: 02/22/2025]
Abstract
HER2 and EGFR are frequently co-expressed in various tumors. While antibody-drug conjugates (ADCs) targeting HER2, such as T-DM1 and T-Dxd, have shown remarkable antitumor effects in clinical responses, their effectiveness is constrained by drug resistance. EGFR amplification or high expression is one of the factors that lead to resistance against HER2-targeted ADCs. Likewise, the amplification of HER2 may lead to the development of resistance to EGFR-targeted therapies. To overcome these challenges, we, therefore, developed a bispecific antibody (B2C4) that targets HER2 and EGFR. B2C4 exhibited strong binding affinity and internalization activity in tumor cells with high expression of HER2 and EGFR, as well as in those with high expression of either target. B2C4 was then conjugated with vc-MMAE to create a bispecific ADC (B2C4-MMAE) with an average DAR of 4.05. By effectively engaging both arms of the bispecific ADC, B2C4-MMAE demonstrated significant antitumor activity in tumor cells and animal models that were unresponsive HER2- or EGFR-targeted ADCs. B2C4-MMAE could serve as an alternative therapeutic option for tumors that are resistant to single-target treatments. Additionally, B2C4-MMAE exhibited potential in treating tumors resistant to T-Dxd, underscoring its promise as a treatment for challenging cases.
Collapse
Affiliation(s)
- Huoying Huang
- Anhui University of Chinese Medicine, Hefei, People's Republic of China
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, People's Republic of China
- Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong, People's Republic of China
| | - Yuxin Zhou
- Anhui University of Chinese Medicine, Hefei, People's Republic of China
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, People's Republic of China
- Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong, People's Republic of China
| | - Chengzhang Shang
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, People's Republic of China
| | - Yifu Zhang
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, People's Republic of China
| | - Yuelei Shen
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, People's Republic of China.
- Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong, People's Republic of China.
| |
Collapse
|
11
|
Salinaro J, Singh K, Sands N, Gill V, Perati S, James N, Sharma S, Nasir A, DiSilvestro P, Miller K, Oliver M, Mathews C. Distribution and concordance of HER2 scores in endometrial and ovarian cancer. Gynecol Oncol 2025; 195:115-121. [PMID: 40088505 DOI: 10.1016/j.ygyno.2025.03.012] [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: 12/08/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025]
Abstract
OBJECTIVES Although multiple HER2 scoring criteria exist, the optimal strategy to identify patients with gynecologic malignancies who may benefit from HER2-directed therapies remains unknown. The objectives of this study were to assess the distribution and concordance of HER2 scores in endometrial and ovarian cancer. METHODS One hundred five tumor specimens from 94 patients with endometrial or epithelial ovarian cancer (EOC) who underwent Caris tumor profiling from 11/2022 to 01/2025 were identified from a retrospective database. Each sample was assigned a HER2 immunohistochemistry (IHC) score of 0, 1+, 2+, or 3+ using breast, endometrial, and gastric criteria. ERBB2 amplification and HER2 IHC scores were abstracted from Caris reports. Patient characteristics and HER2 score distributions were analyzed using descriptive statistics and Fisher's exact test. Matrix correlation coefficients were used to assess HER2 score concordance. RESULTS A total of 105 samples underwent internal triplicate HER2 scoring - 63 EOC and 42 endometrial. A higher percentage of patients with endometrial cancer were HER2-high compared to those with EOC (45.2-50 % vs 20.6 %, p < 0.05). Internal triplicate HER2 score concordance was strong (r ≥ 0.96, p < 0.001) but weaker when compared to Caris scores (r = 0.66). Of the 23 discordant HER2 results, 13 would have changed therapy eligibility (56.5 %). Only 12 patients (12.7 %) had intermediate or high ERBB2 amplification. CONCLUSIONS A clinically significant percentage of patients had HER2-high tumors regardless of tumor type. HER2 score concordance was strong within each sample but weaker when compared to Caris scores. Incorporating multi-site testing and/or validation of external IHC into any gynecologic HER2 scoring algorithm should be considered.
Collapse
Affiliation(s)
- Julia Salinaro
- Program in Women's Oncology, Women and Infants Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA.
| | - Kamaljeet Singh
- Department of Pathology, Women and Infants Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Natalie Sands
- Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Victoria Gill
- Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Shriya Perati
- Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Nicole James
- Program in Women's Oncology, Women and Infants Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Shreenidhi Sharma
- Hassenfeld Child Health and Innovation Institute, School of Public Health, Brown University, Providence, RI, USA
| | | | - Paul DiSilvestro
- Program in Women's Oncology, Women and Infants Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Katherine Miller
- Program in Women's Oncology, Women and Infants Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Matthew Oliver
- Program in Women's Oncology, Women and Infants Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Cara Mathews
- Program in Women's Oncology, Women and Infants Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| |
Collapse
|
12
|
Andriollo P, di Mascio D, Jackson PJM, Hasan MM, Pysz-Hosey I, Procopiou G, Fox KR, Rahman KM, Thurston DE. A novel DNA sequence-selective, guanine mono-alkylating ADC payload suitable for solid tumour treatment. RSC Med Chem 2025:d4md01040j. [PMID: 40352674 PMCID: PMC12059774 DOI: 10.1039/d4md01040j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 03/01/2025] [Indexed: 05/14/2025] Open
Abstract
Pyridinobenzodiazepines (PDDs) are a new class of DNA mono-alkylating antibody-drug conjugate (ADC) payloads that can be linked through their C9 position to a sequence recognition component, guiding them to specific DNA sequences. Compound 18 is a PDD monomer with a unique sequence-selectivity profile and high cytotoxicity in vitro (e.g., IC50 = 0.30 nM in SW60; 1.6 nM in LIM1215 and 0.142 nM in SW48 cell line, after 96 hours incubation). To evaluate its potential as an ADC payload, an amine functionality was introduced into the terminal phenyl ring, and the modified compound was conjugated to trastuzumab (drug-antibody ratio [DAR] = 1.6). The resulting ADC exhibits significant in vivo efficacy in a pancreatic cancer xenograft model using BALB-c mice transplanted with the CAPAN-1 cell line. Complete tumour regression is observed out to 60 days after a single dose of 2 mg kg-1 comparing favourably to a 10 mg kg-1 dose of trastuzumab deruxtecan (Enhertu®). The novel ADC has a good tolerability profile, with a maximum tolerated dose (MTD) above 15 mg kg-1. The tolerability and efficacy profile of compound 18 in an ADC format suggests that PDDs represent a potentially valuable new class of payloads for the treatment of solid tumours.
Collapse
Affiliation(s)
- Paolo Andriollo
- School of Cancer & Pharmaceutical Sciences, King's College London WC2R 2LS UK +44 (0)20 7848 1891
| | | | - Paul J M Jackson
- School of Cancer & Pharmaceutical Sciences, King's College London WC2R 2LS UK +44 (0)20 7848 1891
| | - Md Mahbub Hasan
- School of Cancer & Pharmaceutical Sciences, King's College London WC2R 2LS UK +44 (0)20 7848 1891
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong Chattogram Bangladesh
| | - Ilona Pysz-Hosey
- School of Cancer & Pharmaceutical Sciences, King's College London WC2R 2LS UK +44 (0)20 7848 1891
| | - George Procopiou
- School of Cancer & Pharmaceutical Sciences, King's College London WC2R 2LS UK +44 (0)20 7848 1891
| | - Keith R Fox
- School of Biological Sciences, University of Southampton SO17 1BJ UK
| | - Khondaker Miraz Rahman
- School of Cancer & Pharmaceutical Sciences, King's College London WC2R 2LS UK +44 (0)20 7848 1891
| | - David E Thurston
- School of Cancer & Pharmaceutical Sciences, King's College London WC2R 2LS UK +44 (0)20 7848 1891
| |
Collapse
|
13
|
Jafari P, Forrest M, Segal J, Wang P, Tjota MY. Pan-Cancer Molecular Biomarkers: Practical Considerations for the Surgical Pathologist. Mod Pathol 2025; 38:100752. [PMID: 40058460 DOI: 10.1016/j.modpat.2025.100752] [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: 09/12/2024] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025]
Abstract
Traditional anatomic pathologic classification of cancer is based on tissue of origin and morphologic and immunohistochemical characterization of the malignant cells. With the technological improvements of massively parallel or next-generation sequencing, oncogenic drivers that are shared across different tumor types are increasingly being identified and used as pan-cancer biomarkers. This approach is reflected in the growing list of Food and Drug Administration-approved tumor-agnostic therapies, including pembrolizumab in the setting of microsatellite instability and high tumor mutational burden, larotrectinib and entrectinib for solid tumors with NTRK fusions, and combined dabrafenib-trametinib for BRAF V600E-mutated neoplasms. Several other biomarkers are currently under investigation, including fibroblast growth factor receptor (FGFR), RET, and ROS1 fusions; ERBB2 amplification; and mutations in the AKT1/2/3, NF1, RAS pathway and (mitogen-activated protein kinase (MAPK) pathway. As molecular assays are increasingly incorporated into routine tumor workup, the emergence of additional pan-cancer biomarkers is likely to be a matter more of "when" than "if." In this review, we first explore some of the conceptual and technical considerations at the intersection of surgical and molecular pathology, followed by a brief overview of both established and emerging molecular pan-cancer biomarkers and their diagnostic and clinical applications.
Collapse
Affiliation(s)
- Pari Jafari
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | - Megan Forrest
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | - Jeremy Segal
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | - Peng Wang
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | | |
Collapse
|
14
|
Giacoletto CJ, Valente LJ, Brown L, Patterson S, Gokhale R, Mockus SM, Grody WW, Deng HW, Rotter JI, Schiller MR. New Gain-of-Function Mutations Prioritize Mechanisms of HER2 Activation. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.03.25323043. [PMID: 40093211 PMCID: PMC11908269 DOI: 10.1101/2025.03.03.25323043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
ERBB2 (HER2) is a well-studied oncogene with several driver mutations apart from the well-known amplification defect in some breast cancers. We used the GigaAssay to test the functional effect of HER2 missense mutations on its receptor tyrosine kinase function. The GigaAssay is a modular high-throughput one-pot assay system for simultaneously measuring molecular function of thousands of genetic variants at very high accuracy. The activities of 5,886 mutations were classified, significantly more than mutants previously reported. These variants include 112 new in vitro, 10 known, and 9 new in vivo gain-of-function (GOF) mutations. Many of the GOFs spatially cluster in sequence and structure, supporting the activation mechanisms of heterodimerization with EGFR and release of kinase inhibition by the juxtamembrane domain. Retrospective analysis of patient outcomes from the Genomic Data Commons predicts increased survival with the newly identified HER2 GOF variants.
Collapse
Affiliation(s)
- Christopher J Giacoletto
- Heligenics Inc., 10530 Discovery Dr., Las Vegas, NV 89135 USA
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, 4505 S. Maryland Parkway, Las Vegas, Nevada, 89154 USA
- School of Life Sciences, University of Nevada, Las Vegas, 4505 S. Maryland Parkway, Las Vegas, Nevada, 89154 USA
| | - Liz J Valente
- Heligenics Inc., 10530 Discovery Dr., Las Vegas, NV 89135 USA
| | - Lancer Brown
- Heligenics Inc., 10530 Discovery Dr., Las Vegas, NV 89135 USA
| | - Sara Patterson
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Dr., Farmington, CT 06032
| | - Rewatee Gokhale
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Dr., Farmington, CT 06032
| | | | | | - Hong-Wen Deng
- Tulane Center for Biomedical Informatics and Genomics, Department of Deming Department of Medicine, Tulane University, New Orleans, 70112 USA
| | - Jerome I Rotter
- Heligenics Inc., 10530 Discovery Dr., Las Vegas, NV 89135 USA
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502 USA
| | - Martin R Schiller
- Heligenics Inc., 10530 Discovery Dr., Las Vegas, NV 89135 USA
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, 4505 S. Maryland Parkway, Las Vegas, Nevada, 89154 USA
- School of Life Sciences, University of Nevada, Las Vegas, 4505 S. Maryland Parkway, Las Vegas, Nevada, 89154 USA
| |
Collapse
|
15
|
Landi D, Navai SA, Brock RM, Fousek K, Nawas Z, Sanber K, Chauvin-Fleurence C, Bhat RR, Xu S, Krishnamurthy P, Choe M, Campbell ME, Morris JS, Gad AZ, Shree A, Echeandia Marrero AS, Saadeldin AM, Matthew PR, Mullikin D, Bielamowicz K, Kurenbekova L, Major AM, Salsman VS, Byrd TT, Hicks JM, Zhang YJ, Yustein J, Carisey AF, Joseph SK, Ahmed N, Hegde M. A Checkpoint Reversal Receptor Mediates Bipartite Activation and Enhances CAR T-cell Function. CANCER RESEARCH COMMUNICATIONS 2025; 5:527-548. [PMID: 39973814 PMCID: PMC11955954 DOI: 10.1158/2767-9764.crc-24-0125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 09/18/2024] [Accepted: 02/17/2025] [Indexed: 02/21/2025]
Abstract
SIGNIFICANCE Enhancing CART function and persistence while balancing immune effector-mediated inflammation is crucial. Using our clinically relevant HER2-CAR platform, we demonstrate that tumor-intrinsic signals like the PD-1/PD-L1 immune checkpoint can be leveraged in CART design to modulate immune synapse and metabolic parameters, improving antitumor function without increasing cytokine production.
Collapse
Affiliation(s)
- Daniel Landi
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Shoba A. Navai
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Rebecca M. Brock
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Kristen Fousek
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas
| | - Zeid Nawas
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Khaled Sanber
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Cynthia Chauvin-Fleurence
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Raksha R. Bhat
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Shuo Xu
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Purna Krishnamurthy
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Michelle Choe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Matthew E. Campbell
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Jessica S. Morris
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas
| | - Ahmed Z. Gad
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas
| | - Ankita Shree
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Alesandra S. Echeandia Marrero
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Amr M. Saadeldin
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Development, Disease Models and Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX
| | - Pretty R. Matthew
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Dolores Mullikin
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Kevin Bielamowicz
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Lyazat Kurenbekova
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Angela M. Major
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - Vita S. Salsman
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Tiara T. Byrd
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas
| | - John M. Hicks
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - Yi Jonathan Zhang
- Department of Neurosurgery, Houston Methodist Hospital, Houston, Texas
| | - Jason Yustein
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Alexandre F. Carisey
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Cell & Molecular Biology Department, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Sujith K. Joseph
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Nabil Ahmed
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Meenakshi Hegde
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
16
|
Beauchamp L, Indulkar S, Erak E, Salimian M, Matoso A. Tissue-Based Biomarkers Important for Prognostication and Management of Genitourinary Tumors, Including Surrogate Markers of Genomic Alterations. Surg Pathol Clin 2025; 18:175-189. [PMID: 39890303 DOI: 10.1016/j.path.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2025]
Abstract
A better understanding of the molecular alterations that underlie urologic malignancies and advances in targeted therapies has impacted classification, prognostication, and treatment. In bladder tumors, these advances include the development of antibody-drug conjugates targeting nectin-4 and Trop-2, as well as human epidermal growth factor receptor 2 and immunotherapy. In prostate cancer, assessment of the percentage of Gleason pattern 4, presence of cribriform glands, and molecular alterations, including PTEN and mismatch repair protein loss, have become standard for clinical care. In renal malignancies, alterations in TSC1/2, mammalian target of rapamycin, anaplastic lymphoma kinase, and other genes impact classification and therapeutic decisions.
Collapse
Affiliation(s)
- Leonie Beauchamp
- Departments of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA
| | - Shreeya Indulkar
- Departments of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA
| | - Eric Erak
- Departments of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA
| | - Mohammad Salimian
- Departments of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA
| | - Andres Matoso
- Departments of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA; Department of Urology, The Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA; Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA.
| |
Collapse
|
17
|
Francoeur AA, Monk BJ, Tewari KS. Treatment advances across the cervical cancer spectrum. Nat Rev Clin Oncol 2025; 22:182-199. [PMID: 39753753 DOI: 10.1038/s41571-024-00977-w] [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: 12/02/2024] [Indexed: 02/26/2025]
Abstract
Cervical cancer is preventable with screening and vaccination approaches; however, access to these preventative measures is limited both nationally and globally and thus many women will still develop cervical cancer. Novel treatments and practice-changing research have improved cervical cancer outcomes over the past few decades. In this Review, we discuss clinical trials that have refined or redefined the treatment of cervical cancers across the early stage, locally advanced, persistent, recurrent and/or metastatic disease settings. Advances for patients with early stage disease have been achieved through trials evaluating less extensive and fertility-preserving surgeries, different surgical approaches (open versus minimally invasive), and sentinel versus full pelvic lymph node dissection. We also discuss results from trials testing the use of neoadjuvant, induction and adjuvant chemotherapy as well as immune-checkpoint inhibitors in patients with locally advanced disease. Finally, we review the progress made with systemic chemotherapy and novel therapeutics, including anti-angiogenic agents, immune-checkpoint inhibitors and antibody-drug conjugates, in the setting of metastatic and/or recurrent cervical cancer. The advances highlighted in this manuscript have reduced morbidity and improved overall survival for patients with this challenging-to-treat disease, while also inspiring additional research and trials in the field.
Collapse
Affiliation(s)
- Alex A Francoeur
- Department of Obstetrics and Gynecology, University of California, Irvine, Irvine, CA, USA.
| | - Bradley J Monk
- Florida Cancer Specialists and Research Institute, West Palm Beach, FL, USA
| | - Krishnansu S Tewari
- Department of Obstetrics and Gynecology, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
18
|
Cortés-Salgado A, Moreno-Moreno E, Carretero-Barrio I, Caniego-Casas T, Cristóbal E, Del Campo-Albendea L, Guerra E, Alía V, de Aguado PP, Corraliza V, Palacios J, Pérez-Mies B. HER2 expression in a molecularly defined cohort of endometrial cancer patients: The SPECTRUM study. Gynecol Oncol 2025; 194:33-40. [PMID: 39947031 DOI: 10.1016/j.ygyno.2025.02.005] [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: 11/06/2024] [Revised: 01/30/2025] [Accepted: 02/05/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND HER2 antibody-drug conjugates like Trastuzumab deruxtecan (TDxd) have shown efficacy in HER2-low cancers, however the prevalence and clinical impact of HER2-low in endometrial cancer (EC) remains unclear. This study investigates HER2 expression in EC, focusing on HER2-low frequency, its distribution across histologic and molecular subtypes, intratumoral heterogeneity, and prognostic significance. METHODS This retrospective analysis included EC patients categorized histologically and molecularly, using ASCO/CAP HER2 scoring guidelines for endometrial, gastric, and breast cancers. The main aim was to determine HER2-low frequency by molecular subtype, with secondary goals of assessing HER2 heterogeneity, changes between primary and recurrent tumors, guidelines concordance and the impact of HER2 status on recurrence-free (RFS) and overall survival (OS). RESULTS 193 EC patients were included, 111 low grade endometrioid EC and 82 high grade EC with various histotypes. Based on endometrial criteria, 69.9 % were HER2-negative, 25.8 % HER2-low, and 4.3 % HER2-positive, with HER2-low being most common in POLEmut (66.7 %) and p53abn (45.1 %) subtypes. Low heterogeneity in HER2 expression was observed between different tumors areas and between primary tumors and their recurrences. The endometrial and gastric criteria had high concordance (88.7 %), with endometrial guidelines detecting more HER2-low cases. Neither HER2-low nor HER2-positivity versus HER2-negativity had an impact on RFS or OS, with the stage at diagnosis being the only factor associated with survival, while age was associated only with OS. CONCLUSIONS This study highlights the distribution of HER2-low across EC molecular subtypes, with endometrial criteria proving more sensitive for identifying HER2-low cases, though HER2 status had no prognostic value in this cohort.
Collapse
Affiliation(s)
- Alfonso Cortés-Salgado
- Department of Medical Oncology, Ramón y Cajal University Hospital, Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
| | - Esther Moreno-Moreno
- Department of Pathology, Ramón y Cajal University Hospital, Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain; Faculty of Medicine, Alcalá University, Alcalá de Henares, Spain
| | - Irene Carretero-Barrio
- Department of Pathology, Ramón y Cajal University Hospital, Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain; Faculty of Medicine, Alcalá University, Alcalá de Henares, Spain; Centre for Biomedical Research in Cancer Networks (CIBERONC), Carlos III Health Institute, Madrid, Spain
| | - Tamara Caniego-Casas
- Department of Pathology, Ramón y Cajal University Hospital, Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain; Centre for Biomedical Research in Cancer Networks (CIBERONC), Carlos III Health Institute, Madrid, Spain
| | - Eva Cristóbal
- Department of Pathology, Ramón y Cajal University Hospital, Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain; Centre for Biomedical Research in Cancer Networks (CIBERONC), Carlos III Health Institute, Madrid, Spain
| | - Laura Del Campo-Albendea
- Clinical Biostatistics Unit, Ramón y Cajal University Hospital, Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain; Centre for Biomedical Research in Epidemiology and Public Health (CIBERESP), Carlos III Health Institute, Madrid, Spain
| | - Eva Guerra
- Department of Medical Oncology, Ramón y Cajal University Hospital, Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
| | - Víctor Alía
- Department of Medical Oncology, Ramón y Cajal University Hospital, Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
| | - Patricia Pérez de Aguado
- Department of Medical Oncology, Ramón y Cajal University Hospital, Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
| | - Virginia Corraliza
- Department of Obstetrics and Gynecology, Ramón y Cajal University Hospital, Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
| | - José Palacios
- Department of Pathology, Ramón y Cajal University Hospital, Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain; Faculty of Medicine, Alcalá University, Alcalá de Henares, Spain; Centre for Biomedical Research in Cancer Networks (CIBERONC), Carlos III Health Institute, Madrid, Spain
| | - Belén Pérez-Mies
- Department of Pathology, Ramón y Cajal University Hospital, Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain; Faculty of Medicine, Alcalá University, Alcalá de Henares, Spain; Centre for Biomedical Research in Cancer Networks (CIBERONC), Carlos III Health Institute, Madrid, Spain.
| |
Collapse
|
19
|
Houvast RD, van Duijvenvoorde M, Thijse K, de Steur WO, de Geus-Oei LF, Crobach ASLP, Burggraaf J, Vahrmeijer AL, Kuppen PJK. Selecting Targets for Molecular Imaging of Gastric Cancer: An Immunohistochemical Evaluation. Mol Diagn Ther 2025; 29:213-227. [PMID: 39541080 PMCID: PMC11860997 DOI: 10.1007/s40291-024-00755-5] [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: 10/23/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE Tumor-targeted positron emission tomography (PET) and fluorescence-guided surgery (FGS) could address current challenges in pre- and intraoperative imaging of gastric cancer. Adequate selection of molecular imaging targets remains crucial for successful tumor visualization. This study evaluated the potential of integrin αvβ6, carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5), epidermal growth factor receptor (EGFR), epithelial cell adhesion molecule (EpCAM) and human epidermal growth factor receptor-2 (HER2) for molecular imaging of primary gastric cancer, as well as lymph node and distant metastases. METHODS Expression of αvβ6, CEACAM5, EGFR, EpCAM and HER2 was determined using immunohistochemistry in human tissue specimens of primary gastric adenocarcinoma, healthy surrounding stomach, esophageal and duodenal tissue, tumor-positive and tumor-negative lymph nodes, and distant metastases, followed by quantification using the total immunostaining score (TIS). RESULTS Positive biomarker expression in primary gastric tumors was observed in 86% for αvβ6, 72% for CEACAM5, 77% for EGFR, 93% for EpCAM and 71% for HER2. Tumor expression of CEACAM5, EGFR and EpCAM was higher compared to healthy stomach tissue expression, while this was not the case for αvβ6 and HER2. Tumor-positive lymph nodes could be distinguished from tumor-negative lymph nodes, with accuracy ranging from 82 to 93% between biomarkers. CEACAM5, EGFR and EpCAM were abundantly expressed on distant metastases, with expression in 88-95% of tissue specimens. CONCLUSION Our findings show that CEACAM5, EGFR and EpCAM are promising targets for molecular imaging of primary gastric cancer, as well as visualization of both lymph node and distant metastases. Further clinical evaluation of PET and FGS tracers targeting these antigens is warranted.
Collapse
Affiliation(s)
- Ruben D Houvast
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands.
| | | | - Kira Thijse
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Wobbe O de Steur
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Lioe-Fee de Geus-Oei
- Department of Radiation Science & Technology, Delft University of Technology, Delft, The Netherlands
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Biomedical Photonic Imaging Group, University of Twente, Enschede, The Netherlands
| | - A Stijn L P Crobach
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jacobus Burggraaf
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
- Centre for Human Drug Research, Leiden, The Netherlands
| | | | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
20
|
Kanwal W, Narjis K, Musani S, Nancy F, Qureshi L, Mudasir M, Naseem R, Tooba F, Yousuf J, Farhan K, Javed H, Eljack MMF. Exploring Zanidatamab's efficacy across HER2-positive Malignancies: a narrative review. BMC Cancer 2025; 25:382. [PMID: 40025472 PMCID: PMC11871714 DOI: 10.1186/s12885-025-13749-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 02/17/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND HER2-positive cancers involve amplification or overexpression of the HER2 gene, leading to aggressive tumor growth across several cancer types, including breast, gastric, ovarian, and pancreatic cancers. Detection methods such as immunohistochemistry, next-generation sequencing, and fluorescence in situ hybridization are used, with new advancements like biosensors and circulating tumor DNA offering improved diagnostic potential. Treatment strategies have evolved, including anti-HER2 drugs like trastuzumab and newer agents like zanidatamab, which show promise against HER2-positive malignancies. METHODS A comprehensive search of the following academic databases was performed including PubMed, Cochrane Library, and clinicaltrials.gov. A detailed search string was made. Studies were selected based on whether they contained the keywords and if they reported the details of treatment for zanidatamab. A total of 16 studies were selected. Abstracts were independently examined by one author and critically reviewed by another and if there were any conflicting viewpoints they were discussed until consensus was reached. DISCUSSION Zanidatamab has shown promising clinical outcomes in several HER2-positive cancers, including biliary tract, breast, gastric, and lung cancers, with high disease control rates and progression-free survival. Although it is not yet FDA-approved, it has received priority review for HER2-positive biliary tract cancer, with an FDA decision expected in November 2024. The safety profile of zanidatamab has been well-studied. The most common side effects include diarrhea, infusion reactions, and other mild to moderate treatment-related adverse events. In combination with Palbociclib for HER2-positive breast cancer, more severe side effects were observed, resulting in some patients discontinuing treatment. However, no treatment-related deaths have been reported across trials. While its anticancer efficacy and manageable safety profile are promising, long-term safety and efficacy data are still needed. Early clinical trials demonstrate strong efficacy, though some side effects, such as diarrhea and decreased ejection fraction, were noted. Future research should focus on understanding potential resistance mechanisms and establishing zanidatamab's broader role in the treatment landscape of HER2-positive cancers. CONCLUSION In summary, zanidatamab has shown significant tumor response, progression-free survival, disease control, and improved quality of life in early trials, however, the lack of long-term safety and efficacy data remains a key limitation, requiring further research.
Collapse
Affiliation(s)
| | - Kaneez Narjis
- Isra University of Medical Sciences, Hyderabad, Pakistan
| | - Sarah Musani
- Dow University of Health Sciences, Karachi, Pakistan
| | - Fnu Nancy
- Jinnah Sindh Medical University, Karachi, Pakistan
| | - Laiba Qureshi
- Isra University of Medical Sciences, Hyderabad, Pakistan
| | - Muhammad Mudasir
- Liaquat University of Medical and Health Sciences, Jamshoro, Pakistan
| | - Rohma Naseem
- Liaquat National Hospital and Medical College, Karachi, Pakistan
| | - Fnu Tooba
- Hamdard University, Karachi, Pakistan
| | | | - Kanza Farhan
- Jinnah Sindh Medical University, Karachi, Pakistan
| | - Hadiya Javed
- Dow University of Health Sciences, Karachi, Pakistan
| | | |
Collapse
|
21
|
Aro K, Loukovaara M, Bützow R, Pasanen A. HER2 amplification and HER2 low expression in endometrial carcinoma: prevalence across molecular, histological and clinicopathological risk groups. BJC REPORTS 2025; 3:8. [PMID: 39939712 PMCID: PMC11821901 DOI: 10.1038/s44276-025-00125-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/22/2024] [Accepted: 01/28/2025] [Indexed: 02/14/2025]
Abstract
BACKGROUND Emerging HER2-targeted therapies provide new treatment options for patients with HER2-expressing tumors. This study investigates the prevalence of HER2 amplification and HER2 low expression across a well-characterized cohort of endometrial carcinoma. METHODS HER2 chromogenic in situ hybridization (CISH) was used to detect HER2 amplification in endometrial carcinoma samples. Chromogenic HER2 immunohistochemistry (IHC) was performed. HER2 low was defined as IHC 1 + /2+ and negative CISH. RESULTS CISH confirmed HER2 amplification in 2% (n = 26) of the 1239 endometrial carcinoma samples including all the IHC 3+ cases (n = 13) and 20% of the 2+ cases (n = 55). Amplified cases presented various histotypes but consisted almost exclusively of p53 abnormal tumors. HER2 low 2+ category (n = 44) was heterogeneous with regard to molecular subgroup and histotype with 64.3% of the patients having high-risk disease. HER2 status did not independently predict disease-specific survival. CONCLUSIONS p53 abnormal molecular subgroup predicts HER2 amplification better than histotype. HER2 low cases present a wide range of histotypes and molecular subgroups including many patients with high-risk uterine cancer. Future trials of anti-HER2 therapies will clarify the clinical relevance of HER2 low status, treatment indications and guidelines for HER2 testing in endometrial carcinoma.
Collapse
Affiliation(s)
- Karoliina Aro
- Department of Obstetrics and Gynecology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland.
| | - Mikko Loukovaara
- Department of Obstetrics and Gynecology and Comprehensive Cancer Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Ralf Bützow
- Department of Obstetrics and Gynecology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- Department of Pathology, Helsinki University Hospital and Research Program in Applied Tumor Genomics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Annukka Pasanen
- Department of Pathology, Helsinki University Hospital and Research Program in Applied Tumor Genomics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
22
|
Piha-Paul SA, Tseng C, Tran HT, Naing A, Dumbrava EE, Karp DD, Rodon J, Yap TA, Raghav KP, Damodaran S, Le X, Soliman PT, Lim J, Meric-Bernstam F. Phase I trial of the combination of the pan-ErbB inhibitor neratinib and mTOR inhibitor everolimus in advanced cancer patients with ErbB family gene alterations. ESMO Open 2025; 10:104136. [PMID: 39908697 PMCID: PMC11847258 DOI: 10.1016/j.esmoop.2025.104136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/25/2024] [Accepted: 01/08/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND The ErbB family of receptor tyrosine kinases are key targets for antitumor therapy. Although neratinib, a pan-ErbB kinase inhibitor, is approved in ErbB2-positive breast cancer, drug resistance is common. Preclinical data suggest that combining neratinib with the mTOR inhibitor everolimus may overcome such resistance. PATIENTS AND METHODS Our trial evaluated this combination's safety and efficacy in advanced cancers with ErbB alterations. We conducted a phase I dose-escalation trial of neratinib and everolimus. Primary objectives were to assess safety, tolerability, and dose-limiting toxicities (DLTs) and establish the maximum tolerated dose (MTD). Secondary objectives included objective response by RECIST v1.1 and pharmacokinetic analyses. RESULTS Twenty-two patients (median age 61, median of four prior therapies) with ErbB alterations (mutations 63.6%, amplification 36.3%, or ErbB2-overexpressed by immunohistochemistry 9.1%) were enrolled. Common tumor types included breast (31.8%), colorectal (18.2%), cervical (9.1%), and endometrial (9.1%) cancers. Frequent grade (G) 3 treatment-related adverse events were diarrhea (18.2%), anemia (9.1%), mucositis (9.1%), and acute kidney injury (9.1%). DLTs included G3 mucositis and diarrhea at dose level (DL) 5, and G3 increased creatinine at DL4. The MTD was DL4: neratinib 240 mg with everolimus 7.5 mg. The objective response rate was 19% with partial response in four patients. Stable disease ≥16 weeks was seen in two patients (9.5%), resulting in a clinical benefit rate of 28.6%. CONCLUSION Pharmacokinetic data indicated reduced neratinib clearance possibly due to CYP3A4 pathway saturation by everolimus. Combination therapy with neratinib and everolimus has a tolerable safety profile and clinical activity in ErbB-altered patients. ErbB family receptors and the PI3K pathway are commonly implicated in oncogenesis. This clinical study of neratinib and everolimus demonstrated favorable clinical activity and tolerability.
Collapse
Affiliation(s)
- S A Piha-Paul
- Department of Investigational Cancer Therapeutics (A Phase I Clinical Trials Program), University of Texas MD Anderson Cancer Center, Houston, USA.
| | - C Tseng
- Department of Investigational Cancer Therapeutics (A Phase I Clinical Trials Program), University of Texas MD Anderson Cancer Center, Houston, USA
| | - H T Tran
- Department of Thoracic, Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
| | - A Naing
- Department of Investigational Cancer Therapeutics (A Phase I Clinical Trials Program), University of Texas MD Anderson Cancer Center, Houston, USA
| | - E E Dumbrava
- Department of Investigational Cancer Therapeutics (A Phase I Clinical Trials Program), University of Texas MD Anderson Cancer Center, Houston, USA
| | - D D Karp
- Department of Investigational Cancer Therapeutics (A Phase I Clinical Trials Program), University of Texas MD Anderson Cancer Center, Houston, USA
| | - J Rodon
- Department of Investigational Cancer Therapeutics (A Phase I Clinical Trials Program), University of Texas MD Anderson Cancer Center, Houston, USA; The Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, University of Texas MD Anderson Cancer Center, Houston, USA
| | - T A Yap
- Department of Investigational Cancer Therapeutics (A Phase I Clinical Trials Program), University of Texas MD Anderson Cancer Center, Houston, USA; The Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, University of Texas MD Anderson Cancer Center, Houston, USA; Therapeutics Discovery Division, University of Texas MD Anderson Cancer Center, Houston, USA
| | - K P Raghav
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
| | - S Damodaran
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
| | - X Le
- Department of Thoracic, Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
| | - P T Soliman
- Gynecologic Oncology & Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, USA
| | - J Lim
- Pharmacy Clinic Programs, Division of Pharmacy, University of Texas MD Anderson Cancer Center, Houston, USA
| | - F Meric-Bernstam
- Department of Investigational Cancer Therapeutics (A Phase I Clinical Trials Program), University of Texas MD Anderson Cancer Center, Houston, USA; The Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, University of Texas MD Anderson Cancer Center, Houston, USA; Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
| |
Collapse
|
23
|
Ji P, Chen T, Li C, Zhang J, Li X, Zhu H. Comprehensive review of signaling pathways and therapeutic targets in gastrointestinal cancers. Crit Rev Oncol Hematol 2025; 206:104586. [PMID: 39653094 DOI: 10.1016/j.critrevonc.2024.104586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024] Open
Abstract
Targeted therapy, the milestone in the development of human medicine, originated in 2004 when the FDA approved the first targeted agent bevacizumab for colorectal cancer treatment. This new development has resulted from drug developers moving beyond traditional chemotherapy, and several trials have popped up in the last two decades with an unprecedented speed. Specifically, EGF/EGFR, VEGF/VEGFR, HGF/c-MET, and Claudin 18.2 therapeutic targets have been developed in recent years. Some targets previously thought to be undruggable are now being newly explored, such as the RAS site. However, the efficacy of targeted therapy is extremely variable, especially with the emergence of new drugs and the innovative use of traditional targets for other tumors in recent years. Accordingly, this review provides an overview of the major signaling pathway mechanisms and recent advances in targeted therapy for gastrointestinal cancers, as well as future perspectives.
Collapse
Affiliation(s)
- Pengfei Ji
- Department of Thoracic Surgery, West China Hospital, Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China
| | - Tingting Chen
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Chao Li
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Jinyuan Zhang
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Xiao Li
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| |
Collapse
|
24
|
Korpan M, Puhr HC, Berger JM, Friedrich A, Prager GW, Preusser M, Ilhan-Mutlu A. Current Landscape of Molecular Biomarkers in Gastroesophageal Tumors and Potential Strategies for Co-Expression Patterns. Cancers (Basel) 2025; 17:340. [PMID: 39941712 PMCID: PMC11816248 DOI: 10.3390/cancers17030340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/14/2025] [Accepted: 01/18/2025] [Indexed: 02/16/2025] Open
Abstract
The treatment of metastasized gastroesophageal adenocarcinoma largely depends on molecular profiling based on immunohistochemical procedures. Therefore, the examination of HER2, PD-L1, and dMMR/MSI is recommended by the majority of clinical practice guidelines, as positive expression leads to different treatment approaches. Data from large phase-III trials and consequent approvals in various countries enable physicians to offer their patients several therapy options including immunotherapy, targeted therapy, or both combined with chemotherapy. The introduction of novel therapeutic targets such as CLDN18.2 leads to a more complex decision-making process as a significant number of patients show positive results for the co-expression of other biomarkers besides CLDN18.2. The aim of this review is to summarize the current biomarker landscape of patients with metastatic gastroesophageal tumors, its direct clinical impact on daily decision-making, and to evaluate current findings on biomarker co-expression. Furthermore, possible treatment strategies with multiple biomarker expression are discussed.
Collapse
Affiliation(s)
- Martin Korpan
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Hannah Christina Puhr
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Julia M. Berger
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Alexander Friedrich
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Gerald W. Prager
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Aysegül Ilhan-Mutlu
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
25
|
Zhan T, Betge J, Schulte N, Dreikhausen L, Hirth M, Li M, Weidner P, Leipertz A, Teufel A, Ebert MP. Digestive cancers: mechanisms, therapeutics and management. Signal Transduct Target Ther 2025; 10:24. [PMID: 39809756 PMCID: PMC11733248 DOI: 10.1038/s41392-024-02097-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 10/20/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
Cancers of the digestive system are major contributors to global cancer-associated morbidity and mortality, accounting for 35% of annual cases of cancer deaths. The etiologies, molecular features, and therapeutic management of these cancer entities are highly heterogeneous and complex. Over the last decade, genomic and functional studies have provided unprecedented insights into the biology of digestive cancers, identifying genetic drivers of tumor progression and key interaction points of tumor cells with the immune system. This knowledge is continuously translated into novel treatment concepts and targets, which are dynamically reshaping the therapeutic landscape of these tumors. In this review, we provide a concise overview of the etiology and molecular pathology of the six most common cancers of the digestive system, including esophageal, gastric, biliary tract, pancreatic, hepatocellular, and colorectal cancers. We comprehensively describe the current stage-dependent pharmacological management of these malignancies, including chemo-, targeted, and immunotherapy. For each cancer entity, we provide an overview of recent therapeutic advancements and research progress. Finally, we describe how novel insights into tumor heterogeneity and immune evasion deepen our understanding of therapy resistance and provide an outlook on innovative therapeutic strategies that will shape the future management of digestive cancers, including CAR-T cell therapy, novel antibody-drug conjugates and targeted therapies.
Collapse
Affiliation(s)
- Tianzuo Zhan
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Johannes Betge
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Junior Clinical Cooperation Unit Translational Gastrointestinal Oncology and Preclinical Models, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nadine Schulte
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lena Dreikhausen
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Michael Hirth
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Moying Li
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Philip Weidner
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Antonia Leipertz
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Andreas Teufel
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Matthias P Ebert
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany.
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| |
Collapse
|
26
|
Marei HE, Bedair K, Hasan A, Al-Mansoori L, Caratelli S, Sconocchia G, Gaiba A, Cenciarelli C. Current status and innovative developments of CAR-T-cell therapy for the treatment of breast cancer. Cancer Cell Int 2025; 25:3. [PMID: 39755633 PMCID: PMC11700463 DOI: 10.1186/s12935-024-03615-8] [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: 07/15/2024] [Accepted: 12/12/2024] [Indexed: 01/06/2025] Open
Abstract
Breast cancer will overtake all other cancers in terms of diagnoses in 2024. Breast cancer counts highest among women in terms of cancer incidence and death rates. Innovative treatment approaches are desperately needed because treatment resistance brought on by current clinical drugs impedes therapeutic efficacy. The T cell-based immunotherapy known as chimeric antigen receptor (CAR) T cell treatment, which uses the patient's immune cells to fight cancer, has demonstrated remarkable efficacy in treating hematologic malignancies; nevertheless, the treatment effects in solid tumors, like breast cancer, have not lived up to expectations. We discuss in detail the role of tumor-associated antigens in breast cancer, current clinical trials, barriers to the intended therapeutic effects of CAR-T cell therapy, and potential ways to increase treatment efficacy. Finally, our review aims to stimulate readers' curiosity by summarizing the most recent advancements in CAR-T cell therapy for breast cancer.
Collapse
Affiliation(s)
- Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35116, Egypt.
| | - Khaled Bedair
- Department of Social Sciences, College of Arts and Sciences, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, Qatar
| | - Layla Al-Mansoori
- Biomedical Research Center, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Sara Caratelli
- Institute of Translational Pharmacology-CNR, Rome, Italy
| | | | - Alice Gaiba
- Institute of Translational Pharmacology-CNR, Rome, Italy
| | | |
Collapse
|
27
|
Qian X, Jin X, He J, Zhang J, Hu S. Exploring lipidomic profiles and their correlation with hormone receptor and HER2 status in breast cancer. Oncol Lett 2025; 29:34. [PMID: 39512509 PMCID: PMC11542162 DOI: 10.3892/ol.2024.14781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/26/2024] [Indexed: 11/15/2024] Open
Abstract
Dysregulated lipid metabolism promotes the progression of various cancer types, including breast cancer. The present study aimed to explore the lipidomic profiles of patients with breast cancer, providing insights into the correlation between lipid compositions and tumor subtypes characterized by hormone receptor (HR) and human epidermal growth factor receptor 2 (HER2) status. Briefly, 30 patients with breast cancer were categorized into four groups based on their HR and HER2 status: HR+ no HER2 expression (HER2-0), HR+ HER2-low; HR+ HER2-positive (pos) and HR- HER2-pos. The lipidomic profiles of these patients were analyzed using high-throughput liquid chromatography-mass spectrometry. The data were processed through principal component analysis (PCA), partial least squares-discriminant analysis (PLS-DA) and random forest (RF) classification to assess the lipidomic variations and significant lipid features among these groups. The profiles of the lipids, particularly triglycerides (TG) such as TG(16:0-18:1-18:1)+NH4, were significantly different across the groups. PCA and PLS-DA identified unique lipid profiles in the HR+ HER2-pos and HR+ HER2-0 groups, while RF highlighted phosphatidylinositol-3,4,5-trisphosphate(21:2)+NH4 as a crucial lipid feature for accurate patient grouping. Advanced statistical analysis showed significant correlations between lipid carbon chain length and the number of double bonds within the classifications, providing insights into the role of structural lipid properties in tumor biology. Additionally, a clustering heatmap and network analysis indicated significant lipid-lipid interactions. Pathway enrichment analysis showed critical biological pathways, such as the 'Assembly of active LPL and LIPC lipase complexes', which has high enrichment ratio and statistical significance. In conclusion, the present study underscored that lipidomic profiling is crucial in understanding the metabolic alterations associated with different breast cancer subtypes. These findings highlighted specific lipid features and interactions that may serve as potential biomarkers for breast cancer classification and target pathways for therapeutic intervention. Furthermore, advanced lipidomic analyses can be integrated to decipher complex biological data, offering a foundation for further research into the role of lipid metabolism in cancer progression.
Collapse
Affiliation(s)
- Xiaojun Qian
- Department of Breast and Thyroid Surgery, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Xiaolin Jin
- Health Management Center, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Jiaying He
- Health Management Center, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Junjing Zhang
- Health Management Center, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Shan Hu
- Department of Breast and Thyroid Surgery, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| |
Collapse
|
28
|
Cai X, Cao M, Yang Q, Yu X, Feng XH, Zhao RY. HER2-targeted ADC DX126-262 combined with chemotherapy demonstrates superior antitumor efficacy in HER2-positive gastric cancer. Am J Cancer Res 2024; 14:5752-5768. [PMID: 39803638 PMCID: PMC11711518 DOI: 10.62347/qcdr9612] [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: 09/18/2024] [Accepted: 11/14/2024] [Indexed: 01/16/2025] Open
Abstract
Gastric cancer is a common malignant tumor with high incidence and mortality. The overexpression of Human epidermal growth factor receptor 2 (HER2) is associated with increased metastatic potential and poor clinical outcome in gastric cancer. Despite the proven clinical response rates of approved HER2-targeted therapies, including Trastuzumab combined with chemotherapy, their limited long-term clinical benefits and inevitable disease progression still pose significant challenges to the clinical treatment of gastric cancer. Hence, exploring novel strategies to enhance therapeutic outcomes for HER2-positive patients is extremely crucial and urgent. Here, we reported that DX126-262, a novel HER2-targeted antibody-drug conjugate, generated by conjugating a potent Tubulysin B analogue (Tub-114) to humanized anti-HER2 monoclonal antibody, exhibited a significant synergistic inhibitory effect with both Cisplatin and 5-FU in HER2-positive gastric cancer NCI-N87 cells. Moreover, the triple-drug combination strategy of DX126-262 combined with Cisplatin and 5-FU showed much better in vitro and in vivo therapeutic efficacy than monotherapy or double-drug combination (Cisplatin plus 5-FU) or first-line standard-of-care (SOC, Herceptin plus Cisplatin and 5-FU), and comparable or even superior in vivo efficacy than third-line SOC (DS-8201a) in NCI-N87 cells and xenograft models. Meanwhile, the triple-drug combination therapy did not exhibit superimposed toxicity. Taken together, our findings provide compelling evidence that DX126-262 in combination with Cisplatin and 5-FU exerts synergistic antitumor activity and is a promising strategy to improve the clinical efficacy of HER2-positive advanced or metastatic gastric cancer.
Collapse
Affiliation(s)
- Xiaobo Cai
- Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang UniversityHangzhou 310058, Zhejiang, China
- Hangzhou DAC Biotechnology Co., Ltd.No. 369 Qiaoxin Road, Qiantang District, Hangzhou 310018, Zhejiang, China
| | - Min Cao
- Hangzhou DAC Biotechnology Co., Ltd.No. 369 Qiaoxin Road, Qiantang District, Hangzhou 310018, Zhejiang, China
| | - Qingliang Yang
- Hangzhou DAC Biotechnology Co., Ltd.No. 369 Qiaoxin Road, Qiantang District, Hangzhou 310018, Zhejiang, China
| | - Xiazhen Yu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhou 310003, Zhejiang, China
| | - Xin-Hua Feng
- Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang UniversityHangzhou 310058, Zhejiang, China
| | - Robert Yongxin Zhao
- Hangzhou DAC Biotechnology Co., Ltd.No. 369 Qiaoxin Road, Qiantang District, Hangzhou 310018, Zhejiang, China
| |
Collapse
|
29
|
Papageorgiou D, Liouta G, Sapantzoglou I, Zachariou E, Pliakou D, Papakonstantinou K, Floros T, Pliakou E. HER2-Positive Serous Endometrial Cancer Treatment: Current Clinical Practice and Future Directions. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:2012. [PMID: 39768892 PMCID: PMC11728157 DOI: 10.3390/medicina60122012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/29/2024] [Accepted: 12/04/2024] [Indexed: 01/16/2025]
Abstract
The most common histological subtypes of endometrial cancer consist of endometrioid and uterine serous carcinoma, with the latter being more aggressive and accompanied by poor prognosis. Human epidermal growth factor receptor 2 (HER2) is a transmembrane tyrosine kinase receptor associated with cell proliferation, differentiation, and survival. HER2 positivity can be diagnosed in many solid tumors. It has been found that approximately one-third of the patients diagnosed with serous carcinoma may overexpress HER2/neu protein and/or show the amplification of the c-erBb2 gene. The prognostic and predictive value of HER2 biomarker is nowadays highlighted and the updates of HER2-directed treatment offer new opportunities for improved efficacy and survival. A number of HER2-targeted therapies have become available in recent years and have had promising results, prompting full drug approvals and additional investigation in many cancer types, among which is endometrial cancer. Data from clinical trials combining classical chemotherapy with anti-HER2 agents, mainly trastuzumab, alone or in combination with pertuzumab, do exist and have been incorporated into international guidelines. Moreover, further research with antibody-drug conjugates and tyrosine kinase inhibitors is being conducted. Acquired resistance remains an important problem, and its underlying mechanisms in endometrial cancer are mostly unknown. Studies exploring earlier use of Her2-directed therapy are also on the way. The purpose of this literature review is to describe the available therapies in the current clinical practice and the most prominent research data regarding the future. In any case, a number of unmet medical needs do exist for HER2-positive serous endometrial cancer, and additional research and studies are warranted to provide further understanding and improved outcomes for this tumor type.
Collapse
Affiliation(s)
- Dimitrios Papageorgiou
- Department of Gynecology, Athens Naval and Veterans Hospital, 115 21 Athens, Greece; (D.P.); (K.P.)
| | - Galateia Liouta
- Department of Medical Oncology, General Oncology Hospital of Kifissia “Agioi Anargiroi”, 145 64 Athens, Greece;
| | - Ioakeim Sapantzoglou
- 1st Department of Obstetrics and Gynecology, Alexandra Hospital, National and Kapodistrian University of Athens, 115 28 Athens, Greece;
| | - Eleftherios Zachariou
- 1st Department of Gynecology, Division of Robotic and Laparoscopic Surgery, Metropolitan General Hospital, 155 62 Athens, Greece;
| | - Dimitra Pliakou
- Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece;
| | | | - Theofanis Floros
- 5th Department of Oncology, Metropolitan General Hospital, 155 62 Athens, Greece;
| | - Evangelia Pliakou
- 5th Department of Oncology, Metropolitan General Hospital, 155 62 Athens, Greece;
| |
Collapse
|
30
|
Hwang SY, Jeon KH, Lee HJ, Moon I, Jung S, Kim SA, Jo H, Park S, Ahn M, Kwak SY, Na Y, Kwon Y. Synthesis and biological assessment of chalcone and pyrazoline derivatives as novel inhibitor for ELF3-MED23 interaction. eLife 2024; 13:RP97051. [PMID: 39641248 PMCID: PMC11623927 DOI: 10.7554/elife.97051] [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] [Indexed: 12/07/2024] Open
Abstract
HER2 overexpression significantly contributes to the aggressive nature and recurrent patterns observed in various solid tumors, notably gastric cancers. Trastuzumab, HER2-targeting monoclonal antibody drug, has shown considerable clinical success; however, readily emerging drug resistance emphasizes the pressing need for improved interventions in HER2-overexpressing cancers. To address this, we proposed targeting the protein-protein interaction (PPI) between ELF3 and MED23 as an alternative therapeutic approach to trastuzumab. In this study, we synthesized a total of 26 compounds consisting of 10 chalcones, 7 pyrazoline acetyl, and 9 pyrazoline propionyl derivatives, and evaluated their biological activity as potential ELF3-MED23 PPI inhibitors. Upon systematic analysis, candidate compound 10 was selected due to its potency in downregulating reporter gene activity of ERBB2 promoter confirmed by SEAP activity and its effect on HER2 protein and mRNA levels. Compound 10 effectively disrupted the binding interface between the ELF3 TAD domain and the 391-582 amino acid region of MED23, resulting in successful inhibition of the ELF3-MED23 PPI. This intervention led to a substantial reduction in HER2 levels and its downstream signals in the HER2-positive gastric cancer cell line. Subsequently, compound 10 induced significant apoptosis and anti-proliferative effects, demonstrating superior in vitro and in vivo anticancer activity overall. We found that the anticancer activity of compound 10 was not only restricted to trastuzumab-sensitive cases, but was also valid for trastuzumab-refractory clones. This suggests its potential as a viable therapeutic option for trastuzumab-resistant gastric cancers. In summary, compound 10 could be a novel alternative therapeutic strategy for HER2-overexpressing cancers, overcoming the limitations of trastuzumab.
Collapse
Affiliation(s)
- Soo-Yeon Hwang
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans UniversitySeoulRepublic of Korea
| | - Kyung-Hwa Jeon
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans UniversitySeoulRepublic of Korea
| | - Hwa-Jong Lee
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans UniversitySeoulRepublic of Korea
| | - Inhye Moon
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans UniversitySeoulRepublic of Korea
| | - Sehyun Jung
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans UniversitySeoulRepublic of Korea
| | - Seul-Ah Kim
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans UniversitySeoulRepublic of Korea
| | - Hyunji Jo
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans UniversitySeoulRepublic of Korea
| | - Seojeong Park
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans UniversitySeoulRepublic of Korea
| | - Misun Ahn
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans UniversitySeoulRepublic of Korea
| | - Soo-Yeon Kwak
- College of Pharmacy, CHA UniversityPocheonRepublic of Korea
| | - Younghwa Na
- College of Pharmacy, CHA UniversityPocheonRepublic of Korea
| | - Youngjoo Kwon
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans UniversitySeoulRepublic of Korea
| |
Collapse
|
31
|
Boussios S, Sheriff M, Ovsepian SV. Molecular Biology of Cancer-Interplay of Malignant Cells with Emerging Therapies. Int J Mol Sci 2024; 25:13090. [PMID: 39684799 DOI: 10.3390/ijms252313090] [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: 11/19/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Cancer is currently one of the leading causes of death worldwide, and according to data from the World Health Organization reported in 2020, it ranks as the second leading cause of death globally, accounting for 10 million fatalities [...].
Collapse
Affiliation(s)
- Stergios Boussios
- Faculty of Medicine, Health and Social Care, Canterbury Christ Church University, Canterbury CT1 1QU, UK
- Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, King's College London, Strand, London WC2R 2LS, UK
- Kent Medway Medical School, University of Kent, Canterbury CT2 7LX, UK
- AELIA Organization, 9th Km Thessaloniki-Thermi, 57001 Thessaloniki, Greece
- Department of Medical Oncology, Medway NHS Foundation Trust, Gillingham ME7 5NY, UK
| | - Matin Sheriff
- Department of Urology, Medway NHS Foundation Trust, Gillingham ME7 5NY, UK
| | - Saak V Ovsepian
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Gillingham ME4 4AG, UK
- Faculty of Medicine, Tbilisi State University, Tbilisi 0179, Georgia
| |
Collapse
|
32
|
Andrikopoulou A, Zagouri F, Goula K, Haidopoulos D, Thomakos N, Svarna A, Dimopoulos MA, Liontos M. Real-world evidence of Trastuzumab Deruxtecan (T-DXd) Efficacy in HER2-expressing gynecological malignancies. BMC Cancer 2024; 24:1503. [PMID: 39639215 PMCID: PMC11622448 DOI: 10.1186/s12885-024-13226-1] [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: 08/10/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) overexpression emerges as an attractive therapeutic target in gynecological malignancies. Recently, Trastuzumab-Deruxtecan (T-DXd) has shown substantial efficacy in HER2 overexpressing carcinomas, most prominently in ovarian, endometrial and cervical patients. We have examined the efficacy of T-DXd in patients with metastatic endometrial, ovarian and cervical malignancies that were treated in the Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens.MethodsPatients with gynecological malignancies with HER2 expression 2 + /3 + as assessed by immunohistochemistry (IHC) that were treated with T-DXd were retrospectively identified. Patients received T-DXd intravenously at a dose of 5.4 mg/kg every 3 weeks until disease progression or unacceptable toxicity.ResultsIn total, 10 patients treated with T-DXd for recurrent/metastatic disease were identified. Regarding histology, there were 5 patients with uterine neoplasms (serous carcinoma (n = 3), uterine carcinosarcoma (n = 1), uterine leiomyosarcoma (n = 1)), 1 patient with squamous cervical carcinoma and 4 patients with ovarian cancer (ovarian carcinosarcoma (n = 2), high-grade serous carcinoma (n = 1) and mucinous ovarian carcinoma (n = 1)). Median age was 65.4 years (25th-75th percentile, 58.1-75.2 years). 5 patients had HER2 expression 3 + by IHC and 5 patients had HER2 expression 2 + . Median number of previous lines of therapy was 4 (range 2-6), 2 patients with uterine serous carcinoma were pretreated with trastuzumab and 4 patients had already received immunotherapy. In the entire cohort median progression-free survival (PFS) was 5.4 months (95%CI 0.8-9.8 months). Regarding responses, 5 patients had partial response (including 2 patients that were pretreated with trastuzumab), 1 patient had stable disease at 12 weeks and 4 patients had disease progression at initial assessment. All patients but one that derived clinical benefit had HER2 3 + expression.DiscussionIn the real-world setting, T-DXd showed activity in a cohort of heavily pre-treated patients with HER2-expressing gynecological malignancies.
Collapse
Affiliation(s)
- Angeliki Andrikopoulou
- Department of Clinical Therapeutics, Medical School, Alexandra Hospital, 11528, Athens, Greece.
| | - Flora Zagouri
- Department of Clinical Therapeutics, Medical School, Alexandra Hospital, 11528, Athens, Greece
| | - Kallirroi Goula
- Department of Pathology, Alexandra General Hospital of Athens, Athens, Greece
| | - Dimitrios Haidopoulos
- Obstetrics and Gynecology, National and Kapodistrian University of Athens - 1, Obstetrics and Gynecology Clinic, Athens, Greece
| | - Nikolaos Thomakos
- Obstetrics and Gynecology, National and Kapodistrian University of Athens - 1, Obstetrics and Gynecology Clinic, Athens, Greece
| | - Anna Svarna
- Department of Clinical Therapeutics, Medical School, Alexandra Hospital, 11528, Athens, Greece
| | | | - Michalis Liontos
- Department of Clinical Therapeutics, Medical School, Alexandra Hospital, 11528, Athens, Greece
| |
Collapse
|
33
|
Simó C, Shmuel S, Vanover A, Pereira PMR. [ 64Cu]Cu-NOTA-Trastuzumab and [ 89Zr]Zr-DFO-Trastuzumab in Xenografts with Varied HER2 Expression. Mol Pharm 2024; 21:6311-6322. [PMID: 39471823 PMCID: PMC11611601 DOI: 10.1021/acs.molpharmaceut.4c00777] [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: 07/16/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 11/01/2024]
Abstract
Positron emission tomography (PET) has potential as a complementary technique to biomarker analysis, especially for human epidermal growth factor receptor 2 (HER2)-expressing tumors characterized by high heterogeneity. In this study, zirconium-89 (89Zr) and copper-64 (64Cu) labeled trastuzumab were employed to monitor varying levels of tumoral HER2 expression. Additionally, we studied the use of the cholesterol-depleting lovastatin as a pharmacological approach to enhance cell-surface HER2 expression in tumors with moderate to low HER2 levels, aiming to increase antibody accumulation in these tumor types. Both 89Zr- and 64Cu-labeled trastuzumab effectively monitor HER2 expression levels in xenografts exhibiting varying HER2 expression. No significant difference in tumor uptake was observed between 89Zr- or 64Cu-labeled trastuzumab, and tumor uptake for both radioimmunoconjugates positively correlated with HER2 protein levels. These findings underscore the potential of PET to monitor HER2 protein levels across heterogeneous tumors. Furthermore, our results suggest that further optimization of statin dosing and timing could offer a promising strategy to enhance trastuzumab accumulation in HER2-high, HER2-moderate, and HER2-low tumors.
Collapse
Affiliation(s)
- Cristina Simó
- Department of Radiology,
Mallinckrodt Institute of Radiology, Washington
University School of Medicine, St. Louis, Missouri 63110, United States
| | - Shayla Shmuel
- Department of Radiology,
Mallinckrodt Institute of Radiology, Washington
University School of Medicine, St. Louis, Missouri 63110, United States
| | - Alex Vanover
- Department of Radiology,
Mallinckrodt Institute of Radiology, Washington
University School of Medicine, St. Louis, Missouri 63110, United States
| | - Patrícia M. R. Pereira
- Department of Radiology,
Mallinckrodt Institute of Radiology, Washington
University School of Medicine, St. Louis, Missouri 63110, United States
| |
Collapse
|
34
|
Waliany S, Nagasaka M, Park L, Lam C, Jiang Z, Lin F, Neal JW. Real-World Prevalence, Treatment Patterns, and Outcomes for Patients With HER2 (ERBB2)-Mutant Metastatic Non-Small Cell Lung Cancer, From a US-Based Clinico-Genomic Database. Cancer Med 2024; 13:e70272. [PMID: 39692700 DOI: 10.1002/cam4.70272] [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: 05/16/2024] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 12/19/2024] Open
Abstract
OBJECTIVES Targeted therapies have been shown to improve outcomes in metastatic non-small cell lung cancer (mNSCLC) with driver mutations. We evaluated the real-world prevalence of human epidermal growth factor receptor 2 (HER2; ERBB2) tumor gene mutations among patients with mNSCLC and described historical treatments and outcomes in patients with HER2-mutant mNSCLC, during a period when there was no approved targeted therapy for HER2-mutant mNSCLC. MATERIALS AND METHODS This retrospective observational study used a US nationwide de-identified NSCLC clinico-genomic database. Eligible patients were adults diagnosed with HER2-mutant mNSCLC from January 2014 to July 2021 without co-occuring epidermal growth factor receptor (EGFR) tumor mutations. Descriptive statistics were used to summarize prevalence, baseline characteristics and treatment patterns. Clinical outcomes were estimated with Kaplan-Meier analyses. RESULTS Among 9206 patients with mNSCLC, 164 (1.78%) met the eligibility criteria (mean age: 67.3 years, 63.4% White, 56.7% female, and 53.0% with a smoking history). 132/164 (80.5%) had at least one line of treatment. Platinum-based chemotherapy (45.5%) and immune checkpoint inhibitor (ICI) with chemotherapy (28.0%) were the most frequently used first-line treatments. The median (95% confidence interval [CI]) real-world (rw) progression-free survival in first-line was 5.5 (4.8, 6.2) months and 3.0 (2.3, 4.2) months in second-line. The median rw overall survival in first-line was 13.2 (10.6, 18.4) months and 8.2 (6.6, 13.2) months in second-line. CONCLUSION During this study period, the most common regimens were platinum-based chemotherapy with or without ICI in first and second line, and median rwOS was 13.2 and 8.2 months, respectively. These results indicate the need for more effective targeted therapies in this patient population.
Collapse
Affiliation(s)
- Sarah Waliany
- Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
| | - Misako Nagasaka
- University of California Irvine School of Medicine, Orange, California, USA
| | - Leah Park
- AstraZeneca, Gaithersburg, Maryland, USA
| | - Clara Lam
- AstraZeneca, Gaithersburg, Maryland, USA
| | - Zoe Jiang
- AstraZeneca, Gaithersburg, Maryland, USA
| | - Feng Lin
- Daiichi Sankyo, Basking Ridge, New Jersey, USA
| | - Joel W Neal
- Department of Medicine, Stanford University, Stanford, California, USA
| |
Collapse
|
35
|
Wisniewski A, Humer D, Möller M, Kanje S, Spadiut O, Hober S. Targeted HER2-positive cancer therapy using ADAPT6 fused to horseradish peroxidase. N Biotechnol 2024; 83:74-81. [PMID: 39032630 DOI: 10.1016/j.nbt.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 07/05/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
Targeted cancer therapy is a promising alternative to the currently established cancer treatments, aiming to selectively kill cancer cells while sparing healthy tissues. Hereby, molecular targeting agents, such as monoclonal antibodies, are used to bind to cancer cell surface markers specifically. Although these agents have shown great clinical success, limitations still remain such as low tumor penetration and off-target effects. To overcome this limitation, novel fusion proteins comprised of the two proteins ADAPT6 and Horseradish Peroxidase (HRP) were engineered. Cancer cell targeting is hereby enabled by the small scaffold protein ADAPT6, engineered to specifically bind to human epidermal growth factor receptor 2 (HER2), a cell surface marker overexpressed in various cancer types, while the enzyme HRP oxidizes the nontoxic prodrug indole-3-acetic acid (IAA) which leads to the formation of free radicals and thereby to cytotoxic effects on cancer cells. The high affinity to HER2, as well as the enzymatic activity of HRP, were still present for the ADAPT6-HRP fusion proteins. Further, in vitro cytotoxicity assay using HER2-positive SKOV-3 cells revealed a clear advantage of the fusion proteins over free HRP by association of the fusion proteins directly to the cancer cells and therefore sustained cell killing. This novel strategy of combining ADAPT6 and HRP represents a promising approach and a viable alternative to antibody conjugation for targeted cancer therapy.
Collapse
Affiliation(s)
- Andreas Wisniewski
- Department of Protein Science, KTH-Royal Institute of Technology, SE-10691 Stockholm, Sweden
| | - Diana Humer
- Institute of Chemical, Environmental and Bioscience Engineering, Research Area Biochemical Engineering, AT-1060 Vienna, Austria
| | - Marit Möller
- Department of Protein Science, KTH-Royal Institute of Technology, SE-10691 Stockholm, Sweden
| | - Sara Kanje
- Department of Protein Science, KTH-Royal Institute of Technology, SE-10691 Stockholm, Sweden
| | - Oliver Spadiut
- Institute of Chemical, Environmental and Bioscience Engineering, Research Area Biochemical Engineering, AT-1060 Vienna, Austria
| | - Sophia Hober
- Department of Protein Science, KTH-Royal Institute of Technology, SE-10691 Stockholm, Sweden.
| |
Collapse
|
36
|
Yagisawa M, Taniguchi H, Satoh T, Kadowaki S, Sunakawa Y, Nishina T, Komatsu Y, Esaki T, Sakai D, Doi A, Kajiwara T, Ono H, Asano M, Hirano N, Odegaard J, Fujii S, Nomura S, Bando H, Sato A, Yoshino T, Nakamura Y. Trastuzumab Deruxtecan in Advanced Solid Tumors With Human Epidermal Growth Factor Receptor 2 Amplification Identified by Plasma Cell-Free DNA Testing: A Multicenter, Single-Arm, Phase II Basket Trial. J Clin Oncol 2024; 42:3817-3825. [PMID: 39088783 PMCID: PMC11542975 DOI: 10.1200/jco.23.02626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/01/2024] [Accepted: 05/10/2024] [Indexed: 08/03/2024] Open
Abstract
PURPOSE HERALD/EPOC1806 was conducted as a multicenter phase II trial assessing trastuzumab deruxtecan (T-DXd) therapy for patients with human epidermal growth factor receptor 2 (HER2)-amplified progressive stage solid tumors detected by cell-free DNA (cfDNA) testing. PATIENTS AND METHODS Patients exhibited advanced solid tumors with HER2 amplification that was identified via next-generation sequencing of cfDNA testing, without the requirement for immunohistochemical HER2 testing. The studied group was administered T-DXd at 5.4 mg/kg once every 3 weeks until onset of disease progression or intolerable toxicity. RESULTS Overall, 4,734 patients underwent cfDNA testing from December 2019 to January 2022, and 252 demonstrated HER2 amplification. Finally, the study included 62 patients with 16 cancer types with a median baseline plasma HER2 copy number (CN) of 8.55 (range, 2.4-73.9). Confirmed overall response rate (ORR) by investigator assessment was 56.5% (95% CI, 43.3 to 69.0), thus showing a value beyond the 5% threshold. Responses were evaluated for 13 cancer types, including KRAS-mutant colorectal (1/3), PIK3CA-mutant endometrial (5/6), and tissue HER2-negative gastric (1/2) cancers. Plasma HER2 CN above versus below the baseline median value did not differ for impact response; however, clearance of HER2 amplification in cfDNA on cycle 2 day 1 had higher response values compared with persistence. Median progression-free survival and response duration were 7.0 (95% CI, 4.9 to 9.7) and 8.8 (95% CI, 5.8 to 11.2) months, respectively, with the majority of complications being mild to moderate. Interstitial lung diseases were identified in 16 (26%) patients, including 14 patients with grade 1 disease, one patient with grade 2 disease, and one patient with grade 3 disease. CONCLUSION T-DXd treatment demonstrated high ORR with durable response in patients with advanced HER2-amplified solid tumors determined with cfDNA testing.
Collapse
Affiliation(s)
- Masataka Yagisawa
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology, Japan Community Health Care Organization Sapporo Hokushin Hospital, Sapporo, Japan
| | - Hiroya Taniguchi
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Taroh Satoh
- Center for Cancer Genomics and Precision Medicine, Osaka University Hospital, Osaka, Japan
| | - Shigenori Kadowaki
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Yu Sunakawa
- Department of Clinical Oncology, St Marianna University School of Medicine, Kawasaki, Japan
| | - Tomohiro Nishina
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Yoshito Komatsu
- Division of Cancer Center, Hokkaido University Hospital, Sapporo, Japan
| | - Taito Esaki
- Gastrointestinal and Medical Oncology Division, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Daisuke Sakai
- Center for Cancer Genomics and Precision Medicine, Osaka University Hospital, Osaka, Japan
| | - Ayako Doi
- Department of Clinical Oncology, St Marianna University School of Medicine, Kawasaki, Japan
| | - Takeshi Kajiwara
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Hiromi Ono
- Clinical Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masatoshi Asano
- Clinical Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Nami Hirano
- Clinical Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | | | - Satoshi Fujii
- Department of Molecular Pathology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shogo Nomura
- Department of Biostatistics and Bioinformatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hideaki Bando
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Translational Research Support Section, National Cancer Center Hospital East, Kashiwa, Japan
| | - Akihiro Sato
- Clinical Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshiaki Nakamura
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Translational Research Support Section, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
37
|
Goldberg SD, Satomaa T, Aina O, Aitio O, Burke K, Dudkin V, Geist B, Irrechukwu O, Hänninen AL, Heiskanen A, Helin J, Hiltunen JO, Kinyamu-Akunda J, Klein DM, Kohli N, Kotiranta T, Lähteenmäki T, Niemelä R, Pitkänen V, Pynnönen H, Rittase W, Wiley K, Zhou J, Saarinen J. Trastuzumab-MMAU Antibody-Auristatin Conjugates: Valine-Glucoserine Linker with Stabilized Maleimide Conjugation Improves In Vivo Efficacy and Tolerability. Mol Cancer Ther 2024; 23:1530-1543. [PMID: 38324296 DOI: 10.1158/1535-7163.mct-23-0591] [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: 09/06/2023] [Revised: 11/16/2023] [Accepted: 02/01/2024] [Indexed: 02/08/2024]
Abstract
Antibody-drug conjugates (ADC) have shown impressive clinical activity with approval of many agents in hematologic and solid tumors. However, challenges remain with both efficacy and safety of ADCs. This study describes novel trastuzumab-auristatin conjugates with the hydrophilic monomethylauristatin E (MMAE) prodrug MMAU, and optimization of a glycopeptide linker leading to a wider therapeutic window. Trastuzumab was conjugated with auristatin payloads via a series of linkers using a stabilized maleimide handle. The ADCs were characterized in vitro and their relative in vivo antitumor efficacies were assessed in HER2+ xenograft models. Relative linker stabilities and the mechanism of linker cleavage were studied using in vitro assays. Toxicity and toxicokinetics of the best performing ADC were evaluated in cynomolgus monkey (cyno). The trastuzumab-MMAU ADC with stabilized glycopeptide linker showed maleimide stabilization and higher resistance to cleavage by serum and lysosomal enzymes compared with a valine-citrulline conjugated trastuzumab ADC (trastuzumab-vc-MMAE). A single dose of 1 or 2 mg/kg of trastuzumab-MMAU at drug-to-antibody ratios (DAR) of eight and four respectively resulted in xenograft tumor growth inhibition, with superior efficacy to trastuzumab-vc-MMAE. Trastuzumab-MMAUDAR4 was tolerated at doses up to 12 mg/kg in cyno, which represents 2- to 4-fold higher dose than that observed with vedotin ADCs, and had increased terminal half-life and exposure. The optimized trastuzumab-MMAU ADC showed potent antitumor activity and was well tolerated with excellent pharmacokinetics in nonhuman primates, leading to a superior preclinical therapeutic window. The data support potential utility of trastuzumab-MMAU for treatment of HER2+ tumors.
Collapse
Affiliation(s)
| | | | - Olulanu Aina
- Janssen Pharmaceuticals, Spring House, Pennsylvania
| | | | - Krista Burke
- Janssen Pharmaceuticals, Spring House, Pennsylvania
| | - Vadim Dudkin
- Janssen Pharmaceuticals, Spring House, Pennsylvania
| | - Brian Geist
- Janssen Pharmaceuticals, Spring House, Pennsylvania
| | | | | | | | | | | | | | | | - Neeraj Kohli
- Janssen Pharmaceuticals, Spring House, Pennsylvania
| | | | | | | | | | | | | | | | - Junguo Zhou
- Janssen Pharmaceuticals, Spring House, Pennsylvania
| | | |
Collapse
|
38
|
Liu Y, Oroujeni M, Liao Y, Vorobyeva A, Bodenko V, Orlova A, Konijnenberg M, Carlqvist M, Wahlberg E, Loftenius A, Frejd FY, Tolmachev V. Evaluation of a novel 177Lu-labelled therapeutic Affibody molecule with a deimmunized ABD domain and improved biodistribution profile. Eur J Nucl Med Mol Imaging 2024; 51:4038-4048. [PMID: 39008065 PMCID: PMC11527907 DOI: 10.1007/s00259-024-06840-5] [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: 01/25/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024]
Abstract
PURPOSE Fusion of Affibody molecules with an albumin-binding domain (ABD) provides targeting agents, which are suitable for radionuclide therapy. To facilitate clinical translation, the low immunogenic potential of such constructs with targeting properties conserved is required. METHODS The HER2-targeting Affibody molecule ZHER2:2891 was fused with a deimmunized ABD variant and DOTA was conjugated to a unique C-terminal cysteine. The novel construct, PEP49989, was labelled with 177Lu. Affinity, specificity, and in vivo targeting properties of [177Lu]Lu-PEP49989 were characterised. Experimental therapy in mice with human HER2-expressing xenografts was evaluated. RESULTS The maximum molar activity of 52 GBq/µmol [177Lu]Lu-PEP49989 was obtained. [177Lu]Lu-PEP49989 bound specifically to HER2-expressing cells in vitro and in vivo. The HER2 binding affinity of [177Lu]Lu-PEP49989 was similar to the affinity of [177Lu]Lu-ABY-027 containing the parental ABD035 variant. The renal uptake of [177Lu]Lu-PEP49989 was 1.4-fold higher, but hepatic and splenic uptake was 1.7-2-fold lower than the uptake of [177Lu]Lu-ABY-027. The median survival of xenograft-bearing mice treated with 21 MBq [177Lu]Lu-PEP49989 (> 90 days) was significantly longer than the survival of mice treated with vehicle (38 days) or trastuzumab (45 days). Treatment using a combination of [177Lu]Lu-PEP49989 and trastuzumab increased the number of complete tumour remissions. The renal and hepatic toxicity was minimal to mild. CONCLUSION In preclinical studies, [177Lu]Lu-PEP49989 demonstrated favourable biodistribution and a strong antitumour effect, which was further enhanced by co-treatment with trastuzumab.
Collapse
Affiliation(s)
- Yongsheng Liu
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, 751 85, Sweden
| | - Maryam Oroujeni
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, 751 85, Sweden
- Affibody AB, Solna, 171 65, Sweden
| | - Yunqi Liao
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, 751 85, Sweden
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, 751 85, Sweden
| | - Vitalina Bodenko
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, 751 85, Sweden
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, Uppsala, 751 23, Sweden
| | - Mark Konijnenberg
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | | | | | | | - Fredrik Y Frejd
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, 751 85, Sweden
- Affibody AB, Solna, 171 65, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, 751 85, Sweden.
| |
Collapse
|
39
|
Nguyen K, Fama K, Mercado G, Myat Y, Thein K. Histology Agnostic Drug Development: An Updated Review. Cancers (Basel) 2024; 16:3642. [PMID: 39518080 PMCID: PMC11544807 DOI: 10.3390/cancers16213642] [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: 09/07/2024] [Revised: 10/23/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
Recent advancements in oncology have led to the development of histology-agnostic therapies, which target genetic alterations irrespective of the tumor's tissue of origin. This review aimed to provide a comprehensive update on the current state of histology-agnostic drug development, focusing on key therapies, including pembrolizumab, larotrectinib, entrectinib, dostarlimab, dabrafenib plus trametinib, selpercatinib, trastuzumab deruxtecan, and reprotrectinib. We performed a detailed analysis of each therapy's mechanism of action, clinical trial outcomes, and associated biomarkers. The review further explores challenges in drug resistance, such as adaptive signaling pathways and neoantigen variability, as well as diagnostic limitations in identifying optimal patient populations. While these therapies have demonstrated efficacy in various malignancies, significant hurdles remain, including intratumoral heterogeneity and resistance mechanisms that diminish treatment effectiveness. We propose considerations for refining trial designs and emerging biomarkers, such as tumor neoantigen burden, to enhance patient selection. These findings illustrate the transformative potential of histology-agnostic therapies in precision oncology but highlight the need for continued research to optimize their use and overcome existing barriers.
Collapse
Affiliation(s)
- Kevin Nguyen
- Touro University Nevada College of Osteopathic Medicine, 874 American Pacific Dr, Henderson, NV 89014, USA; (K.N.)
| | - Karina Fama
- Touro University Nevada College of Osteopathic Medicine, 874 American Pacific Dr, Henderson, NV 89014, USA; (K.N.)
| | - Guadalupe Mercado
- Touro University Nevada College of Osteopathic Medicine, 874 American Pacific Dr, Henderson, NV 89014, USA; (K.N.)
| | - Yin Myat
- University College Dublin School of Medicine, Belfield, D04 V1W8 Dublin, Ireland
- One Brooklyn Health—Interfaith Medical Center Campus, 1545, Atlantic Avenue, Brooklyn, NY 11213, USA
| | - Kyaw Thein
- Touro University Nevada College of Osteopathic Medicine, 874 American Pacific Dr, Henderson, NV 89014, USA; (K.N.)
- Comprehensive Cancer Centers of Nevada—Central Valley, 3730 S Eastern Ave, Las Vegas, NV 89169, USA
- Kirk Kerkorian School of Medicine, University of Nevada, Las Vegas (UNLV), 4505 S. Maryland Pkwy, Las Vegas, NV 89154, USA
| |
Collapse
|
40
|
Lazar V, Raymond E, Magidi S, Bresson C, Wunder F, Berindan-Neagoe I, Tijeras-Rabaland A, Raynaud J, Onn A, Ducreux M, Batist G, Lassen U, Cilius Nielsen F, Schilsky RL, Rubin E, Kurzrock R. Identification of a central network hub of key prognostic genes based on correlation between transcriptomics and survival in patients with metastatic solid tumors. Ther Adv Med Oncol 2024; 16:17588359241289200. [PMID: 39429467 PMCID: PMC11487509 DOI: 10.1177/17588359241289200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 09/18/2024] [Indexed: 10/22/2024] Open
Abstract
Background Dysregulated pathways in cancer may be hub addicted. Identifying these dysregulated networks for targeting might lead to novel therapeutic options. Objective Considering the hypothesis that central hubs are associated with increased lethality, identifying key hub targets within central networks could lead to the development of novel drugs with improved efficacy in advanced metastatic solid tumors. Design Exploring transcriptomic data (22,000 gene products) from the WINTHER trial (N = 101 patients with various metastatic cancers), in which both tumor and normal organ-matched tissue were available. Methods A retrospective in silico analysis of all genes in the transcriptome was conducted to identify genes different in expression between tumor and normal tissues (paired t-test) and to determine their association with survival outcomes using survival analysis (Cox proportional hazard regression algorithm). Based on the biological relevance of the identified genes, hub targets of interest within central networks were then pinpointed. Patients were grouped based on the expression level of these genes (K-mean clustering), and the association of these groups with survival was examined (Cox proportional hazard regression algorithm, Forest plot, and Kaplan-Meier plot). Results We identified four key central hub genes-PLOD3, ARHGAP11A, RNF216, and CDCA8, for which high expression in tumor tissue compared to analogous normal tissue had the most significant correlation with worse outcomes. The correlation was independent of tumor or treatment type. The combination of the four genes showed the highest significance and correlation with the poorer outcome: overall survival (hazard ratio (95% confidence interval (CI)) = 10.5 (3.43-31.9) p = 9.12E-07 log-rank test in a Cox proportional hazard regression model). Findings were validated in independent cohorts. Conclusion The expression of PLOD3, ARHGAP11A, RNF216, and CDCA8 constitute, when combined, a prognostic tool, agnostic of tumor type and previous treatments. These genes represent potential targets for intercepting central hub networks in various cancers, offering avenues for novel therapeutic interventions.
Collapse
Affiliation(s)
- Vladimir Lazar
- Worldwide Innovative Network Association—WIN Consortium, Villejuif, France
| | - Eric Raymond
- Groupe Hospitalier Saint Joseph, Oncology Department Paris, France
| | - Shai Magidi
- Worldwide Innovative Network Association—WIN Consortium, 24, rue Albert Thuret, Chevilly-Larue 94850, France
| | - Catherine Bresson
- Worldwide Innovative Network Association—WIN Consortium, Villejuif, France
| | - Fanny Wunder
- Worldwide Innovative Network Association—WIN Consortium, Villejuif, France
| | - Ioana Berindan-Neagoe
- The Oncology Institute “Prof. Dr. Ion Chiricuta,” Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | | | - Jacques Raynaud
- Worldwide Innovative Network Association—WIN Consortium, Villejuif, France
| | - Amir Onn
- Sheba Medical Center, Institute of Pulmonology, Tel HaShomer, Ramat-Gan, Israel
| | - Michel Ducreux
- Gustave Roussy, Department of Medical Oncology, Villejuif, France
- University Paris-Saclay, Department of Medical Oncology, Orsay, France
| | - Gerald Batist
- Segal Cancer Centre, Department of Oncology, Jewish General Hospital, McGill University, Montréal, QC, Canada
| | | | | | | | - Eitan Rubin
- Ben-Gurion University of the Negev, The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, Be’er-Sheva, Israel
| | | |
Collapse
|
41
|
Zhang T, Febres-Aldana CA, Liu Z, Dix JM, Cheng R, Dematteo RG, Lui AJW, Khodos I, Gili L, Mattar MS, Lisanti J, Kwong C, Linkov I, Tipping MJ, de Stanchina E, Odintsov I, Ladanyi M, Somwar R. HER2 Antibody-Drug Conjugates Are Active against Desmoplastic Small Round Cell Tumor. Clin Cancer Res 2024; 30:4701-4713. [PMID: 39120576 PMCID: PMC11479846 DOI: 10.1158/1078-0432.ccr-24-1835] [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: 06/11/2024] [Revised: 07/16/2024] [Accepted: 08/07/2024] [Indexed: 08/10/2024]
Abstract
PURPOSE Desmoplastic small round cell tumor (DSRCT) is a rare but highly aggressive soft tissue sarcoma that arises in the abdominopelvic cavity of young males. Since the discovery of EWSR1::WT1 fusion as the driver of DSRCT, no actionable genomic alterations have been identified, limiting disease management to a combination of surgery, chemotherapy, and radiation, with very poor outcomes. Herein, we evaluated ERBB2/HER2 expression in DSRCT as a therapeutic target. EXPERIMENTAL DESIGN ERBB2/HER2 expression was assessed in clinical samples and patient-derived xenografts (PDX) using RNA sequencing, RT-qPCR, and a newly developed HER2 IHC assay (clone 29D8). Responses to HER2 antibody-drug conjugates (ADC)-trastuzumab deruxtecan (T-DXd) and trastuzumab emtansine-were evaluated in DSRCT PDX, cell line, and organoid models. Drug internalization was demonstrated by live microscopy. Apoptosis was evaluated by Western blotting and caspase activity assays. RESULTS ERBB2/HER2 was detectable in DSRCT samples from patients and PDXs, with higher sensitivity RNA assays and improved IHC detectability using clone 29D8. Treatment of ERBB2/HER2-expressing DSRCT PDX, cell line, and organoid models with T-DXd or trastuzumab emtansine resulted in tumor regression. This therapeutic response was long-lasting in T-DXd-treated xenografts and was mediated by rapid HER2 ADC complex internalization and cytotoxicity, triggering p53-mediated apoptosis and growth arrest. Xenograft regression was associated with bystander payload effects triggering global tumor niche responses proportional to HER2 status. CONCLUSIONS ERBB2/HER2 is a therapeutic target in DSRCT. HER2 ADCs may represent novel options for managing this exceptionally aggressive sarcoma, possibly fulfilling an urgent and historically unmet need for more effective clinical therapy.
Collapse
Affiliation(s)
- Tom Zhang
- New York Medical College, Valhalla, NY, 10595, USA
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Christopher A. Febres-Aldana
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Zebing Liu
- Department of Pathology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Jenna-Marie Dix
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Ryan Cheng
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Raymond G. Dematteo
- Department of Pharmacy, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Allan JW Lui
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Cancer Research UK, University of Cambridge, Cambridge, CB2 0RE, UK
| | - Inna Khodos
- Antitumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Leo Gili
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Marissa S. Mattar
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Jeanine Lisanti
- Antitumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Charlene Kwong
- Antitumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Irina Linkov
- Pathology Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Murray J. Tipping
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Igor Odintsov
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 021105, USA
| | - Marc Ladanyi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Romel Somwar
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| |
Collapse
|
42
|
Zalaquett Z, Rita Hachem MC, Assi A, Mohanna R, Farhat M, Noujaim C, Kourie HR. Cardiac toxicity of HER-2 targeting antibody-drug conjugates: overview and clinical implications. Future Oncol 2024; 20:3151-3167. [PMID: 39373602 DOI: 10.1080/14796694.2024.2407756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 09/19/2024] [Indexed: 10/08/2024] Open
Abstract
Antibody-drug conjugates (ADCs) have recently emerged as a promising therapeutic option that combine the specificity of monoclonal antibodies and the cytotoxic effect of chemotherapy. With numerous ADCs approved and on the market, a particular concern of ADCs that target HER-2 has been their cardiac side effects, in view of the crucial role of HER-2 in cardiac development and physiology. While rarely toxic and generally safe, numerous publications have outlined the consistent association of trastuzumab emtansine (T-DM1) and trastuzumab deruxtecan (T-DXd) with the development of cardiac toxicity. Despite not being clinically relevant in most cases, cardiac baseline evaluation, monitoring and early detection of cardiac adverse events remain pivotal with HER-2 targeting ADCs. This review aims to summarize and better characterize the complete cardiac toxicity profile of HER-2 ADCs, with the goal of improving clinical understanding of this adverse event, leading to better recognition, monitoring and management.
Collapse
Affiliation(s)
- Ziad Zalaquett
- Hôtel-Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon
| | | | - Ahmad Assi
- Hôtel-Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Rami Mohanna
- Hôtel-Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Mohamad Farhat
- Hôtel-Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon
| | | | - Hampig-Raphael Kourie
- Hôtel-Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon
| |
Collapse
|
43
|
Cheng X, Li P, Jiang R, Meng E, Wu H. ADC: a deadly killer of platinum resistant ovarian cancer. J Ovarian Res 2024; 17:196. [PMID: 39367438 PMCID: PMC11451100 DOI: 10.1186/s13048-024-01523-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: 04/18/2024] [Accepted: 09/25/2024] [Indexed: 10/06/2024] Open
Abstract
Platinum is a key component of ovarian cancer systemic therapy. However, most patients will eventually face a recurrence, leading to chemotherapy resistance, especially against platinum. For individuals with platinum-resistant ovarian cancer (PROC), treatment options are limited, and their survival prospects are grim. The emergence of antibody-drug conjugates (ADCs) shows promises as a future treatment for PROC. This review synthesizes current research on the effectiveness of ADCs in treating PROC. It encapsulates the advancements and clinical trials of novel ADCs that target specific antigens such as Folate Receptor alpha (FRα), MUC16, NaPi2b, Mesothelin, Dipeptidase 3(DPEP3), and human epidermal growth factor receptor 2 (HER2), as well as tissue factor, highlighting their potential anti-tumor efficacy and used in combination with other therapies. The ADCs landscape in ovarian cancer therapeutics is swiftly evolving, promising more potent and efficacious treatment avenues.
Collapse
Affiliation(s)
- Xu Cheng
- The Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing City, Jiangsu Province, China
| | - Ping Li
- The Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing City, Jiangsu Province, China
| | - Rongqi Jiang
- The Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing City, Jiangsu Province, China
| | - Enqing Meng
- The Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing City, Jiangsu Province, China
| | - Hao Wu
- The Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing City, Jiangsu Province, China.
| |
Collapse
|
44
|
Bhagyalalitha M, Handattu Shankaranarayana A, Arun Kumar S, Singh M, Pujar KG, Bidye D, Veeranna Pujar G. Advances in HER2-Targeted Therapies: From monoclonal antibodies to dual inhibitors developments in cancer treatment. Bioorg Chem 2024; 151:107695. [PMID: 39137598 DOI: 10.1016/j.bioorg.2024.107695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 07/28/2024] [Accepted: 08/04/2024] [Indexed: 08/15/2024]
Abstract
HER2 receptors, overexpressed in certain human cancers, have drawn significant attention in cancer research due to their correlation with poor survival rates. Researchers have developed monoclonal antibodies like Trastuzumab and Pertuzumab against HER2 receptors, which have proven highly beneficial in cancer therapy. Bispecific antibodies like Zanidatamab and antibody-drug conjugates like T-DM1 have been developed to overcome the resistance associated with monotherapy. Small molecules such as Lapatinib, Neratinib, and Pyrotinib were initially developed for treating breast cancer. However, ongoing research is investigating their potential use in other types of cancer, often in combination with other medications. EGFR/HER2 dual-targeted drugs have overcome drug resistance associated with HER2-targeted monotherapy. This comprehensive review covers the structural characteristics of HER2, the HER family signaling pathway mechanism, recent findings regarding HER2 receptor involvement in various cancers, and diverse HER2-targeted therapies. This information provides a comprehensive understanding of HER2-targeted strategies in the evolving field of cancer treatment.
Collapse
Affiliation(s)
- Meduri Bhagyalalitha
- Computer Aided Drug Design Laboratory, Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Sri Shivarathreeshwara Nagara, Mysuru 570015 India
| | - Akshatha Handattu Shankaranarayana
- Computer Aided Drug Design Laboratory, Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Sri Shivarathreeshwara Nagara, Mysuru 570015 India
| | - Sethu Arun Kumar
- Computer Aided Drug Design Laboratory, Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Sri Shivarathreeshwara Nagara, Mysuru 570015 India
| | - Manisha Singh
- Computer Aided Drug Design Laboratory, Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Sri Shivarathreeshwara Nagara, Mysuru 570015 India
| | - Karthik G Pujar
- Computer Aided Drug Design Laboratory, Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Sri Shivarathreeshwara Nagara, Mysuru 570015 India
| | - Durgesh Bidye
- Computer Aided Drug Design Laboratory, Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Sri Shivarathreeshwara Nagara, Mysuru 570015 India
| | - Gurubasavaraj Veeranna Pujar
- Computer Aided Drug Design Laboratory, Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Sri Shivarathreeshwara Nagara, Mysuru 570015 India.
| |
Collapse
|
45
|
Quaquarini E, Grillo F, Gervaso L, Arpa G, Fazio N, Vanoli A, Parente P. Prognostic and Predictive Roles of HER2 Status in Non-Breast and Non-Gastroesophageal Carcinomas. Cancers (Basel) 2024; 16:3145. [PMID: 39335117 PMCID: PMC11430748 DOI: 10.3390/cancers16183145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/01/2024] [Accepted: 09/07/2024] [Indexed: 09/30/2024] Open
Abstract
The oncogene ERBB2, also known as HER2 or c-ERB2, is located on chromosome 17 (q12). It encodes a tyrosine kinase receptor, the human epidermal growth factor receptor 2 (HER2), involved in neoplastic proliferation, tumor angiogenesis, and invasiveness. Over the past years, the introduction of various anti-HER2 therapies has significantly improved outcomes for patients with HER2-positive breast and gastroesophageal carcinomas. More recently, the introduction of a new antibody-drug conjugate, that is trastuzumab deruxtecan, expanded the therapeutic options to low-HER2 breast and gastroesophageal tumors. HER2 protein overexpression is investigated using immunohistochemistry, gene amplification using fluorescence in situ hybridization, and gene mutation using next-generation sequencing. This review evaluated the predictive and prognostic role of HER2 status in various types of epithelial malignant cancers beyond breast and gastroesophageal cancers. We critically analyzed the key published studies, focusing on utilized scoring systems and assays used, and analyzed clinical parameters and therapeutic approaches. Although the evidence about prognostic and predictive roles of HER2 in carcinomas other than breast and gastroesophageal has been widely increasing over the last decade, it still remains investigational, revealing a tumor site-related prognostic and predictive value of the different types of HER2 alterations. However, standardized and validated scoring system assays have not been well-established for many organs.
Collapse
Affiliation(s)
- Erica Quaquarini
- Medical Oncology Unit of Pavia Institute, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy;
| | - Federica Grillo
- Anatomic Pathology Unit, University of Genova and Policlinico San Martino Hospital, 16132 Genova, Italy;
| | - Lorenzo Gervaso
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IRCCS, 20141 Milan, Italy; (L.G.); (N.F.)
| | - Giovanni Arpa
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy;
- Anatomic Pathology Unit of Pavia Institute, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy
| | - Nicola Fazio
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IRCCS, 20141 Milan, Italy; (L.G.); (N.F.)
| | - Alessandro Vanoli
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy;
- Anatomic Pathology Unit, Fondazione IRCCS San Matteo Hospital, 27100 Pavia, Italy
| | - Paola Parente
- Pathology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy;
| |
Collapse
|
46
|
Panaampon J, Sungwan P, Fujikawa S, Sampattavanich S, Jirawatnotai S, Okada S. Trastuzumab, a monoclonal anti-HER2 antibody modulates cytotoxicity against cholangiocarcinoma via multiple mechanisms. Int Immunopharmacol 2024; 138:112612. [PMID: 38968862 DOI: 10.1016/j.intimp.2024.112612] [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: 04/30/2024] [Revised: 06/16/2024] [Accepted: 06/30/2024] [Indexed: 07/07/2024]
Abstract
Cholangiocarcinoma (CCA) is an aggressive and fatal cancer. The prognosis is very poor and no optimal chemotherapy has been established. Human epidermal growth factor receptor 2 (HER2, neu, and erbB2) is highly-expressed in breast cancer and is expressed in many other tumors but poorly expressed in CCA. The anti-HER2 antibody, trastuzumab, has been used for the treatment of HER2-positive breast and gastric cancer. In this study, we examined the surface expression of HER2 on seven Thai liver-fluke-associated CCA cell lines by flow cytometry, and found all of these CCA cells were weakly positive for HER2. MTT assay revealed that trastuzumab directly suppressed the growth of CCA. By using FcR-bearing recombinant Jurkat T-cell-expressing firefly luciferase gene under the control of NFAT response elements, we defined the activities of antibody-dependent cytotoxicity (ADCC) and antibody-dependent cell phagocytosis (ADCP). ADCC was confirmed by using expanded NK cells. ADCP was confirmed by using mouse peritoneal macrophages and human monocyte-derived macrophages as effector cells. Rabbit serum was administered to test the complement-dependent cytotoxicity (CDC) activity of trastuzumab. Finally, we evaluated the efficacy of trastuzumab in in vivo patient-derived cell xenograft and patient-derived xenograft (PDX) models. Our results showed that a distinct population of CCA (liver-fluke-associated CCA) expressed HER2. Trastuzumab demonstrated a potent inhibitory effect on even HER2 weakly positive CCA both in vitro and in vivo via multiple mechanisms. Thus, HER2 is a promising target in anti-CCA therapy, and trastuzumab can be considered a promising antibody immunotherapy agent for the treatment of CCA.
Collapse
Affiliation(s)
- Jutatip Panaampon
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan; Division of Hematologic Neoplasia, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Prin Sungwan
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Sawako Fujikawa
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Somponnat Sampattavanich
- Siriraj Center of Research Excellence for Precision Medicine and Systems Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Siwanon Jirawatnotai
- Siriraj Center of Research Excellence for Precision Medicine and Systems Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan; Institute of Industrial Nanomaterials, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto, 860-8555, Japan.
| |
Collapse
|
47
|
Cho D, Lord SJ, Ward R, IJzerman M, Mitchell A, Thomas DM, Cheyne S, Martin A, Morton RL, Simes J, Lee CK. Criteria for assessing evidence for biomarker-targeted therapies in rare cancers-an extrapolation framework. Ther Adv Med Oncol 2024; 16:17588359241273062. [PMID: 39229469 PMCID: PMC11369883 DOI: 10.1177/17588359241273062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 07/09/2024] [Indexed: 09/05/2024] Open
Abstract
Background Advances in targeted therapy development and tumor sequencing technology are reclassifying cancers into smaller biomarker-defined diseases. Randomized controlled trials (RCTs) are often impractical in rare diseases, leading to calls for single-arm studies to be sufficient to inform clinical practice based on a strong biological rationale. However, without RCTs, favorable outcomes are often attributed to therapy but may be due to a more indolent disease course or other biases. When the clinical benefit of targeted therapy in a common cancer is established in RCTs, this benefit may extend to rarer cancers sharing the same biomarker. However, careful consideration of the appropriateness of extending the existing trial evidence beyond specific cancer types is required. A framework for extrapolating evidence for biomarker-targeted therapies to rare cancers is needed to support transparent decision-making. Objectives To construct a framework outlining the breadth of criteria essential for extrapolating evidence for a biomarker-targeted therapy generated from RCTs in common cancers to different rare cancers sharing the same biomarker. Design A series of questions articulating essential criteria for extrapolation. Methods The framework was developed from the core topics for extrapolation identified from a previous scoping review of methodological guidance. Principles for extrapolation outlined in guidance documents from the European Medicines Agency, the US Food and Drug Administration, and Australia's Medical Services Advisory Committee were incorporated. Results We propose a framework for assessing key assumptions of similarity of the disease and treatment outcomes between the common and rare cancer for five essential components: prognosis of the biomarker-defined cancer, biomarker test analytical validity, biomarker actionability, treatment efficacy, and safety. Knowledge gaps identified can be used to prioritize future studies. Conclusion This framework will allow systematic assessment, standardize regulatory, reimbursement and clinical decision-making, and facilitate transparent discussions between key stakeholders in drug assessment for rare biomarker-defined cancers.
Collapse
Affiliation(s)
- Doah Cho
- National Health and Medical Research Council Clinical Trials Centre, Faculty of Medicine and Health, University of Sydney, Australia
- Faculty of Medicine and Health, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Locked Bag 77, Camperdown, NSW 1450, Australia
| | - Sarah J. Lord
- Faculty of Medicine and Health, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Robyn Ward
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Maarten IJzerman
- Faculty of Medicine, Dentistry and Health Sciences, Centre for Health Policy, University of Melbourne Centre for Cancer Research, Parkville, VIC, Australia
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Andrew Mitchell
- Department of Health Economics Wellbeing and Society, The Australian National University, Canberra, ACT, Australia
| | - David M. Thomas
- Centre for Molecular Oncology, University of New South Wales, Sydney, NSW, Australia
| | - Saskia Cheyne
- Faculty of Medicine and Health, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Andrew Martin
- Faculty of Medicine and Health, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
- Centre for Clinical Research, University of Queensland, St Lucia, QLD, Australia
| | - Rachael L. Morton
- Faculty of Medicine and Health, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - John Simes
- Faculty of Medicine and Health, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Chee Khoon Lee
- Faculty of Medicine and Health, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
48
|
Shalaby N, Xia Y, Kelly JJ, Sanchez-Pupo R, Martinez F, Fox MS, Thiessen JD, Hicks JW, Scholl TJ, Ronald JA. Imaging CAR-NK cells targeted to HER2 ovarian cancer with human sodium-iodide symporter-based positron emission tomography. Eur J Nucl Med Mol Imaging 2024; 51:3176-3190. [PMID: 38722382 PMCID: PMC11368970 DOI: 10.1007/s00259-024-06722-w] [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: 02/20/2024] [Accepted: 04/14/2024] [Indexed: 09/03/2024]
Abstract
Chimeric antigen receptor (CAR) cell therapies utilize CARs to redirect immune cells towards cancer cells expressing specific antigens like human epidermal growth factor receptor 2 (HER2). Despite their potential, CAR T cell therapies exhibit variable response rates and adverse effects in some patients. Non-invasive molecular imaging can aid in predicting patient outcomes by tracking infused cells post-administration. CAR-T cells are typically autologous, increasing manufacturing complexity and costs. An alternative approach involves developing CAR natural killer (CAR-NK) cells as an off-the-shelf allogeneic product. In this study, we engineered HER2-targeted CAR-NK cells co-expressing the positron emission tomography (PET) reporter gene human sodium-iodide symporter (NIS) and assessed their therapeutic efficacy and PET imaging capability in a HER2 ovarian cancer mouse model.NK-92 cells were genetically modified to express a HER2-targeted CAR, the bioluminescence imaging reporter Antares, and NIS. HER2-expressing ovarian cancer cells were engineered to express the bioluminescence reporter Firefly luciferase (Fluc). Co-culture experiments demonstrated significantly enhanced cytotoxicity of CAR-NK cells compared to naive NK cells. In vivo studies involving mice with Fluc-expressing tumors revealed that those treated with CAR-NK cells exhibited reduced tumor burden and prolonged survival compared to controls. Longitudinal bioluminescence imaging demonstrated stable signals from CAR-NK cells over time. PET imaging using the NIS-targeted tracer 18F-tetrafluoroborate ([18F]TFB) showed significantly higher PET signals in mice treated with NIS-expressing CAR-NK cells.Overall, our study showcases the therapeutic potential of HER2-targeted CAR-NK cells in an aggressive ovarian cancer model and underscores the feasibility of using human-derived PET reporter gene imaging to monitor these cells non-invasively in patients.
Collapse
Affiliation(s)
- Nourhan Shalaby
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.
| | - Ying Xia
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - John J Kelly
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Rafael Sanchez-Pupo
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Francisco Martinez
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Matthew S Fox
- Lawson Health Research Institute, London, ON, Canada
- Saint Joseph's Health Care, London, ON, Canada
| | - Jonathan D Thiessen
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Lawson Cyclotron and Radiochemistry Facility, London, ON, Canada
- Saint Joseph's Health Care, London, ON, Canada
| | - Justin W Hicks
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Lawson Health Research Institute, London, ON, Canada
- Lawson Cyclotron and Radiochemistry Facility, London, ON, Canada
| | - Timothy J Scholl
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Ontario Institute for Cancer Research, London, ON, Canada
| | - John A Ronald
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Lawson Health Research Institute, London, ON, Canada
| |
Collapse
|
49
|
Verma S, Chapman A, Pickard LA, Porplycia D, McConkey H, Jarosz P, Sinfield J, Lauzon-Young C, Cecchini MJ, Howlett C, Grindrod N, Sadikovic B, Welch SA, Breadner D. Evaluation of HER2 immunohistochemistry expression in non-standard solid tumors from a Single-Institution Prospective Cohort. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:1100-1109. [PMID: 39351438 PMCID: PMC11438559 DOI: 10.37349/etat.2024.00265] [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: 03/14/2024] [Accepted: 07/29/2024] [Indexed: 10/04/2024] Open
Abstract
Aim Human epidermal growth factor receptor-2 (HER2) is a well-established prognostic and predictive biomarker. It is an FDA-approved therapeutic target for HER2 positive breast, gastroesophageal, and more recently, lung and colon cancers. It is an emerging biomarker in biliary tract, bladder, cervical, endometrial, ovarian, and pancreatic cancers. The emergence of new indications warrants further characterization of HER2 expression in diverse cancer populations. This study investigated HER2 expression in solid tumour samples and the feasibility of obtaining these results. Methods Prospective consent was obtained at a Canadian tertiary academic cancer center from adult oncology patients who were referred for molecular genetic testing of malignant tissue samples. Standard HER2-targeted malignancies were considered breast and gastroesophageal, and were excluded from this study. Between July 2020 and November 2023, 499 samples of solid tumors underwent immunohistochemistry (IHC) HER2 staining. A median turnaround time (TAT) of 14 days would be considered feasible for clinical decision making. Results The mean age (± SD) of participants was 67 ± 12.5 years, with 270 (54%) male and 229 (46%) female. HER2 protein expression was measured in 42 unique cancer types. IHC levels of 0, 1+, 2+, and 3+ were reported and were 43%, 12%, 35%, and 10% of all analyzable samples respectively (tissue inadequate in 3% of samples). The median TAT for HER2 expression results from time of request to result in release was 18 (interquartile range, 11 to 30) days. Conclusions HER2 protein expression varies widely between different cancer types. TAT for HER2 IHC results was a median of 18 days, which is close to our feasibility cut-off.
Collapse
Affiliation(s)
- Saurav Verma
- Division of Medical Oncology, Department of Oncology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
- Verspeeten Family Cancer Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Amanda Chapman
- Department of Medicine, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Lee-Anne Pickard
- Verspeeten Family Cancer Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Danielle Porplycia
- Verspeeten Family Cancer Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Haley McConkey
- Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON N6A 5W9, Canada
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Patricia Jarosz
- Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - James Sinfield
- Verspeeten Family Cancer Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Carolyn Lauzon-Young
- Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON N6A 5W9, Canada
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Matthew J Cecchini
- Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Christopher Howlett
- Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Natalie Grindrod
- Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Bekim Sadikovic
- Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON N6A 5W9, Canada
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Stephen A Welch
- Division of Medical Oncology, Department of Oncology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
- Verspeeten Family Cancer Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Daniel Breadner
- Division of Medical Oncology, Department of Oncology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
- Verspeeten Family Cancer Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada
| |
Collapse
|
50
|
Verma P, Gupta P, Gupta N, Srinivasan R, Gupta P, Dutta U, Sharma S, Uppal R, Nada R, Lal A. HER2/ERBB2 overexpression in advanced gallbladder carcinoma: comprehensive evaluation by immunocytochemistry and fluorescence in situ hybridisation on fine-needle aspiration cytology samples. J Clin Pathol 2024; 77:614-621. [PMID: 37221046 DOI: 10.1136/jcp-2023-208940] [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: 04/24/2023] [Accepted: 05/12/2023] [Indexed: 05/25/2023]
Abstract
AIMS Advanced gallbladder carcinoma (AGBC) carries a poor prognosis with dismal survival. There are no data regarding HER2/ERBB2 expression in AGBC. This study evaluated the overexpression of HER2/ERBB2 in cytological aspirates from AGBCs to identify potential patients for whom anti-HER2 targeted therapies can benefit. METHODS This prospective, case-control study was performed on 50 primary AGBC cases. A detailed cytomorphological assessment, followed by immunocytochemistry (ICC) for HER2/ERBB2, was performed on AGBC cell blocks. A similar number of age-matched and gender-matched resected chronic cholecystitis specimens were included as controls. Fluorescence in situ hybridisation (FISH) was performed in equivocal cases. RESULTS A total of 10 (20%) cases showed positive (3+), 19 (38%) equivocal (2+) expression and 21 (42%) were negative on HER2/ERBB2 ICC. None of the equivocal cases demonstrated HER2 amplification by FISH. Among the controls, none showed positive (3+) immunoexpression, 23 (46%) demonstrated equivocal expression and 27 (54%) were negative. On statistical analysis, HER2/ERBB2 overexpression was significantly associated with AGBC compared with the controls. Of all the clinical, radiological and cytomorphological parameters, the predominant papillary or acinar arrangements of the tumour cells were significantly associated with HER2/ERBB2 overexpression. CONCLUSIONS This is the first study to evaluate the expression of HER2/ERBB2 on cytological aspirates in AGBC using ICC and FISH. HER2/ERBB2 overexpression(20%) was significantly associated with AGBC. Furthermore, predominant papillary or acinar arrangements of tumour cells in the cytological smears were significantly associated with HER2/ERBB2 overexpression. They can serve as potential predictors of HER2/ERBB2 overexpression to select AGBC patients for anti-HER2 targeted therapies.
Collapse
Affiliation(s)
- Pragya Verma
- Department of Pathology, PGIMER, Chandigarh, India
| | - Parikshaa Gupta
- Department of Cytology and Gynecologic Pathology, PGIMER, Chandigarh, India
| | - Nalini Gupta
- Department of Cytology and Gynecologic Pathology, PGIMER, Chandigarh, India
| | - Radhika Srinivasan
- Department of Cytology and Gynecologic Pathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Pankaj Gupta
- Department of Radiodiagnosis, PGIMER, Chandigarh, India
| | - Usha Dutta
- Department of Gastroenterology, PGIMER, Chandigarh, India
| | - Shelly Sharma
- Department of Cytology and Gynecologic Pathology, PGIMER, Chandigarh, India
| | - Radha Uppal
- Department of Cytology and Gynecologic Pathology, PGIMER, Chandigarh, India
| | - Ritambhra Nada
- Histopathology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Anupam Lal
- Department of Radiodiagnosis, PGIMER, Chandigarh, India
| |
Collapse
|