1
|
Liang QB, Zhang ZL. An electrochemical fluorescence dual-mode strategy for HER2-positive breast cancer cell detection. Talanta 2025; 292:127974. [PMID: 40112589 DOI: 10.1016/j.talanta.2025.127974] [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: 01/30/2025] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
Breast cancer is one of the most prevalent malignancies worldwide, with HER2-overexpressing subtypes exhibiting increased aggressiveness and poorer prognosis. Accurate identification of HER2-positive subtypes is essential for the effective implementation of HER2-targeted therapy. In this study, an electrochemical fluorescence dual-mode strategy was developed for the high sensitive detection of HER2-positive breast cancer cells. Immunofluorescent quantum dot probes (IFQDs) with both fluorescence and enzyme catalysis were constructed. It labelled HER2 sites on the cell membrane to enable fluorescent imaging and cell counting. Furthermore, alkaline phosphatase (ALP) on the probe surface catalyzed the reduction of silver on the surface of the Au NPs@ITO electrode through enzyme-induced metallization, thereby enabling quantitative detection of the cells via stripping voltammetry. The application of two methods, namely enzyme-induced metallization and enrichment of signal species on the electrode surface, significantly enhanced the sensitivity of this analytical strategy. The self-monitoring of dual signals achieved more accurate analytical performance. The dual-mode strategy demonstrated satisfactory results in identifying breast cancer cells with varying HER2 expression levels and even in complex samples. It indicated that the electrochemical fluorescence dual-mode strategy had potential for typing and quantitative detection of cells with varying HER2 expression levels.
Collapse
Affiliation(s)
- Qiao-Bin Liang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, PR China
| | - Zhi-Ling Zhang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, PR China.
| |
Collapse
|
2
|
Akbari B, Hasan MM, Islam SM. Advances in targeted therapy for triple-negative breast cancer: a review of key antigens and recent advances. J Drug Target 2025:1-34. [PMID: 40515614 DOI: 10.1080/1061186x.2025.2520306] [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: 03/21/2025] [Revised: 05/16/2025] [Accepted: 06/09/2025] [Indexed: 06/16/2025]
Abstract
The most aggressive subtype of breast cancer is triple-negative breast cancer (TNBC), which affects about 10-15% of all breast cancer cases. TNBC is associated with a poor prognosis and drug resistance due to the lack of oestrogen, progesterone, and HER2 receptors. Developing targeted immunotherapy for TNBC was challenged by identifying TNBC-specific antigens that can be suitable targets for antibody and nanobody-based therapies. Evidence from cancer- targeted therapy demonstrates that treatment outcomes are more successful when the target antigen is either overexpressed in tumour tissue or exhibits tumour specificity. Several antigens have been overexpressed in TNBC, including programmed cell death protein 1 (PD-1), programmed death-ligand 1(PD-L1), mesothelin (MSLN), trophoblast cell-surface antigen 2 (Trop-2), tumour endothelial marker 8 (TEM8), etc. There have been investigations targeting these antigens with antibodies, nanobodies, small molecules, peptides, and miniproteins for targeted treatment of TNBC. Antibodies known as Aembrolizumab, Atezolizumab, and Sacituzumab Govitecan-hziy have been approved by the FDA, and many are under investigation. The present review discusses the antigens with high expression in TNBC, their role in cancer development and progression, and the targeted therapies like antibodies, recombinant proteins, and antibody-drug conjugates (ADC).
Collapse
Affiliation(s)
- Bahman Akbari
- Department of Medical Biotechnology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Chemistry, Delaware State University, Dover, DE, USA
| | - Md Mehedi Hasan
- Department of Chemistry, Delaware State University, Dover, DE, USA
| | - Shahidul M Islam
- Department of Chemistry, Delaware State University, Dover, DE, USA
| |
Collapse
|
3
|
Ding N, Hermans KEPE, van Nijnatten TJA, Engelen SME, Tol J, Kooreman L, Vrancken-Peeters MJTFD, Siesling S, Voogd AC, Tjan-Heijnen VCG, Geurts SME. Overall survival of patients with de Novo HER2-positive metastatic breast cancer in the Netherlands from 2008 to 2017: A population-based cohort study of systemically treated patients. Eur J Cancer 2025; 222:115475. [PMID: 40306118 DOI: 10.1016/j.ejca.2025.115475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/09/2025] [Accepted: 04/22/2025] [Indexed: 05/02/2025]
Abstract
AIM This study aims to determine whether real-world overall survival (OS) of patients with de Novo HER2-positive (HER2 +) metastatic breast cancer (MBC) in the Netherlands improved over time. METHODS Data of patients diagnosed with de Novo HER2 + MBC in 2008-2017 were retrieved from the Netherlands Cancer Registry. OS was estimated per two-year period using the Kaplan-Meier method and compared using the log-rank test for trend and the multivariable Cox proportional hazards analysis, adjusted for baseline characteristics and local therapy. First-given systemic and local therapy use was determined per two-year period. RESULTS Among 1458 patients included, 99 % (1452/1458) were female and 53 % (775/1458) aged 50-74 years at diagnosis. Comparing patients diagnosed with de Novo HER2 + MBC in 2008-2009 versus 2016-2017, the median OS improved from 30·9 months (95 %CI:25·0-35·4) to 57·3 months (95 %CI:46·7-68·1) (p-value for trend<0·001) (adjusted hazard rate ratio =0·49, 95 %CI:0·40-0·59). The use of any HER2-targeted therapy increased from 64 % (178/279) to 84 % (287/340), while the use of pertuzumab-based therapy increased from 0 % to 67 % (227/340) over the same period. The use of surgery remained stable over time, while radiotherapy use for both the primary tumour and metastases increased in recent years, from 16 % (44/279) and 15 % (42/279) in 2008-2009-22 % (76/340) and 26 % (88/340) in 2016-2017, respectively. CONCLUSION OS has doubled over ten years, for patients diagnosed with de Novo HER2 + MBC in the Netherlands between 2008 and 2017. No change in metastatic presentation was observed over time, suggesting the improved OS is most likely explained by treatment improvements.
Collapse
Affiliation(s)
- Nan Ding
- Department of Medical Oncology, GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre, PO Box 5800, Maastricht, AZ 6202, the Netherlands
| | - Karlijn E P E Hermans
- Department of Medical Oncology, GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre, PO Box 5800, Maastricht, AZ 6202, the Netherlands
| | - Thiemo J A van Nijnatten
- Department of Radiology and Nuclear Medicine, GROW, Maastricht University Medical Centre, PO Box 5800, Maastricht, AZ 6202, the Netherlands
| | - Sanne M E Engelen
- Department of Surgery, Maastricht University Medical Centre, PO Box 5800, Maastricht, AZ 6202, the Netherlands
| | - Jolien Tol
- Department of Medical Oncology, Jeroen Bosch Hospital, P.O. Box 90153, s-Hertogenbosch, ME 5200, the Netherlands
| | - Loes Kooreman
- Department of Pathology, GROW, Maastricht University Medical Centre, PO Box 5800, Maastricht, AZ 6202, the Netherlands
| | - Marie-Jeanne T F D Vrancken-Peeters
- Department of Surgical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands & Department of Surgery, Amsterdam University Medical Centre, the Netherlands
| | - Sabine Siesling
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation, Utrecht, the Netherlands
| | - Adri C Voogd
- Department of Epidemiology, Maastricht University, PO Box 616, Maastricht, MD 6200, the Netherlands
| | - Vivianne C G Tjan-Heijnen
- Department of Medical Oncology, GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre, PO Box 5800, Maastricht, AZ 6202, the Netherlands
| | - Sandra M E Geurts
- Department of Medical Oncology, GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre, PO Box 5800, Maastricht, AZ 6202, the Netherlands.
| |
Collapse
|
4
|
Wei S, Ma W, Xie S, Liu S, Xie N, Li W, Zhang B, Liu J. Hyperoside Protects Trastuzumab-Induced Cardiotoxicity via Activating the PI3K/Akt Signaling Pathway. Cardiovasc Drugs Ther 2025; 39:481-490. [PMID: 37943365 DOI: 10.1007/s10557-023-07522-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/25/2023] [Indexed: 11/10/2023]
Abstract
PURPOSE Trastuzumab is a landmark agent in the treatment of human epidermal growth factor receptor-2(HER2)-positive breast cancer. Nevertheless, trastuzumab also comes with unexpected cardiac side effects. Hyperoside is a natural product that serves beneficial roles in cardiovascular disease. This study aimed to explore the effect and mechanism of hyperoside in trastuzumab-induced cardiotoxicity. METHODS A female C57BL/6 mice cardiotoxicity model was established via intraperitoneally injecting with trastuzumab (10 mg/kg/day, once every other day, cumulative dosage to 40 mg/kg) with or without hyperoside (15 or 30 mg/kg/day) administration. In vitro, the H9c2 cells were exposed to 1 μM trastuzumab with or without hyperoside (100 or 200 μM) administration. Cardiac function was evaluated by echocardiographic, myocardial enzymes levels, and pathological section examinations. TUNEL staining and Annexin V-FITC/ propidium iodide flow cytometry were used to analyze the cardiomyocyte apoptosis. RESULTS Compared to the control group, the LVEF, LVFS was decreased and the concentrations of cTnT, CK, CK-MB and LDH in mice were significantly increased after treatment with trastuzumab. Collagen deposition and cardiomyocyte hypertrophy were observed in the myocardium of the trastuzumab group. However, these changes were all reversed by different doses of hyperoside. In addition, hyperoside attenuated trastuzumab-induced myocardium apoptosis and H9c2 cells apoptosis through inhibiting the expressions of cleaved caspase-3 and Bax. Trastuzumab abolished the PI3K/Akt signaling pathway in mice and H9c2 cells, while co-treatment of hyperoside effectively increased the ratio of p-Akt/Akt. CONCLUSION Hyperoside inhibited trastuzumab-induced cardiotoxicity through activating the PI3K/Akt signaling pathway. Hyperoside may be a promising therapeutic approach to trastuzumab-induced cardiotoxicity.
Collapse
Affiliation(s)
- Shanshan Wei
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China
| | - Wanjun Ma
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Suifen Xie
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China
| | - Sa Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China
| | - Ning Xie
- Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha, 410013, China
| | - Wenqun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China
| | - Bikui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China
| | - Jian Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
5
|
Abdulkareem NM, Bhat R, Castillo M, Jung SY, Vasaikar S, Nanda S, Ruiz A, Shea M, Cao W, Veeraraghavan J, Kim HY, Bawa-Khalfe T, Hussain T, Liu X, Gunaratne P, Schiff R, Trivedi MV. Interactions between ADGRF1 (GPR110) and extracellular matrix proteins govern its effects on tumorigenesis in HER2-positive breast cancer. Br J Pharmacol 2025; 182:2524-2541. [PMID: 39965212 DOI: 10.1111/bph.17463] [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: 03/07/2024] [Revised: 12/22/2024] [Accepted: 12/28/2024] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND AND PURPOSE We and others have previously shown that ADGRF1, an adhesion G protein-coupled receptor, is overexpressed and associated with poor survival in many cancers, including human epidermal growth factor receptor-2 (HER2) breast cancer (BC). Also, we have reported the tumour-promoting function of ADGRF1 using preclinical models of HER2+ BC. In this study, we investigated the effect of ADGRF1 overexpression in an orthotopic in vivo model as well as downstream signalling of ADGRF1 in HER2+ BC. EXPERIMENTAL APPROACH We utilized a doxycycline (Dox)-induced ADGRF1 overexpression system in HER2+ BC cell lines and performed various in vitro and in vivo studies. Following ADGRF1 overexpression in the presence/absence of Matrigel, laminin-111 or collagen-IV, we performed the mammosphere assay to assess the tumorigenicity of breast epithelial cells, as well as cAMP/IP1 assays and RNA-sequencing, to understand the receptor function and pharmacology. We conducted cross-linking-aided immunoprecipitation and mass spectrometry to confirm the physical interaction between ADGRF1 and the extracellular matrix proteins present in Matrigel. KEY RESULTS We found that ADGRF1 switched from a tumour-promoting to tumour-suppressive function upon interaction with laminin-111. Interaction of ADGRF1 with laminin-111 resulted in inhibition of Gαs coupling and STAT3 phosphorylation, induction of senescence, increase in HER2 expression, and improvement of sensitivity to anti-HER2 drugs in HER2+ BC. CONCLUSIONS ADGRF1 switches from a tumour-promoting to tumour-suppressive function upon interaction with laminin-111, leading to improvements in sensitivity to anti-HER2 drugs. Leveraging ADGRF1 interactions with laminin-111 may allow the design of novel therapies against ADGRF1 in HER2+ BC.
Collapse
Affiliation(s)
- Noor Mazin Abdulkareem
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas, USA
| | - Raksha Bhat
- Department of Pharmacy Practice and Translational Research, University of Houston, Houston, Texas, USA
| | - Micah Castillo
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Sung Yun Jung
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Suhas Vasaikar
- Clinical Biomarker and Diagnostics, Seagen, Bothell, Washington, USA
| | - Sarmistha Nanda
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
| | - Alexis Ruiz
- Department of Pharmacy Practice and Translational Research, University of Houston, Houston, Texas, USA
| | - Martin Shea
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
| | - Wangjia Cao
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas, USA
| | - Jamunarani Veeraraghavan
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Hee-Yong Kim
- Laboratory of Molecular Signaling, National Institute of Alcohol Abuse and Alcoholism, Rockville, Maryland, USA
| | - Tasneem Bawa-Khalfe
- Center for Nuclear Receptors & Cell Signaling, Department of Biology & Biochemistry, University of Houston, Houston, Texas, USA
| | - Tahir Hussain
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas, USA
| | - Xinli Liu
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas, USA
| | - Preethi Gunaratne
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Rachel Schiff
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Meghana V Trivedi
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas, USA
- Department of Pharmacy Practice and Translational Research, University of Houston, Houston, Texas, USA
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
6
|
Mohamed G, Hamdy O, Alkallas A, Tahoun Y, Gomaa MM, Moaz I, Orabi A, Elzohery YH, Zakaria AS, Eltohamy MI. Role of artificial intelligence -based machine learning model in predicting HER2/neu gene status in breast cancer. Pathol Res Pract 2025; 270:155927. [PMID: 40233530 DOI: 10.1016/j.prp.2025.155927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 03/09/2025] [Accepted: 03/26/2025] [Indexed: 04/17/2025]
Abstract
Our study investigated the predictive efficacy of AI-based Machine Learning (ML) model for determining HER2 status in a population of 3424 breast cancer patients. Multivariate logistic regression analysis identified several independent variables that were predictive of HER2 positivity, namely age ≤ 40 years, tumor multicentricity, high tumor grade, high-grade DCIS, N3 stage disease, and negative ER status (p < 0.05). These findings suggest that patients presenting with these factors may benefit from more aggressive and targeted therapies. Furthermore, XGBoost ML model was trained using the dataset of 3324 patients, which was divided into an 80 % training set and a 20 % test set. The model achieved an impressive accuracy of 95 % on both training and test sets, as evidenced by the area under the curve (AUC) values of 0.95. The model ranked the presence of DCIS, DCIS component (major versus minor), DCIS grade, multiplicity of the tumor, and ER status as the top four variables for predicting HER2/neu status. To validate the performance of the proposed model, blind HER2 status data from an external validation cohort of 100 cases were utilized. Notably, the model demonstrated a sensitivity of 90.5 %, indicating its ability to accurately identify HER2-positive cases, and a specificity of 84.4 %, suggesting its capability to correctly classify HER2-negative cases. These results highlight the promising predictive efficacy of AI-based ML in determining HER2 status in breast cancer patients. The model's ability to accurately identify HER2-positive cases can assist in guiding treatment decisions, ensuring that patients receive appropriate and targeted therapies. However, further research with larger datasets is necessary to validate and generalize these findings.
Collapse
Affiliation(s)
- Ghada Mohamed
- Department of Pathology, National Cancer Institute, Cairo University, Egypt.
| | - Omar Hamdy
- Faculty of Engineering, Computer department, Cairo University, Egypt
| | - Anwar Alkallas
- Data analyst, Baheya Foundation for Early Detection And Management Of Breast Cancer, Egypt
| | | | - Mohammed Mohammed Gomaa
- Radiodiagnosis Department, National Cancer Institute, Cairo University, Egypt; Radiodiagnosis Department, Baheya Foundation for Early Detection and Management of Breast Cancer, Egypt
| | - Inas Moaz
- Epidemiology and preventive medicine department, National Liver Institute, Menoufia university, Egypt
| | - Ahmed Orabi
- Surgical oncology Department, National Cancer Institute, Cairo University, Egypt
| | | | - Al-Shimaa Zakaria
- Department of Pathology, National Cancer Institute, Cairo University, Egypt
| | | |
Collapse
|
7
|
Robbins CJ, Bates KM, Rimm DL. HER2 testing: evolution and update for a companion diagnostic assay. Nat Rev Clin Oncol 2025; 22:408-423. [PMID: 40195456 DOI: 10.1038/s41571-025-01016-y] [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: 03/13/2025] [Indexed: 04/09/2025]
Abstract
Human epidermal growth factor receptor 2 (HER2; encoded by ERBB2) testing has been a cornerstone of patient selection for HER2-targeted therapies, principally in breast cancer but also in several other solid tumours. Since the introduction of HercepTest as the original companion diagnostic for trastuzumab, HER2 assessment methods have evolved substantially, incorporating various testing modalities, from western blots, immunohistochemistry and fluorescence in situ hybridization, to early chromogenic quantitative methods and, probably in the future, fully quantitative methods. The advent of highly effective HER2-targeted antibody-drug conjugates with clinical activity at low levels of HER2 expression, such as trastuzumab deruxtecan, has necessitated the re-evaluation of HER2 testing, particularly for HER2-low tumours. In this Review, we provide an in-depth overview of the evolution of HER2 testing, the current clinical guidelines for HER2 testing across various solid tumours, challenges associated with current testing methodologies and the emerging potential of quantitative techniques. We discuss the importance of accurately defining HER2-low expression for therapeutic decision-making and how newer diagnostic approaches, such as quantitative immunofluorescence and RNA-based assays, might address the limitations of traditional immunohistochemistry-based methods. As the use of HER2-targeted therapies continues to expand to a wider range of tumour types, ensuring the precision and accuracy of HER2 testing will be crucial for guiding treatment strategies and improving patient outcomes.
Collapse
Affiliation(s)
- Charles J Robbins
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Katherine M Bates
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - David L Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
8
|
Takeda T, Matsumoto J, Sakai T, Iwata N, Hamano H, Koyama T, Ariyoshi N, Zamami Y. Comprehensive analysis of adverse event profile changes with pertuzumab addition to trastuzumab-based breast cancer therapy: Disproportionality analysis using VigiBase. Br J Clin Pharmacol 2025. [PMID: 40344285 DOI: 10.1002/bcp.70094] [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] [Revised: 03/23/2025] [Accepted: 04/21/2025] [Indexed: 05/11/2025] Open
Abstract
AIMS Pertuzumab is used in combination with trastuzumab-based therapy for HER2-positive breast cancer. However, real-world safety information on pertuzumab remains limited. This study assessed the safety of adding pertuzumab to trastuzumab-based therapy for HER2-positive breast cancer using real-world data. METHODS VigiBase, the World Health Organization's global database of adverse events (AEs), containing reports from November 1967 to December 2023, was used. Signals for pertuzumab-associated AEs in breast cancer cases were detected using the reporting odds ratio (ROR). RESULTS Signals of trastuzumab plus pertuzumab relative to trastuzumab alone were detected in gastrointestinal disorders (ROR: 1.45, 95% confidence interval: 1.26-1.67), including diarrhoea (3.49, 2.83-4.30); infections and infestations (1.54, 1.24-1.91); and skin and subcutaneous tissue disorders (ROR: 1.63, 1.40-1.90), including pruritus (1.96, 1.51-2.55) and rash (1.63, 1.20-2.23). Further, signals of trastuzumab plus docetaxel plus pertuzumab relative to those of trastuzumab plus docetaxel were detected in gastrointestinal disorders (1.63, 1.38-1.93), including nausea (1.72, 1.24-2.39) and vomiting (1.48, 1.01-2.17), and in nervous system disorders (1.50, 1.20-1.87), including paraesthesia (2.60, 1.33-5.08) and peripheral sensory neuropathy (5.94, 1.79-19.71). The frequency of AEs causing or prolonging hospitalization was increased with trastuzumab plus pertuzumab compared to that with trastuzumab alone (1.18, 1.00-1.38). CONCLUSIONS AE profiles after the addition of pertuzumab to trastuzumab-based therapy were comprehensively identified. The findings in this study highlight the importance of considering these AEs when selecting pertuzumab combination therapy to ensure the safety of patients with breast cancer.
Collapse
Affiliation(s)
- Tatsuaki Takeda
- Department of Education and Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
| | - Jun Matsumoto
- Department of Education and Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
- Department of Personalized Medicine and Preventive Healthcare Sciences, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Tomonori Sakai
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
- Department of Clinical Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Naohiro Iwata
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
| | - Hirofumi Hamano
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
- Department of Clinical Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Toshihiro Koyama
- Department of Health Data Science, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Noritaka Ariyoshi
- Department of Education and Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Department of Personalized Medicine and Preventive Healthcare Sciences, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yoshito Zamami
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
- Department of Clinical Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
9
|
Zhang D, Wang Z, Inuzuka H, Wei W. Proximity-induced membrane protein degradation for cancer therapies. RSC Med Chem 2025:d5md00141b. [PMID: 40365034 PMCID: PMC12066958 DOI: 10.1039/d5md00141b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
The selective modulation of membrane proteins presents a significant challenge in drug development, particularly in cancer therapies. However, conventional small molecules and biologics often face significant hurdles in effectively targeting membrane-bound proteins, largely due to the structural complexity of these proteins and their involvement in intricate cellular processes. In light of these limitations, proximity-induced protein modulation has recently emerged as a transformative approach. It leverages molecule-induced proximity strategies to commandeer endogenous cellular machinery for precise protein manipulation. One of these modulatory strategies is protein degradation, wherein membrane-targeting degraders derived from proximity-induction approaches offer a unique therapeutic avenue by inducing the irreversible removal of key oncogenic and immune-regulatory proteins to combat cancer. This review explores the fundamental principles underlying proximity-driven membrane protein degradation, highlighting key strategies such as LYTACs, PROTABs, TransTACs, and IFLD that are reshaping targeted cancer therapy. We discuss recent technological advancements in the application of proximity-induced degraders across breast cancer, lung cancer, immunotherapy, and other malignancies, underscoring how these innovative approaches have demonstrated significant therapeutic potential. Lastly, while these emerging technologies offer significant promise, they still face substantial limitations, including drug delivery, selectivity, and resistance mechanisms that need to be addressed to achieve successful clinical translation.
Collapse
Affiliation(s)
- Dingpeng Zhang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School Boston MA 02215 USA
| | - Zhen Wang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School Boston MA 02215 USA
| | - Hiroyuki Inuzuka
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School Boston MA 02215 USA
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School Boston MA 02215 USA
| |
Collapse
|
10
|
Atampugbire G, Adomako EEA, Quaye O. In Vitro Antiviral Assays: A Review of Laboratory Methods. Assay Drug Dev Technol 2025; 23:165-179. [PMID: 39804198 DOI: 10.1089/adt.2024.075] [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: 05/31/2025] Open
Abstract
Viral diseases remain a significant challenge for global health with rising fatalities each year. In vitro assays are crucial techniques that have been utilized by researchers in the quest to develop antiviral therapies. These assays mimic the internal conditions of a living system and make it possible to study how antiviral compounds interact with such systems in a laboratory setting. Thus, the importance of in vitro assays cannot be overemphasized, as they provide an accurate means for assessing the efficacy of potential antiviral compounds. This review offers an overview of in vitro antiviral assays, the different types of cell lines used, and emerging techniques and applications that have been developed in recent times. The current review also assesses challenges that are encountered in antiviral drug research, as well as emerging technologies like microfluidics and three-dimensional cell cultures. The integration of computational models and multiparametric assays into antiviral research was noted to significantly improve antiviral drug development process.
Collapse
Affiliation(s)
- Gabriel Atampugbire
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana
| | | | - Osbourne Quaye
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana
| |
Collapse
|
11
|
Abdulaziz O, Khan FR, Alharthi NS, Alhuthali HM, Hazazi A, Alzahrani HA, Gharib AF, Alsalmi OA, Hawsawi NM, Alhazmi AY. Computational insights into overcoming resistance mechanisms in targeted therapies for advanced breast cancer: focus on EGFR and HER2 co-inhibition. J Biomol Struct Dyn 2025; 43:4215-4226. [PMID: 38234016 DOI: 10.1080/07391102.2024.2301766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 12/30/2023] [Indexed: 01/19/2024]
Abstract
In the present study, the formation of a heterodimer involving both epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 (HER2) has been explored as a potential therapeutic mechanism to inhibit the progression of breast cancer. Virtual screening using molecular docking resulted in the three hit compounds (ZINC08382411, ZINC08382438, and ZINC08382292) with minimum binding scores and commonly binding to both receptors. Further, MD simulation analysis of these complexes illustrated the high stability of these compounds with EGFR and HER2. RMSD showed that ZINC08382411 displayed the most stable RMSD of 2 - 3 Å when bound to both receptors, suggesting to have strong compatibility with the active site of the receptor. Hydrogen bond analysis showed that ZINC08382411 forms the maximum number of H-bonds (2 to 3) in both EGFR and HER2 bound complexes, with the highest occupancy of 62% and 79%, respectively. Binding free energy calculation showed that ZINC08382411 possesses maximum affinity towards both the receptors with ΔGbind = -129.628 and -164.063 kJ/mol, respectively. This approach recognizes the significance of EGFR and HER2 in breast cancer development and aims to disrupt their collaborative signaling, which is known to promote the antagonistic behavior of cancer cells. By focusing on this EGFR/HER2 heterodimer, the study offers a promising avenue for identifying a potential candidate (ZINC08382411) that may inhibit breast cancer cell growth and potentially improve patient outcomes. The study's findings may contribute to the ongoing efforts to advance breast cancer treatment strategies.
Collapse
Affiliation(s)
- Osama Abdulaziz
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Taif University, Taif Province, Kingdom of Saudi Arabia
| | - Farhan R Khan
- Department of Clinical Laboratory Science,College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | - Nahed S Alharthi
- Department of Medical Laboratory, College of Applied Medical Sciences in Al-Kharj, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Hayaa M Alhuthali
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Ali Hazazi
- Department of Pathology and Laboratory Medicine, Security Forces Hospital Program, Riyadh, Kingdom of Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia
| | - Hind A Alzahrani
- Department of Basic Sciences, College of Applied of Medical Sciences, Albaha University, Albaha, Saudi Arabia
| | - Amal F Gharib
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Ohud A Alsalmi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Nahed M Hawsawi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Abdulfattah Y Alhazmi
- Pharmaceutical Practices Department, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
12
|
Bang I, Hattori T, Leloup N, Corrado A, Nyamaa A, Koide A, Geles K, Buck E, Koide S. Selective targeting of oncogenic hotspot mutations of the HER2 extracellular domain. Nat Chem Biol 2025; 21:706-715. [PMID: 39438724 DOI: 10.1038/s41589-024-01751-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/13/2024] [Indexed: 10/25/2024]
Abstract
Oncogenic mutations in the extracellular domain (ECD) of cell-surface receptors could serve as tumor-specific antigens that are accessible to antibody therapeutics. Such mutations have been identified in receptor tyrosine kinases including HER2. However, it is challenging to selectively target a point mutant, while sparing the wild-type protein. Here we developed antibodies selective to HER2 S310F and S310Y, the two most common oncogenic mutations in the HER2 ECD, via combinatorial library screening and structure-guided design. Cryogenic-electron microscopy structures of the HER2 S310F homodimer and an antibody bound to HER2 S310F revealed that these antibodies recognize the mutations in a manner that mimics the dimerization arm of HER2 and thus inhibit HER2 dimerization. These antibodies as T cell engagers selectively killed a HER2 S310F-driven cancer cell line in vitro, and in vivo as a xenograft. These results validate HER2 ECD mutations as actionable therapeutic targets and offer promising candidates toward clinical development.
Collapse
Affiliation(s)
- Injin Bang
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Takamitsu Hattori
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
| | - Nadia Leloup
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Alexis Corrado
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Atekana Nyamaa
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Akiko Koide
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
- Division of Hematology Oncology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Ken Geles
- Black Diamond Therapeutics, New York, NY, USA
| | | | - Shohei Koide
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA.
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
13
|
Li H, Yan M, Li Z, Cui X, Ji X, Fang F, Zhang Y, Wang Y, Guo X, Jing M, Gao Z, Cao H, Dong F, Wu J, Jiang C, Duan Y, Li X, Jiang Y, Jiang L, E Y, Jia Y, Zhang L, Peng P, Sun T. Pyrotinib and Nab-Paclitaxel in HER2-Positive Breast Cancer (PANHER Trial): A Prospective, Single-Arm, Phase II Trial. Cancer Sci 2025. [PMID: 40263948 DOI: 10.1111/cas.70086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/17/2025] [Accepted: 04/06/2025] [Indexed: 04/24/2025] Open
Abstract
Trastuzumab and pertuzumab combined with chemotherapy represent the standard therapy for first-line treatment of HER2-positive metastatic breast cancer (BC). Due to challenges related to availability and cost in China, it is necessary to explore treatments involving tyrosine kinase inhibitors (TKIs). In this multicenter, single-arm, open-label phase II trial, patients with HER2-positive BC were enrolled from seven hospitals in China. Patients received oral pyrotinib 400 mg once daily and intravenous nab-paclitaxel 125 mg/m2 on days 1, 8, and 15 of each 28-day cycle until disease progression or intolerable toxicity. The primary endpoint was the objective response rate (ORR). Between December 2019 and December 2021, 51 patients were enrolled. Among all enrolled patients, 48 had at least one response evaluation. Of these evaluable patients, 39 patients achieved responses, resulting in a positive study outcome. The ORR was 76.5% (95% CI, 62.5%-87.2%), and the disease control rate was 94.1% (95% CI, 83.8-98.8). As of January 24, 2024, the median follow-up duration was 29.2 months (IQR, 24.9-33.2). The median progression-free survival was 14.6 months (95% CI, 8.0-24.2), and the median overall survival was not reached. Forty-nine patients (96.1%) developed grade ≥ 3 adverse events (AEs). The most common grade ≥ 3 AEs were decreased neutrophil count (43.1%), decreased white blood cell count (43.1%), and diarrhea (23.5%). Pyrotinib combined with nab-paclitaxel demonstrated promising efficacy for HER2-positive advanced breast cancer, with an acceptable safety profile. Trial Registration: chictr.org, ChiCTR1900023653.
Collapse
Affiliation(s)
- Huan Li
- Department of Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Min Yan
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Zhaohui Li
- Department of Oncology, Anshan Cancer Hospital, Anshan, China
| | - Xiujie Cui
- Department of Oncology, Chaoyang Central Hospital, Chaoyang, China
| | - Xuening Ji
- Department of Oncology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Fengqi Fang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yuyang Zhang
- Department of Oncology, General Hospital of Fushun Mining Bureau of Liaoning Health Industry Group, Fushun, China
| | - Yan Wang
- Department of Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Xiangyu Guo
- Department of Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Mingxi Jing
- Department of Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Zhichao Gao
- Department of Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Hui Cao
- Department of Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Fangyuan Dong
- Department of Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Jie Wu
- Department of Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Cui Jiang
- Department of Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Yangyang Duan
- Department of Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Xiaorui Li
- Department of Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Yujun Jiang
- Department of Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Lei Jiang
- Department of Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Ying E
- Department of Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Yufeng Jia
- Department of Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Liang Zhang
- Department of Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Pai Peng
- Oncology Business Unit, Jiangsu Hengrui Pharmaceuticals Co., Ltd., Shanghai, China
| | - Tao Sun
- Department of Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| |
Collapse
|
14
|
Tang L, Que H, Wei Y, Yang T, Tong A, Wei X. Replicon RNA vaccines: design, delivery, and immunogenicity in infectious diseases and cancer. J Hematol Oncol 2025; 18:43. [PMID: 40247301 PMCID: PMC12004886 DOI: 10.1186/s13045-025-01694-2] [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: 01/02/2025] [Accepted: 03/23/2025] [Indexed: 04/19/2025] Open
Abstract
Replicon RNA (RepRNA) represents a cutting-edge technology in the field of vaccinology, fundamentally transforming vaccine design and development. This innovative approach facilitates the induction of robust immune responses against a range of infectious diseases and cancers. RepRNA vaccines leverage the inherent capabilities of RNA-dependent RNA polymerase associated with self-replicating repRNA, allowing for extreme replication within host cells. This process enhances antigen production and subsequently stimulates adaptive immunity. Additionally, the generation of double-stranded RNA during RNA replication can activate innate immune responses. Numerous studies have demonstrated that repRNA vaccines elicit potent humoral and cellular immune responses that are broader and more durable than those generated by conventional mRNA vaccines. These significant immune responses have been shown to provide protection in various models for infectious diseases and cancers. This article will explore the design and delivery of RepRNA vaccines, the mechanisms of immune activation, preclinical studies addressing infectious diseases and tumors, and related clinical trials that focus on safety and immunogenicity.
Collapse
Affiliation(s)
- Lirui Tang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Haiying Que
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Ting Yang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, 610041, People's Republic of China.
| | - Aiping Tong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
15
|
Erdogan M, Kelten Talu C, Guzeloz Z, Demir G, Eyiler F, Akay S, Yilmaz E, Unal OU. Role of Progesterone Receptor Level in Predicting Axillary Lymph Node Metastasis in Clinical T1-T2N0 Luminal Type Breast Cancer. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:710. [PMID: 40283001 PMCID: PMC12028829 DOI: 10.3390/medicina61040710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 03/29/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025]
Abstract
Background and Objectives: Axillary lymph node metastasis and the number of metastatic lymph nodes are important prognostic factors which are directly related to overall survival in women with breast cancer. Several factors have been identified to predict the likelihood of axillary lymph node metastasis in early-stage breast cancer. High PR expression is often more prevalent in the luminal A subgroup, which is associated with a better prognosis. The aim of this study was to determine the relationship between the percentage of PR expression and the likelihood of axillary metastasis in Her-2-negative, clinical T1-T2N0 luminal type breast cancer. Materials and Methods: A hundred and ninety-nine cases with luminal type, Her-2-negative, clinically and radiologically axilla-negative T1-T2 breast cancer who received radiotherapy were evaluated retrospectively. The pathological specimens were assessed by an experienced pathologist. Results: The statistical evaluation showed that tumor diameter greater than 2 cm, (p = 0.003), presence of lymphovascular invasion (p = 0.001), and PR expression level below 80% (p = 0.037) were identified as significant predictors of lymph node positivity in breast cancer patients. Conclusions: Percentage of progesterone receptor expression along with other molecular biological markers and clinicopathological parameters should be evaluated altogether when predicting axillary metastasis risk before surgery.
Collapse
Affiliation(s)
- Mihriban Erdogan
- Department of Radiation Oncology, University of Health Sciences Izmir College of Medicine and Izmir City Hospital, 35540 Izmir, Turkey; (Z.G.); (G.D.); (F.E.)
| | - Canan Kelten Talu
- Department of Pathology, University of Health Sciences İzmir Tepecik Training and Research Hospital, 35020 Izmir, Turkey; (C.K.T.); (E.Y.)
| | - Zeliha Guzeloz
- Department of Radiation Oncology, University of Health Sciences Izmir College of Medicine and Izmir City Hospital, 35540 Izmir, Turkey; (Z.G.); (G.D.); (F.E.)
| | - Gonul Demir
- Department of Radiation Oncology, University of Health Sciences Izmir College of Medicine and Izmir City Hospital, 35540 Izmir, Turkey; (Z.G.); (G.D.); (F.E.)
| | - Ferhat Eyiler
- Department of Radiation Oncology, University of Health Sciences Izmir College of Medicine and Izmir City Hospital, 35540 Izmir, Turkey; (Z.G.); (G.D.); (F.E.)
| | - Seval Akay
- Department of Medical Oncology, Izmir City Hospital, 35540 Izmir, Turkey; (S.A.); (O.U.U.)
| | - Ezgi Yilmaz
- Department of Pathology, University of Health Sciences İzmir Tepecik Training and Research Hospital, 35020 Izmir, Turkey; (C.K.T.); (E.Y.)
| | - Olcun Umit Unal
- Department of Medical Oncology, Izmir City Hospital, 35540 Izmir, Turkey; (S.A.); (O.U.U.)
| |
Collapse
|
16
|
Han J, Zhai X, Tao X, Li Y, Zhao Z, Yu Z, Dong D, Yang S, Lv L. Pharmacovigilance study of adverse reactions of anti-HER-2 drugs for the treatment of HER-2-positive breast cancer based on the FAERS database. Breast Cancer Res 2025; 27:54. [PMID: 40205546 PMCID: PMC11983758 DOI: 10.1186/s13058-025-02013-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: 11/27/2024] [Accepted: 03/29/2025] [Indexed: 04/11/2025] Open
Abstract
OBJECTIVE There are three categories of drugs that treat human epidermal growth factor receptor type 2 (HER-2) positive breast cancer: monoclonal antibodies (mAbs), antibody-drug conjugates (ADCs), and tyrosine kinase inhibitors (TKIs). The purpose of this study is to analyze and compare the adverse reactions of three classes of anti-HER-2 drugs to various body systems in patients based on the FDA Adverse Event Reporting System (FAERS). METHODS All data reports were extracted from the FAERS between 2004 and 2024. Data mining of adverse events associated with anti-HER-2 drugs was carried out using disproportionality analysis. A multivariate logistic regression analysis was conducted to explore the risk factors associated with AEs leading to hospitalization. RESULTS A total of 47,799 patients were screened for the three classes of drugs, among which ADC drugs caused the largest proportion of deaths. MAb has the strongest ADR signals associated with "cardiac disorders". Moreover, trastuzumab was associated with a greater risk of cardiotoxicity. Logistic regression analysis revealed that the treatment with mAbs should be wary of serious adverse reactions in "infections and infestations" and "metabolism and nutrition disorders". Moreover, "endocrine disorders" were the factor associated with the highest risk of prolonged hospitalization due to trastuzumab deruxtecan (T-DXd). The safety of tucatinib among TKI drugs is greater than that of other drugs. CONCLUSION In general, from the perspective of the effects of the three classes of drugs on the various body systems of patients, we should focus on mAb-associated "cardiac disorders", ADC-associated "hepatobiliary disorders", "respiratory, thoracic and mediastinal disorders", and TKI-associated "gastrointestinal disorders.
Collapse
Affiliation(s)
- Jinming Han
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, 116024, China
| | - Xiaohan Zhai
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, 116024, China
| | - Xufeng Tao
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, 116024, China
| | - Yunming Li
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, 116024, China
| | - Ziqi Zhao
- Department of Breast Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhan Yu
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, 116024, China
| | - Deshi Dong
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, 116024, China
| | - Shilei Yang
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, 116024, China
| | - Linlin Lv
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, 116024, China.
| |
Collapse
|
17
|
Zhou S, Qin X, Xing W, Xu Z, Wei C, Ren Y, Gong Z. Differences in treatment response and survival between HER2(2+)/FISH-positive and HER2(3+) breast cancer patients after dual-target neoadjuvant therapy: a matched case-control study. Front Oncol 2025; 15:1530793. [PMID: 40255431 PMCID: PMC12006182 DOI: 10.3389/fonc.2025.1530793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/14/2025] [Indexed: 04/22/2025] Open
Abstract
Background The efficacy of neoadjuvant therapy (NAT) comprising dual-target drugs has been confirmed among patients with human epidermal growth factor receptor 2 (HER2)-positive breast cancer (BC). Therefore, we explored the differences in responses to NAT and prognosis between patients with HER2(3+) and HER2(2+)/fluorescence in-situ hybridization (FISH)-positive BC after TCbHP-based dual-target NAT. Methods Data from patients with HER2-positive invasive BC who underwent NAT and radical surgery between January 2019 and December 2022 at the Peking University First Hospital and Cancer Hospital of Chinese Academy of Medical Sciences were retrospectively summarized. Propensity score matching (PSM) was used to reduce confounding effects. Pathological complete response (pCR) and invasive disease-free survival (IDFS) were evaluated to respectively reflect therapeutic response and patients' survival status. Results We selected 132 BC patients (66 pairs) through PSM form a cohort of 308 patients. The pCR rate of patients in the HER2(3+) group was significantly higher than that in the HER2(2+)/FISH-positive group after NAT (P<0.001). Univariate and multivariate logistic regression analyses determined that pCR was significantly affected by tumor grade, hormone receptor (HR) status, HER2 status (P<0.05). The 3-year IDFS rate of HER2(3+) BC patients was better than that of HER2(2+)/FISH-positive BC patient (P=0.083), although the difference was not statistically significant. Furthermore, multivariable Cox regression analysis exhibited that positive lymph node, HER2(3+), and pCR were independent prognostic factors for IDFS. Conclusion HER2(2+)/FISH-positive BC patients exhibited worse treatment response and prognosis than HER2(3+) BC patients after dual-target NAT, indicating that HER2 expression level is a crucial factor influencing the therapeutic efficacy and prognosis of BC patients after TCbHP-based dual-target NAT.
Collapse
Affiliation(s)
- Sicheng Zhou
- Department of Thyroid and Breast Surgery, Peking University First Hospital, Beijing, China
| | - Xuhui Qin
- Department of General Surgery, Zanhuang County Hospital of Traditional Chinese Medicine, Shijiazhuang, China
| | - Wei Xing
- Department of General Surgery, Hebei Provincial Hospital of Chinese Medicine/The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Zhao Xu
- Department of General Surgery, Hebei Provincial Hospital of Chinese Medicine/The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Chunlv Wei
- Department of General Surgery, Hebei Provincial Hospital of Chinese Medicine/The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yining Ren
- Department of General Surgery, Hebei Provincial Hospital of Chinese Medicine/The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Zixing Gong
- Department of General Surgery, Hebei Provincial Hospital of Chinese Medicine/The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, China
| |
Collapse
|
18
|
Hao C, Wang X, Shi Y, Tong Z, Li S, Liu X, Zhang L, Zhang J, Meng W, Zhang L. Combination Therapy of Pyrotinib and Metronomic Vinorelbine in HER2+ Advanced Breast Cancer after Trastuzumab Failure (PROVE): A Prospective Phase 2 Study. Cancer Res Treat 2025; 57:434-442. [PMID: 39118524 PMCID: PMC12016823 DOI: 10.4143/crt.2024.340] [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: 04/03/2024] [Accepted: 08/08/2024] [Indexed: 08/10/2024] Open
Abstract
PURPOSE Approximately 50%-74% of patients with metastatic human epidermal growth factor receptor 2 (HER2)-positive breast cancer do not respond to trastuzumab, with 75% of treated patients experiencing disease progression within a year. The combination of pyrotinib and capecitabine has showed efficacy in these patients. This study evaluates the efficacy and safety of pyrotinib combined with metronomic vinorelbine for trastuzumab-pretreated HER2-positive advanced breast cancer patients. MATERIALS AND METHODS In this phase 2 trial, patients aged 18-75 years with HER2-positive advanced breast cancer who had previously failed trastuzumab treatment were enrolled to receive pyrotinib 400 mg daily in combination with vinorelbine 40mg thrice weekly. The primary endpoint was progression-free survival (PFS), while secondary endpoints included objective response rate (ORR), disease control rate (DCR), overall survival (OS), and safety. RESULTS From October 21, 2019, to January 21, 2022, 36 patients were enrolled and received at least one dose of study treatment. At the cutoff date, 20 experienced disease progression or death. With a median follow-up duration of 35 months, the median PFS was 13.5 months (95% confidence interval [CI], 8.3 to 18.5). With all patients evaluated, an ORR of 38.9% (95% CI, 23.1 to 56.5) and a DCR of 83.3% (95% CI, 67.2 to 93.6) were achieved. The median OS was not reached. Grade 3 adverse events (AEs) were observed in 17 patients, with diarrhea being the most common (27.8%), followed by vomiting (8.3%) and stomachache (5.6%). There were no grade 4/5 AEs. CONCLUSION Pyrotinib combined with metronomic vinorelbine showed promising efficacy and an acceptable safety profile in HER2-positive advanced breast cancer patients after trastuzumab failure.
Collapse
Affiliation(s)
- Chunfang Hao
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Department of Breast Oncology, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
| | - Xu Wang
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| | - Yehui Shi
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| | - Zhongsheng Tong
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| | - Shufen Li
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| | - Xiaodong Liu
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| | - Lan Zhang
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| | - Jie Zhang
- Department of Breast Oncology, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
| | - Wenjing Meng
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| | - Li Zhang
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
19
|
Yao Z, Bai L, Nie Y. Fluorescence in situ hybridization protocol for cardiomyocytes. J Mol Cell Cardiol 2025; 201:44-51. [PMID: 39954939 DOI: 10.1016/j.yjmcc.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 02/06/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
Measuring cardiomyocyte nuclear ploidy is crucial for evaluating broader aspects of cardiac development, function, and disease progression. Fluorescence in situ hybridization (FISH) remains the gold standard for ploidy identification; however, its application in cardiomyocytes is hindered by their unique cellular complexities. Here, we describe a detailed cardiomyocyte-specific FISH (cardioFISH) protocol. CardioFISH incorporates a tailored enzymatic digestion strategy to enhance nuclear accessibility while preserving cellular integrity and minimizing sarcomere-derived autofluorescence. Additionally, we introduce a 3D nuclear visualization framework for comprehensive cardioFISH signal analysis, addressing the limitations imposed by the large nuclear dimensions of cardiomyocytes, where signals are frequently distributed across multiple imaging planes. This two-day cardioFISH protocol is applicable to various stages of cardiomyocyte development and provides a powerful tool for advancing studies of cardiomyocyte ploidy.
Collapse
Affiliation(s)
- Zehao Yao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lina Bai
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China.
| | - Yu Nie
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen 518057, China; National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Fuwai Central-China Hospital, Central China Branch of National Center for Cardiovascular Diseases, Zhengzhou 450046, China.
| |
Collapse
|
20
|
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
|
21
|
Rahadi A, Mahardya RTK, Listiani P, Herlinawaty E, Nugraha RR, Budiman DR, Suharlim C. Calibration of transition probabilities to model survival of adjuvant trastuzumab for early breast cancer in Indonesia. Int J Technol Assess Health Care 2025; 41:e18. [PMID: 40135279 PMCID: PMC11955306 DOI: 10.1017/s0266462325000157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 02/06/2025] [Accepted: 02/24/2025] [Indexed: 03/27/2025]
Abstract
OBJECTIVES Cost-effectiveness models fully informed by real-world epidemiological parameters yield the best results, but they are costly to obtain. Model calibration using real-world data/evidence (RWD/E) on routine health indicators can provide an alternative to improve the validity and acceptability of the results. We calibrated the transition probabilities of the reference chemotherapy treatment using RWE on patient overall survival (OS) to model the survival benefit of adjuvant trastuzumab in Indonesia. METHODS A Markov model comprising four health states was initially parameterized using the reference-treatment transition probabilities, obtained from published international evidence. We then calibrated these probabilities, targeting a 2-year OS of 86.11 percent from the RWE sourced from hospital registries. We compared projected OS duration and life-years gained (LYG) before and after calibration for the Nelder-Mead, Bound Optimization BY Quadratic Approximation, and generalized reduced gradient (GRG) nonlinear optimization methods. RESULTS The pre-calibrated transition probabilities overestimated the 2-year OS (92.25 percent). GRG nonlinear performed best and had the smallest difference with the RWD/E OS. After calibration, the projected OS duration was significantly lower than their pre-calibrated estimates across all optimization methods for both standard chemotherapy (~7.50 vs. 11.00 years) and adjuvant trastuzumab (~9.50 vs. 12.94 years). LYG measures were, however, similar (~2 years) for the pre-calibrated and calibrated models. CONCLUSIONS RWD/E calibration resulted in realistically lower survival estimates. Despite the little difference in LYG, calibration is useful to adapt external evidence commonly used to derive transition probabilities to the policy context, thereby enhancing the validity and acceptability of the modeling results.
Collapse
Affiliation(s)
- Arie Rahadi
- Management Sciences for Health, Arlington, VA, USA
| | | | - Putri Listiani
- Center for Health Financing Policy and Insurance Management, Gadjah Mada University, Sleman, Yogyakarta, Indonesia
| | - Eva Herlinawaty
- Center for Health Financing and Decentralization Policy, Ministry of Health Republic of Indonesia, Central Jakarta, Jakarta, Indonesia
| | | | - Dani Ramdhani Budiman
- Center for Health Financing and Decentralization Policy, Ministry of Health Republic of Indonesia, Central Jakarta, Jakarta, Indonesia
| | | |
Collapse
|
22
|
Lin Z, Wang X, Hua G, Zhong F, Cheng W, Qiu Y, Chi Z, Zeng H, Wang X. Identification of mitochondrial permeability transition-related lncRNAs as quantitative biomarkers for the prognosis and therapy of breast cancer. Front Genet 2025; 16:1510154. [PMID: 40206506 PMCID: PMC11979797 DOI: 10.3389/fgene.2025.1510154] [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: 11/07/2024] [Accepted: 03/05/2025] [Indexed: 04/11/2025] Open
Abstract
Breast cancer (BC) continues to pose a global health threat and presents challenges for treatment due to its high heterogeneity. Recent advancements in the understanding of mitochondrial permeability transition (MPT) and the regulatory roles of long non-coding RNAs (lncRNAs) offer potential insights for the stratification and personalized treatment of BC. Although the association between MPT and lncRNAs has not been widely studied, a few research studies have indicated a regulatory impact of lncRNAs on MPT, further deepening the understanding of the tumor. To identify reliable biomarkers associated with MPT for managing BC, bulk RNA-seq data of MPT-related lncRNAs acquired from The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression (GTEx) project were utilized to assess BC patients. A scoring system, termed the MPT-related score (MPTRscore), was developed using LASSO-Cox regression on data from 1,029 BC patients from TCGA-BRCA. Meanwhile, the superior prognostic accuracy of the MPTRscore was demonstrated by comparing it with biomarkers, including PAM50 subtyping for standardization. Subsequently, a clinical prediction model was created by incorporating the MPTRscore and clinical variables. This analysis revealed two distinct MPTRscore groups characterized by different biomolecular processes, tumor microenvironment (TME) patterns, and clinical outcomes. The MPTRscore was further investigated through unsupervised consensus clustering of TCGA-BRCA based on MPTRscore-related prognostic genes. Additionally, the MPTRscore was identified as an independent prognostic factor for BC and showed guiding utility in immunotherapy and chemotherapy response. Specifically, patients with a low MPTRscore exhibited better prognosis and treatment responses compared to those with a high MPTRscore. Significantly, the relevance of clustering results and MPTRscore was found to be mediated by lncRNA transcript RP11-573D15.8-018. In conclusion, MPTRscore-related clusters were identified in BC, and an integrative score was developed as a biomarker for predicting BC prognosis and therapeutic response. Additionally, molecular interactions underlying the relationship between MPTRscore-related clusters and MPTRscore were uncovered, proving insights for BC stratification. These findings may aid in prognosis determination and therapeutic decision-making for BC patients.
Collapse
Affiliation(s)
- Zhongshu Lin
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- School of Biological and Behavioural Science, Queen Mary University of London, London, United Kingdom
- Queen Mary College, Nanchang University, Nanchang, China
| | - Xinlu Wang
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Guanxiang Hua
- School of Biological and Behavioural Science, Queen Mary University of London, London, United Kingdom
- Queen Mary College, Nanchang University, Nanchang, China
| | - Fangmin Zhong
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Wangxinjun Cheng
- School of Biological and Behavioural Science, Queen Mary University of London, London, United Kingdom
- Queen Mary College, Nanchang University, Nanchang, China
| | - Yuxiang Qiu
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhe Chi
- School of Biological and Behavioural Science, Queen Mary University of London, London, United Kingdom
- Queen Mary College, Nanchang University, Nanchang, China
| | - Huan Zeng
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xiaozhong Wang
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
23
|
Palma M. Advancing Breast Cancer Treatment: The Role of Immunotherapy and Cancer Vaccines in Overcoming Therapeutic Challenges. Vaccines (Basel) 2025; 13:344. [PMID: 40333213 PMCID: PMC12030785 DOI: 10.3390/vaccines13040344] [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: 02/11/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 05/09/2025] Open
Abstract
Breast cancer (BC) remains a significant global health challenge due to its complex biology, which complicates both diagnosis and treatment. Immunotherapy and cancer vaccines have emerged as promising alternatives, harnessing the body's immune system to precisely target and eliminate cancer cells. However, several key factors influence the selection and effectiveness of these therapies, including BC subtype, tumor mutational burden (TMB), tumor-infiltrating lymphocytes (TILs), PD-L1 expression, HER2 resistance, and the tumor microenvironment (TME). BC subtypes play a critical role in shaping treatment responses. Triple-negative breast cancer (TNBC) exhibits the highest sensitivity to immunotherapy, while HER2-positive and hormone receptor-positive (HR+) subtypes often require combination strategies for optimal outcomes. High TMB enhances immune responses by generating neoantigens, making tumors more susceptible to immune checkpoint inhibitors (ICIs); whereas, low TMB may indicate resistance. Similarly, elevated TIL levels are associated with better immunotherapy efficacy, while PD-L1 expression serves as a key predictor of checkpoint inhibitor success. Meanwhile, HER2 resistance and an immunosuppressive TME contribute to immune evasion, highlighting the need for multi-faceted treatment approaches. Current breast cancer immunotherapies encompass a range of targeted treatments. HER2-directed therapies, such as trastuzumab and pertuzumab, block HER2 dimerization and enhance antibody-dependent cellular cytotoxicity (ADCC), while small-molecule inhibitors, like lapatinib and tucatinib, suppress HER2 signaling to curb tumor growth. Antibody-drug conjugates (ADCs) improve tumor targeting by coupling monoclonal antibodies with cytotoxic agents, minimizing off-target effects. Meanwhile, ICIs, including pembrolizumab, restore T-cell function, and CAR-macrophage (CAR-M) therapy leverages macrophages to reshape the TME and overcome immunotherapy resistance. While immunotherapy, particularly in TNBC, has demonstrated promise by eliciting durable immune responses, its efficacy varies across subtypes. Challenges such as immune-related adverse events, resistance mechanisms, high costs, and delayed responses remain barriers to widespread success. Breast cancer vaccines-including protein-based, whole-cell, mRNA, dendritic cell, and epitope-based vaccines-aim to stimulate tumor-specific immunity. Though clinical success has been limited, ongoing research is refining vaccine formulations, integrating combination therapies, and identifying biomarkers for improved patient stratification. Future advancements in BC treatment will depend on optimizing immunotherapy through biomarker-driven approaches, addressing tumor heterogeneity, and developing innovative combination therapies to overcome resistance. By leveraging these strategies, researchers aim to enhance treatment efficacy and ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Marco Palma
- Institute for Globally Distributed Open Research and Education (IGDORE), 03181 Torrevieja, Spain
| |
Collapse
|
24
|
Ying F, Zhou X, Chen M, Huang L, Gao L, Zhao Q, Zhang Y. Preclinical study of inetetamab combined with atezolizumab to synergistically inhibit HER2 and PD-L1 in the treatment of ovarian cancer. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200938. [PMID: 40034965 PMCID: PMC11874541 DOI: 10.1016/j.omton.2025.200938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/14/2024] [Accepted: 01/14/2025] [Indexed: 03/05/2025]
Abstract
Epithelial ovarian cancer (EOC) is the deadliest gynecological malignancy. Precision treatments are crucial for improving patient survival. This research explored the potential anti-tumor effects of combining inetetamab and atezolizumab for HER2+ EOC patients. The expressions of human epidermal growth factor receptor 2 (HER2) and programmed cell death ligand 1 (PD-L1) in EOC cells were evaluated. EOC cell-derived subcutaneous and peritoneal dissemination mouse models were used to evaluate the anti-tumor effects of inetetamab, with or without atezolizumab. The correlations between the expressions of HER2 and PD-L1 as well as the infiltration of T cells in tumors from patients and mice were analyzed by immunohistochemistry. Inetetamab suppressed the growth of HER2+ tumors in mouse models. HER2 overexpression increased PD-L1 levels in EOC cells. The expression level of HER2 is positively related to that of PD-L1 in the tumors of EOC patients as well as the infiltration of both CD4+ and CD8+ T cells. The combination of inetetamab and atezolizumab impeded the growth of HER2+ EOC tumors in vivo and induced a long-term anti-tumor effect with the elevated infiltration of CD103+CD8+ cells. These findings suggest that the combination of inetetamab and atezolizumab could be a promising precision treatment strategy for HER2+ EOC patients.
Collapse
Affiliation(s)
- Feiquan Ying
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xuyang Zhou
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Medical Ultrasound, Yueyang Central Hospital, Yueyang 414000, China
| | - Mengqing Chen
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lin Huang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lingling Gao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qing Zhao
- Department of Obstetrics and Gynecology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Yuan Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
25
|
Bar Y, Fell G, Dedeoglu A, Moffett N, Vidula N, Spring L, Wander SA, Bardia A, Ko N, Moy B, Ellisen LW, Isakoff SJ. Dynamic HER2-low status among patients with triple negative breast cancer (TNBC) and the impact of repeat biopsies. NPJ Breast Cancer 2025; 11:27. [PMID: 40069204 PMCID: PMC11897376 DOI: 10.1038/s41523-025-00741-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
Trastuzumab deruxtecan (T-DXd) is approved for HER2-low (HER2 immunohistochemistry (IHC)1+ or 2+ with non-amplified in situ hybridization (ISH)), but not HER2-0 (IHC 0) metastatic breast cancer. The impact of repeat biopsies (Bxs) in identifying new potential candidates with triple negative breast cancer (TNBC) for T-DXd treatment remains unknown. 512 consecutive patients with TNBC at diagnosis were included in the study cohort. Bxs were categorized as core, surgical, or metastatic based on the timing and method of biopsy (Bx) acquisition, and the total number of Bxs was determined for each patient. Additionally, matched biopsies were identified, and the rate of discordance in HER2 status was calculated. The proportion of patients with at least one HER2-low result increased as the number of successive Bxs increased [59%, 73%, 83%, 83%, and 100% when 1 (196 patients), 2 (231 patients), 3 (48 patients), 4 (29 patients), and ≥ 5 (8 patients) Bxs were obtained, respectively]. Among patients without a prior HER2-low result, approximately one-third demonstrated HER2-low status with each additional successive Bx. HER2 status exhibited variability between matched Bxs, with observed discordance rates of 26%, 44%, and 33% between matched core-surgical, early-metastatic, and two metastatic matched Bxs, respectively. Our findings indicate that HER2 status can vary between different Bxs taken during the disease course of patients with TNBC with the highest discordance rate observed between the primary and metastatic Bxs. For patients with metastastic HER2-0 TNBC, repeat Bxs can increase the chance of obtaining a HER2-low result, thereby offering patients a promising therapeutic option.
Collapse
Affiliation(s)
- Yael Bar
- Massachusetts General Hospital Cancer Center, Boston, MA, USA.
- Tel Aviv Sourasky Medical Center and The Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Geoffrey Fell
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Aylin Dedeoglu
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Natalie Moffett
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Neelima Vidula
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Laura Spring
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Seth A Wander
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Aditya Bardia
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - Naomi Ko
- Section of Hematology/Oncology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Beverly Moy
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Leif W Ellisen
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Steven J Isakoff
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
26
|
Carpio AR, Talubo ND, Tsai PW, Chen BY, Tayo LL. Berries as Nature's Therapeutics: Exploring the Potential of Vaccinium Metabolites in Gastric Cancer Treatment Through Computational Insights. Life (Basel) 2025; 15:406. [PMID: 40141751 PMCID: PMC11944152 DOI: 10.3390/life15030406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/02/2025] [Accepted: 03/03/2025] [Indexed: 03/28/2025] Open
Abstract
Berries from the Vaccinium genus, known for their rich array of bioactive metabolites, are recognized for their antioxidant, anti-inflammatory, and anticancer properties. These compounds, including anthocyanins, flavonoids, and phenolic acids, have attracted significant attention for their potential health benefits, particularly in cancer prevention and treatment. Gastric cancer (GC), a leading cause of cancer-related deaths worldwide, remains challenging to treat, especially in its advanced stages. This study investigates the therapeutic potential of Vaccinium species in GC treatment using computational methods. RNA sequencing revealed upregulated genes associated with GC, while network pharmacology and molecular docking approaches identified strong interactions between cyanidin 3-O-glucoside (C3G), a key bioactive metabolite. Furthermore, molecular dynamics simulations of the HSP90AA1-C3G complex demonstrated stable binding and structural integrity, suggesting that C3G may inhibit HSP90AA1, a protein involved in cancer progression. These findings highlight the therapeutic potential of Vaccinium metabolites, offering a novel approach to GC treatment by targeting key molecular pathways. This research provides valuable insights into the role of berries as natural therapeutics, supporting their integration into future gastric cancer treatment strategies.
Collapse
Affiliation(s)
- Angelica Rachel Carpio
- School of Chemical, Biological, and Materials Engineering and Sciences, School of Graduate Studies, Mapúa University, Manila 1002, Philippines; (A.R.C.); (N.D.T.)
| | - Nicholas Dale Talubo
- School of Chemical, Biological, and Materials Engineering and Sciences, School of Graduate Studies, Mapúa University, Manila 1002, Philippines; (A.R.C.); (N.D.T.)
| | - Po-Wei Tsai
- Department of Food Science, National Taiwan Ocean University, Keelung 202, Taiwan;
| | - Bor-Yann Chen
- Department of Chemical and Materials Engineering, National I-Lan University, I-Lan 260, Taiwan
| | - Lemmuel L. Tayo
- Department of Biology, School of Health Sciences, Mapúa University, Makati 1200, Philippines
| |
Collapse
|
27
|
Waks AG, Chen EL, Graham N, Frey AM, Almeida K, Attaya V, Ryding C, Abbass I, Fung A, Sussell J, Cortazar P, Harvey C, Leth D, Faggen M, Sinclair N, Walsh J, Tung N, Sinclair S, Lo S, Yardley D, Valero V, Meisel J, Ballinger TJ, Adams S, Carey LA, Rauch JK, Abramson VG, Williams NO, Chen WY, Leone JP, Schumer ST, Tayob N, Tolaney SM. Subcutaneous vs Intravenous Trastuzumab/Pertuzumab: A Time and Motion Substudy of a Phase II Trial of Adjuvant Trastuzumab/Pertuzumab for Stage I HER2+ Breast Cancer (ADEPT trial). JCO Oncol Pract 2025; 21:351-357. [PMID: 39028923 PMCID: PMC11925349 DOI: 10.1200/op.24.00021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/18/2024] [Accepted: 06/11/2024] [Indexed: 07/21/2024] Open
Abstract
PURPOSE The time required for in-clinic drug administration can substantially affect breast cancer patients' quality of life. Subcutaneous (SC) drug administration, as opposed to intravenous (IV), may reduce this time commitment. This study sought to estimate the difference in time burden between IV and SC administration of trastuzumab and pertuzumab (HP). METHODS We prospectively enrolled a subcohort of patients participating in the ADEPT trial (ClinicalTrials.gov identifier: NCT04569747, investigating adjuvant HP plus endocrine therapy for stage I human epidermal growth factor receptor 2-positive breast cancer) to this single-arm crossover time and motion substudy. Patients received two cycles of IV HP followed by two cycles of SC HP. During each cycle, time points in drug preparation and administration were captured. The primary end point was total patient time in the treatment chair. Additional end points included total patient treatment experience time and total pharmacy workflow time. A sample size of 22 patients was estimated to provide 90.7% power with two-sided alpha .05 to detect a difference of 70 minutes in the primary end point by treatment arm (IV v SC). RESULTS Twenty-two patients were enrolled. The mean total patient time in the treatment chair was 61.8 minutes shorter with SC versus IV HP (22.5 v 84.3 minutes; P < .0001). The mean total patient treatment experience time (incorporating time spent waiting for treatment initiation and time spent in the treatment chair) was 81.8 minutes shorter for SC administration (96 v 177.8 minutes; P < .0001). The pharmacy workflow time was 78.2 minutes shorter for SC versus IV formulation (41 v 119.2 minutes; P < .0001). CONCLUSION SC administration of HP shortened patient time burden by approximately 1 hour. SC drug administration can facilitate faster workflows for health care professionals and improve patients' breast cancer treatment experience.
Collapse
Affiliation(s)
- Adrienne G. Waks
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA
| | | | - Noah Graham
- Data Sciences, Dana-Farber Cancer Institute, Boston, MA
| | - Anna Mae Frey
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Kenneth Almeida
- Pharmacy and Clinical Support, Dana-Farber Cancer Institute, Boston, MA
| | - Victoria Attaya
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Current Affiliation: Olema Oncology, San Francisco, CA
| | - Cari Ryding
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | | | | | | | - Caroline Harvey
- Pharmacy and Clinical Support, Dana-Farber Cancer Institute, Boston, MA
| | - Denise Leth
- Pharmacy and Clinical Support, Dana-Farber Cancer Institute, Boston, MA
| | - Meredith Faggen
- Dana-Farber Brigham Cancer Center in Clinical Affiliation With South Shore Hospital, Weymouth, MA
| | - Natalie Sinclair
- Dana-Farber Brigham Cancer Center—Foxborough and Milford, Foxborough, MA
| | - Jeanna Walsh
- Dana-Farber/New Hampshire Oncology-Hematology, Londonderry, NH
| | - Nadine Tung
- Harvard Medical School, Boston, MA
- Beth Israel Deaconess Medical Center, Boston, MA
| | | | - Steve Lo
- Bennett Cancer Center, Stamford Health, Stamford, CT
| | | | - Vicente Valero
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jane Meisel
- Winship Cancer Institute at Emory University, Atlanta, GA
| | | | - Sylvia Adams
- Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - Lisa A. Carey
- Department of Medicine, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | | | | | | - Wendy Y. Chen
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Jose P. Leone
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Susan T. Schumer
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Nabihah Tayob
- Harvard Medical School, Boston, MA
- Data Sciences, Dana-Farber Cancer Institute, Boston, MA
| | - Sara M. Tolaney
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
28
|
Pourjamal N, Le Joncour V, Vereb G, Honkamaki C, Isola J, Leyton JV, Laakkonen P, Joensuu H, Barok M. Disitamab vedotin in preclinical models of HER2-positive breast and gastric cancers resistant to trastuzumab emtansine and trastuzumab deruxtecan. Transl Oncol 2025; 53:102284. [PMID: 39837059 PMCID: PMC11788861 DOI: 10.1016/j.tranon.2025.102284] [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: 09/20/2024] [Revised: 12/27/2024] [Accepted: 01/13/2025] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Most HER2-positive breast or gastric cancers eventually become resistant to the approved anti-HER2 antibody-drug conjugates (ADC) trastuzumab emtansine (T-DM1) and trastuzumab deruxtecan (T-DXd). Disitamab vedotin (DV) is a novel anti-HER2 ADC that binds to a different epitope on HER2 compared to trastuzumab. We assessed the efficacy of DV in breast and gastric cancer cell lines and xenografts, including tumor models resistant to T-DM1 and T-DXd. Additionally, we investigated whether combining two anti-HER2 ADCs could enhance the efficacy of the individual ADCs. METHODS The efficacy of DV, T-DM1, and T-DXd, both as single agents and in combinations, was assessed using an AlamarBlue cell proliferation assay in HER2-positive breast and gastric cancer cell lines, including those resistant to T-DM1 and T-DXd. The efficacy of DV was evaluated also in breast and gastric cancer SCID mouse xenografts that had progressed on T-DM1 and/or T-DXd. ADC combinations were tested in breast and gastric cancer xenografts. RESULTS DV was effective in cell lines resistant to T-DM1 and/or T-DXd, and it inhibited the growth of breast and gastric cancer xenografts that had progressed on T-DM1 and/or T-DXd. The combinations of DV plus T-DM1 and DV plus T-DXd showed greater efficacy than the corresponding single agents in both breast and gastric cancer cell lines and xenografts. CONCLUSIONS DV was effective in treating breast and gastric cancer xenograft tumors resistant to T-DM1 and/or T-DXd. The combination of DV with T-DM1 or T-DXd demonstrated promising activity.
Collapse
Affiliation(s)
- Negar Pourjamal
- Helsinki University Hospital and University of Helsinki, Helsinki, Finland; Laboratory of Molecular Oncology, Biomedicum, University of Helsinki, Helsinki, Finland
| | - Vadim Le Joncour
- Neuroscience Center, Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - György Vereb
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; HUN-REN-UD Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Cilla Honkamaki
- Helsinki University Hospital and University of Helsinki, Helsinki, Finland; Laboratory of Molecular Oncology, Biomedicum, University of Helsinki, Helsinki, Finland
| | - Jorma Isola
- Laboratory of Cancer Biology, Medical Faculty, University of Tampere, Tampere, Finland
| | - Jeffrey V Leyton
- School of Pharmaceutical Sciences, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Pirjo Laakkonen
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Laboratory Animal Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland; ICAN Digital Precision Medicine Flagship Program, University of Helsinki, Helsinki, Finland
| | - Heikki Joensuu
- Helsinki University Hospital and University of Helsinki, Helsinki, Finland; Laboratory of Molecular Oncology, Biomedicum, University of Helsinki, Helsinki, Finland; Department of Oncology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Mark Barok
- Helsinki University Hospital and University of Helsinki, Helsinki, Finland; Laboratory of Molecular Oncology, Biomedicum, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
29
|
Madoz-Gúrpide J, Serrano-López J, Sanz-Álvarez M, Morales-Gallego M, Rodríguez-Pinilla SM, Rovira A, Albanell J, Rojo F. Adaptive Proteomic Changes in Protein Metabolism and Mitochondrial Alterations Associated with Resistance to Trastuzumab and Pertuzumab Therapy in HER2-Positive Breast Cancer. Int J Mol Sci 2025; 26:1559. [PMID: 40004024 PMCID: PMC11855744 DOI: 10.3390/ijms26041559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/30/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
HER2 (human epidermal growth factor receptor 2) is overexpressed in approximately 15-20% of breast cancers, leading to aggressive tumour growth and poor prognosis. Anti-HER2 therapies, such as trastuzumab and pertuzumab, have significantly improved the outcomes for patients with HER2-positive breast cancer by blocking HER2 signalling. However, intrinsic and acquired resistance remains a major clinical challenge, limiting the long-term effectiveness of these therapies. Understanding the mechanisms of resistance is essential for developing strategies to overcome it and improve the therapeutic outcomes. We generated multiple HER2-positive breast cancer cell line models resistant to trastuzumab and pertuzumab combination therapy. Using mass spectrometry-based proteomics, we conducted a comprehensive analysis to identify the mechanisms underlying resistance. Proteomic analysis identified 618 differentially expressed proteins, with a core of 83 overexpressed and 118 downregulated proteins. Through a series of advanced bioinformatics analyses, we identified significant protein alterations and signalling pathways potentially responsible for the development of resistance, revealing key alterations in the protein metabolism, mitochondrial function, and signalling pathways, such as MAPK, TNF, and TGFβ. These findings identify mitochondrial activity and detoxification processes as pivotal mechanisms underlying the resistance to anti-HER2 therapy. Additionally, we identified key proteins, including ANXA1, SLC2A1, and PPIG, which contribute to the tumour progression and resistance phenotype. Our study suggests that targeting these pathways and proteins could form the basis of novel therapeutic strategies to overcome resistance in HER2-positive breast cancer.
Collapse
Affiliation(s)
- Juan Madoz-Gúrpide
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain (M.M.-G.); (S.M.R.-P.)
| | - Juana Serrano-López
- Department of Haematology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain;
| | - Marta Sanz-Álvarez
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain (M.M.-G.); (S.M.R.-P.)
| | - Miriam Morales-Gallego
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain (M.M.-G.); (S.M.R.-P.)
| | - Socorro María Rodríguez-Pinilla
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain (M.M.-G.); (S.M.R.-P.)
| | - Ana Rovira
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), 08003 Barcelona, Spain;
| | - Joan Albanell
- Department of Medical Oncology, Hospital del Mar—CIBERONC, 08003 Barcelona, Spain;
| | - Federico Rojo
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain (M.M.-G.); (S.M.R.-P.)
| |
Collapse
|
30
|
Shokoohi M, Sedaghatshoar S, Arian H, Mokarami M, Habibi F, Bamarinejad F. Genetic advancements in breast cancer treatment: a review. Discov Oncol 2025; 16:127. [PMID: 39918655 PMCID: PMC11805739 DOI: 10.1007/s12672-025-01884-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/03/2025] [Indexed: 02/09/2025] Open
Abstract
Breast cancer (BC) remains a leading cause of cancer-related deaths among women globally, highlighting the urgent need for more effective and targeted therapies. Traditional treatments, including surgery, chemotherapy, and radiation, face limitations such as drug resistance, metastasis, and severe side effects. Recent advancements in gene therapy, particularly CRISPR/Cas9 technology and Oncolytic Virotherapy (OVT), are transforming the BC treatment landscape. CRISPR/Cas9 enables precise gene editing to correct mutations in oncogenes like HER2 and MYC, directly addressing tumor growth and immune evasion. Simultaneously, OVT leverages genetically engineered viruses to selectively destroy cancer cells and stimulate robust antitumor immune responses. Despite their potential, gene therapies face challenges, including off-target effects, delivery issues, and ethical concerns. Innovations in delivery systems, combination strategies, and integrating gene therapy with existing treatments offer promising solutions to overcome these barriers. Personalized medicine, guided by genomic profiling, further enhances treatment precision by identifying patient-specific mutations, such as BRCA1 and BRCA2, allowing for more tailored and effective interventions. As research progresses, the constructive interaction between gene therapy, immunotherapy, and traditional approaches is paving the way for groundbreaking advancements in BC care. Continued collaboration between researchers and clinicians is essential to translate these innovations into clinical practice, ultimately improving BC patients' survival rates and quality of life.
Collapse
Affiliation(s)
- Marzieh Shokoohi
- Department of Life Sciences Engineering, Faculty of New Sciences & Technologies, University of Tehran, Tehran, Iran.
- Amino Techno Gene Virtual Private Laboratory, Tehran, Iran.
| | - Sadaf Sedaghatshoar
- Kent School of Social Work and Family Science, University of Louisville, Louisville, KY, USA
| | - Homaira Arian
- Pharmaceutical Biotechnology Department, Pharmacy Faculty, Anadolu University, Eskishehir, Turkey.
| | - Milad Mokarami
- Student Research Committee, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Fatemeh Habibi
- Department of Speech Therapy, School of Rehabilitation, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Bamarinejad
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
31
|
Dowsing B, Dehbi HM, Chung R, Pedra J, Worn O, Artico J, Schmid P, Roylance R, Kellman P, Moon JC, Crake T, Westwood M, Ghosh A, Andres MS, Nazir MS, Lyon AR, Chen D, Walker M, Manisty CH. HER-SAFE study design: an open-label, randomised controlled trial to investigate the safety of withdrawal of pharmacological treatment for recovered HER2-targeted therapy-related cardiac dysfunction. BMJ Open 2025; 15:e091917. [PMID: 39909533 PMCID: PMC11800297 DOI: 10.1136/bmjopen-2024-091917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 01/20/2025] [Indexed: 02/07/2025] Open
Abstract
INTRODUCTION A quarter of breast cancers show human epidermal growth factor-2 (HER2) overexpression, where targeted therapy dramatically improves survival. However, cancer therapy-related cardiac dysfunction (CTRCD) occurs in up to 15% of patients. With the interruption of HER2 therapy, if necessary, and the initiation of heart failure therapy (HFT), HER2 CTRCD recovers in over 80% of cases. The need to continue HFT in 'recovered' HER2 CTRCD following completion of HER2 therapy is unclear and there are potential significant impacts on patient's quality of life (QoL). The Randomised Controlled Trial for the Safety of Withdrawal of Pharmacological Treatment for Recovered HER2 Targeted Therapy Related Cardiac Dysfunction (HER-SAFE) aims to evaluate whether HFT can be safely withdrawn in non-high cardiovascular (CV) risk patients with 'recovered' HER2 CTRCD. METHODS AND ANALYSIS This is a multicentre, open-label randomised controlled trial investigating whether withdrawal of HFT is non-inferior to continuation in non-high CV risk, breast cancer survivors with recovered HER2 CTRCD after cancer treatment completion. The primary endpoint is the incidence of guideline-defined cardiac dysfunction or clinical heart failure. Secondary endpoints include changes in cardiac blood biomarkers, cardiovascular magnetic resonance (CMR)-derived strain and tissue mapping and heart failure symptom questionnaires. The study will recruit 90 participants who will undergo serial clinical assessment over 12 months with advanced cardiovascular imaging (CMR scans with automated analysis at baseline, 6 and 12 months), cardiac biomarker measurement (six time points over 12 months), plus complete heart failure QoL and medication disutility questionnaires. This is the first multicentre study to address this significant clinical issue. ETHICS AND DISSEMINATION This study was approved by the research ethics committee (London-London Bridge, 23/LO/0152). The results will be disseminated in peer-reviewed scientific journals. TRIAL REGISTRATION NUMBER NCT05880160.
Collapse
Affiliation(s)
- Benjamin Dowsing
- Institute of Cardiovascular Science, University College London, London, UK
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | | | - Robin Chung
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
- Whittington Health NHS Trust, London, UK
| | - Joanna Pedra
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | - Orla Worn
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | - Jessica Artico
- Institute of Cardiovascular Science, University College London, London, UK
- Imperial College Healthcare NHS Trust Cardiology Service, London, UK
| | - Peter Schmid
- Queen Mary University of London, London, UK
- Barts Health NHS Trust, London, UK
| | | | - Peter Kellman
- National Heart Lung and Blood Institute, Bethesda, Maryland, USA
| | - James C Moon
- Institute of Cardiovascular Science, University College London, London, UK
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | - Tom Crake
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | - Mark Westwood
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | - Arjun Ghosh
- Institute of Cardiovascular Science, University College London, London, UK
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | - Maria Sol Andres
- Cardio-Oncology Centre of Excellence, Royal Brompton Hospital, London, UK
| | - Muhummad Sohaib Nazir
- King's College London School of Biomedical Engineering & Imaging Sciences, London, UK
- Cardio-Oncology Centre of Excellence, Royal Brompton and Harefield Hospitals, London, UK
| | - Alexander R Lyon
- Cardio-Oncology Centre of Excellence, Royal Brompton Hospital, London, UK
| | - Daniel Chen
- Institute of Cardiovascular Science, University College London, London, UK
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | - Malcolm Walker
- Institute of Cardiovascular Science, University College London, London, UK
| | - Charlotte H Manisty
- Institute of Cardiovascular Science, University College London, London, UK
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| |
Collapse
|
32
|
Yang J, Zhou MY, Yu B, Lin Q, Yao Y, Wu HL, Zhu QW, Ye M, Xie HY, Wu JW, Cai G, Cai R, Qi WX, Chen JY, Cao L. A multicenter retrospective study of early cardiac toxicity in operable breast cancer patients receiving concurrent dual or mono anti-HER2 therapy with postoperative radiation therapy. Breast 2025; 79:103879. [PMID: 39798278 PMCID: PMC11773200 DOI: 10.1016/j.breast.2025.103879] [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: 11/07/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 01/15/2025] Open
Abstract
PURPOSE This study aims to assess whether dual anti-HER2 therapy with trastuzumab and pertuzumab increases early cardiac toxicity compared to trastuzumab alone in breast cancer (BC) patients receiving postoperative radiation therapy (RT). METHODS Consecutive operable BC patients receiving postoperative RT and trastuzumab with or without pertuzumab between January 2017 and September 2020 at seven tertiary hospitals in China were retrospectively reviewed. Cardiac examinations included echocardiography, electrocardiogram (ECG), NT-proBNP, and cTnI at baseline before RT and during the follow-up. The cardiac event is any new-onset symptomatic heart disease or abnormality in the cardiac examination after RT. RESULTS In total, 681 patients were enrolled in the analysis, of whom 567 were treated with trastuzumab-alone and 124 patients received dual anti-HER2 therapy. The median follow-up was 11 months. Multivariate analysis showed that left-sided breast cancer (HR 2.38; 95%CI 1.65-3.44, p < 0.001) and IMN RT (HR 1.47; 95 % CI 1.01-2.15, P-value = 0.047) are independent risk factors for ECG abnormalities. Age >50 years is an independent risk factor for developing LVDD (HR 5.16; 95%CI 1.17-22.73, P-value = 0.030). Dosimetric analysis showed that patients who developed subclinical cardiac events had increased mean heart dose (412.0 ± 249.6 vs. 347.2 ± 242.6 cGy, P-value = 0.010). Among right-sided patients or patients receiving anthracycline-based chemotherapy, the dual-targeted cohort had a higher risk of developing ECG abnormalities compared to the trastuzumab-only cohort. CONCLUSION Compared with trastuzumab-only, dual anti-HER2 therapy does not increase early cardiac toxicity in combination with postoperative RT in BC patients. Cardiac radiation exposure remains the primary risk factor for early toxicity.
Collapse
Affiliation(s)
- Jing Yang
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Proton Therapy, Shanghai, 201801, China
| | - Meng-Yun Zhou
- Department of Radiotherapy, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| | - Bo Yu
- Department of Radiotherapy, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin, China
| | - Qing Lin
- Department of Radiation Oncology, Shanghai Tenth People's Hospital, Shanghai, China
| | - Yuan Yao
- Department of Radiotherapy, Shanghai Ninth People's Hospital, Shanghai, China
| | - Hua-Ling Wu
- Department of Radiation Oncology, Shanghai Tenth People's Hospital, Shanghai, China
| | - Qi-Wei Zhu
- Department of Radiation Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Ming Ye
- Department of Radiation Oncology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hua-Ying Xie
- Department of Radiation Oncology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jian-Wei Wu
- Department of Radiotherapy, Shanghai Ninth People's Hospital, Shanghai, China
| | - Gang Cai
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Proton Therapy, Shanghai, 201801, China
| | - Rong Cai
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Proton Therapy, Shanghai, 201801, China
| | - Wei-Xiang Qi
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Proton Therapy, Shanghai, 201801, China
| | - Jia-Yi Chen
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Proton Therapy, Shanghai, 201801, China.
| | - Lu Cao
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Proton Therapy, Shanghai, 201801, China.
| |
Collapse
|
33
|
Sangwan N, Gangwal A, Jain P, Langtso C, Srivastava S, Dhawan U, Baweja R, Singh Y. Anthrax: Transmission, Pathogenesis, Prevention and Treatment. Toxins (Basel) 2025; 17:56. [PMID: 39998073 PMCID: PMC11860322 DOI: 10.3390/toxins17020056] [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: 11/30/2024] [Revised: 01/13/2025] [Accepted: 01/22/2025] [Indexed: 02/26/2025] Open
Abstract
Bacillus anthracis is a deadly pathogen that under unfavourable conditions forms highly resistant spores which enable them to survive for a long period of time. Spores of B. anthracis are transmitted through the contaminated soil or animal products and enter to the host through the skin, lungs or oral route and can cause cutaneous, injection, inhalation and gastrointestinal anthrax, respectively. The disease is caused by the toxin which is produced by them once they germinate within the host cell. Anthrax toxin is the major virulence factor which has the ability to kill the host cell. The role of protein kinases and phosphatases of B. anthracis in toxin production and other virulence related properties have also been reported. There are two vaccines, BioThrax and CYFENDUSTM, which are approved by the FDA-USA to prevent anthrax disease. Recently, anthrax toxin has also been shown to be a potential candidate for cancer therapeutics. Through present review, we aim to provide insights into sporulation, transmission and pathogenesis of B. anthracis as well as the current state of its prevention, treatment, vaccines and possible therapeutic uses in cancer.
Collapse
Affiliation(s)
- Nitika Sangwan
- Department of Biomedical Science, Bhaskaracharya College of Applied Sciences, University of Delhi, Delhi 110075, India
| | - Aakriti Gangwal
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, CA 94305, USA
| | - Preksha Jain
- Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi 110007, India
| | - Chokey Langtso
- Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi 110007, India
| | - Shruti Srivastava
- Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi 110007, India
| | - Uma Dhawan
- Department of Biomedical Science, Bhaskaracharya College of Applied Sciences, University of Delhi, Delhi 110075, India
| | - Renu Baweja
- Department of Biochemistry, Shivaji College, University of Delhi, Delhi 110027, India
| | - Yogendra Singh
- Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi 110007, India
| |
Collapse
|
34
|
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
|
35
|
Li F, Li J, Ji C, Wu S, Zhang S, Wang T, Bian L, Jiang Z. Novel anti-HER2 ADCs vs dual anti-HER2 antibody for HER2-positive metastatic breast cancer failed to tyrosine kinase inhibitor. Oncologist 2025; 30:oyae144. [PMID: 39786451 PMCID: PMC11783281 DOI: 10.1093/oncolo/oyae144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 05/16/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Both novel anti-human epidermal growth factor receptor 2 (HER2) antibody-drug conjugates (ADCs) and pertuzumab and trastuzumab (HP) combined with chemotherapy(C) regimens are the choice of treatment for HER2 positive metastatic breast cancer (MBC) after tyrosine kinase inhibitors (TKIs). Our team's previous research has shown significant therapeutic effects of novel anti-HER2 ADCs in patients with TKIs treatment failure. Unfortunately, there is currently no data available to compare novel anti-HER2 ADCs with HP combined with chemotherapy regimens. This study was conducted to compare the efficacy and safety of novel anti-HER2 ADCs with that of the HP combined with chemotherapy regimen in patients for whom TKI treatment failed. MATERIALS AND METHODS HER2-positive MBC who used novel anti-HER2 ADCs and HP combined with a chemotherapy regimen from January 2019 to August 2023 were included, and all patients received TKIs. The primary study endpoint was progression-free survival (PFS), while the secondary study endpoints were objective response rate (ORR), clinical benefit rate (CBR), and safety. RESULTS A total of 150 patients, of which 83 are in the novel anti-HER2 ADCs group and 67 are in the HP combined with chemotherapy. Among these novel anti-HER2 ADCs, 36 patients received treatment with trastuzumab deruxtecan (T-Dxd), and 47 patients received treatment with other new types of ADCs. The median PFS of the novel anti-HER2 ADCs group and HP combined with the chemotherapy group were 7.0 months and 8.9 months, respectively, with ORR of 51.8% and 26.9%, and CBR of 69.9% and 65.7%, respectively. In subgroup, patients receiving T-Dxd showed improvement in PFS compared to the HP combined with chemotherapy group. The most common grade 3-4 adverse events in the novel anti-HER2 ADCs group and the HP combined with chemotherapy group were neutropenia and gastrointestinal symptoms. CONCLUSIONS In HER2-positive MBC for whom TKI treatment has failed, novel anti-HER2 ADCs and the HP combined with chemotherapy regimen both showed moderate efficacy and tolerable toxicity. Novel anti-HER2 ADCs are the preferred treatment recommendation for TKI failure patients. Meanwhile, based on the results of this study, the HP combined with chemotherapy regimen may also be an option, especially for patients with low accessibility.
Collapse
Affiliation(s)
- Feng Li
- Department of Breast Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, People’s Republic of China
| | - Jianbin Li
- Department of Breast Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, People’s Republic of China
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing 100071, People’s Republic of China
| | - Chenchen Ji
- Department of Breast Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, People’s Republic of China
- Department of Physical Examination, Beijing Electric Power Hospital, Beijing 100000, People’s Republic of China
| | - Song Wu
- Department of Breast Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, People’s Republic of China
| | - Shaohua Zhang
- Department of Breast Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, People’s Republic of China
| | - Tao Wang
- Department of Breast Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, People’s Republic of China
| | - Li Bian
- Department of Breast Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, People’s Republic of China
| | - Zefei Jiang
- Department of Breast Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, People’s Republic of China
| |
Collapse
|
36
|
Saeed S, Hassan AF, Suliman A, Moustafa AEA, Alali F. Methanolic Leaves Extract of Ziziphus spina-christi Inhibits Cell Proliferation and Migration of HER2-Positive Breast Cancer via p38 MAPK Signaling Pathway. Int J Mol Sci 2025; 26:654. [PMID: 39859369 PMCID: PMC11765879 DOI: 10.3390/ijms26020654] [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: 11/27/2024] [Revised: 01/01/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) is a subtype of breast cancer that is associated with poor prognosis and low survival rates. The discovery of novel anti-cancer agents to manage this subtype of cancer is still needed. Ziziphus spina-christi (ZSC) is a plant species that is native to Qatar. It exerts various biological activities, including cytotoxicity as it contains different essential bioactive constituents, mainly rutin and quercetin. To examine the outcome of ZSC on HER2-positive breast cancer, we standardized the ZSC methanolic leaves extracted by Reverse Phase High-Performance Liquid Chromatography (RP-HPLC) analysis using the flavonoids rutin and quercetin as marker compounds. Here we used two HER2-positive breast cancer cell lines, ZR-75-1 and SK-BR-3, and the chorioallantoic membrane as an angiogenesis model. We found that ZSC extract significantly reduces viability, alters the normal morphological phenotype of HER2-positive breast cancer cells, and inhibits cell migration as well as colony formation; this is accompanied by deregulating different apoptotic markers such as Bax/Bcl-2 and NF-κB in both cell lines. Additionally, ZSC methanolic extract significantly represses the angiogenesis of the chorioallantoic membrane model. Moreover, the molecular pathway investigations pointed out that ZSC extract represses the activity of HER2 and p38 MAPK which could be the main pathways behind the effect of ZSC in HER2-positive cells. Collectively, our results support the potential role of ZSC in the management of HER2-positive breast cancer and form the basis for future investigations.
Collapse
Affiliation(s)
- Sumayyah Saeed
- College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar; (S.S.); (A.F.H.); (A.S.)
| | - Arij Fouzat Hassan
- College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar; (S.S.); (A.F.H.); (A.S.)
| | - Azza Suliman
- College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar; (S.S.); (A.F.H.); (A.S.)
| | - Ala-Eddin Al Moustafa
- College of Medicine, QU Health, Qatar University, Doha 2713, Qatar
- Oncology Department, McGill University, Montreal, QC H4A 3T2, Canada
| | - Feras Alali
- College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar; (S.S.); (A.F.H.); (A.S.)
| |
Collapse
|
37
|
Cheng TC, Hung MC, Wang LH, Tu SH, Wu CH, Yen Y, Chen CL, Whang-Peng J, Lee WJ, Liao YC, Lee YC, Pan MH, Lin HK, Tzeng HE, Guo P, Chu CY, Chen LC, Ho YS. Histamine N-methyltransferase (HNMT) as a potential auxiliary biomarker for predicting adaptability to anti-HER2 drug treatment in breast cancer patients. Biomark Res 2025; 13:7. [PMID: 39789599 PMCID: PMC11720525 DOI: 10.1186/s40364-024-00715-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Up to 23% of breast cancer patients recurred within a decade after trastuzumab treatment. Conversely, one trial found that patients with low HER2 expression and metastatic breast cancer had a positive response to trastuzumab-deruxtecan (T-Dxd). This indicates that relying solely on HER2 as a single diagnostic marker to predict the efficacy of anti-HER2 drugs is insufficient. This study highlights the interaction between histamine N-methyltransferase (HNMT) and HER2 as an adjunct predictor for trastuzumab response. Furthermore, modulation of HER2 expression by HNMT may explain why those with low HER2 expression still respond to T-Dxd. METHODS We investigated the impact of HNMT protein expression on the efficacy of anti-HER2 therapy in both in vivo and ex vivo models of patient-derived xenografts and cell line-derived xenografts. Our analysis included Förster resonance energy transfer (FRET) to assess the interaction strength between HNMT and HER2 proteins in trastuzumab-resistant and sensitive tumor tissues. Additionally, we used fluorescence lifetime imaging microscopy (FLIM), cleaved luciferase, and immunoprecipitation to study the interaction dynamics of HNMT and HER2. Furthermore, we evaluated the influence of HNMT activity on the binding of anti-HER2 antibodies to their targets through flow cytometry. We also observed the nuclear translocation of HNMT/HER2-ICD cells using fluorescent double staining and DeltaVision microscopy. Finally, ChIP sequencing was employed to identify target genes affected by the HNMT/HER2-ICD complex. RESULTS This study highlights HNMT as a potential auxiliary biomarker for diagnosing HER2 + breast cancer. FRET analysis demonstrated a significant interaction between HNMT and HER2 protein in trastuzumab-sensitive tumor tissue (n = 50), suggesting the potential of HNMT as a predictor of treatment response. Mechanistic studies revealed that the interaction between HNMT and HER2 contributes to increased HER2 protein expression at the transcriptional level, thereby impacting the efficacy of anti-HER2 therapy. Furthermore, a subset of triple-negative breast cancers characterized by HNMT overexpression was found to be sensitive to HER2 antibody-drug conjugates such as T-Dxd. CONCLUSIONS These findings offer crucial insights for clinicians evaluating candidates for anti-HER2 therapy, especially for HER2-low breast cancer patients who could gain from T-Dxd treatment. Identifying HNMT expression could help clinicians pinpoint patients who would benefit from anti-HER2 therapy.
Collapse
Affiliation(s)
- Tzu-Chun Cheng
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, China Medical University, Taichung, Taiwan
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Lu-Hai Wang
- Institute of Integrated Medicine and Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Shih-Hsin Tu
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University; & Department of Surgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chih-Hsiung Wu
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University; & Department of Surgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yun Yen
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chi-Long Chen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Pathology, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Jacqueline Whang-Peng
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wen-Jui Lee
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, China Medical University, Taichung, Taiwan
| | - You-Cheng Liao
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yu-Ching Lee
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Min-Hsiung Pan
- Institute of Food Sciences and Technology, National Taiwan University, Taipei, Taiwan
| | - Hui-Kuan Lin
- Department of Pathology, Duke University Medical Center, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Huey-En Tzeng
- Division of Hematology/Medical Oncology, Department of Medicine, Taichung Veterans General Hospital, Taichung City, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung-Hsing University, Taichung, Taiwan
| | - Peixuan Guo
- Center for RNA Nanobiotechnology and Nanomedicine, College of Pharmacy, College of Medicine, Dorothy M. Davis Heart and Lung Research Institute, and James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Cheng-Ying Chu
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- CRISPR Gene Targeting Core, Taipei Medical University, Taipei, 110, Taiwan
| | - Li-Ching Chen
- Department of Biological Science & Technology, College of Life Sciences, China Medical University, Taichung, Taiwan.
| | - Yuan-Soon Ho
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, China Medical University, Taichung, Taiwan.
| |
Collapse
|
38
|
Kan WL, Weekley CM, Nero TL, Hercus TR, Yip KH, Tumes DJ, Woodcock JM, Ross DM, Thomas D, Terán D, Owczarek CM, Liu NW, Martelotto LG, Polo JM, Pant H, Tvorogov D, Lopez AF, Parker MW. The β Common Cytokine Receptor Family Reveals New Functional Paradigms From Structural Complexities. Immunol Rev 2025; 329:e13430. [PMID: 39748163 DOI: 10.1111/imr.13430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025]
Abstract
Cytokines are small proteins that are critical for controlling the growth and activity of hematopoietic cells by binding to cell surface receptors and transmitting signals across membranes. The β common (βc) cytokine receptor family, consisting of the granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin (IL)-3, and IL-5 cytokine receptors, is an architype of the heterodimeric cytokine receptor systems. We now know that signaling by cytokine receptors is not always an "all or none" phenomenon. Subtle alterations of the cytokine:receptor complex can result in differential or selective signaling and underpin a variety of diseases including chronic inflammatory conditions and cancers. Structural biology techniques, such as X-ray crystallography and cryo-electron microscopy alongside cell biology studies, are providing detailed insights into cytokine receptor signaling. Recently, we found that the IL-3 receptor ternary complex forms higher-order assemblies, like those found earlier for the GM-CSF receptor, and demonstrated that functionally distinct biological signals arise from different IL-3 receptor oligomeric assemblies. As we enhance our understanding of the structural nuances of cytokine-receptor interactions, we foresee a new era of theranostics whereby structurally guided mechanism-based manipulation of cytokine signaling through rational/targeted protein engineering will harness the full potential of cytokine biology for precision medicine.
Collapse
Affiliation(s)
- Winnie L Kan
- Cytokine Receptor Laboratory, Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, South Australia, Australia
| | - Claire M Weekley
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria, Australia
| | - Tracy L Nero
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria, Australia
| | - Timothy R Hercus
- Cytokine Receptor Laboratory, Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, South Australia, Australia
| | - Kwok Ho Yip
- Cytokine Receptor Laboratory, Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, South Australia, Australia
| | - Damon J Tumes
- Cytokine Receptor Laboratory, Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, South Australia, Australia
| | - Joanna M Woodcock
- Cytokine Receptor Laboratory, Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, South Australia, Australia
| | - David M Ross
- Discipline of Medicine, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), University of Adelaide, Adelaide, South Australia, Australia
- Acute Leukemia Laboratory, Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, South Australia, Australia
| | - Daniel Thomas
- Discipline of Medicine, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), University of Adelaide, Adelaide, South Australia, Australia
| | - David Terán
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria, Australia
| | - Catherine M Owczarek
- CSL, Bio21 Molecular Science and Biotechnology Institute, Parkville, Victoria, Australia
| | - Nora W Liu
- Adelaide Centre for Epigenetics, School of Biomedicine, University of Adelaide, Adelaide, South Australia, Australia
- Cancer Epigenetics Program, South Australian immunoGENomics Cancer Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Luciano G Martelotto
- Adelaide Centre for Epigenetics, School of Biomedicine, University of Adelaide, Adelaide, South Australia, Australia
- Cancer Epigenetics Program, South Australian immunoGENomics Cancer Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Jose M Polo
- Adelaide Centre for Epigenetics, School of Biomedicine, University of Adelaide, Adelaide, South Australia, Australia
- Cancer Epigenetics Program, South Australian immunoGENomics Cancer Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Harshita Pant
- Discipline of Medicine, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
- Adelaide Centre for Epigenetics, School of Biomedicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Denis Tvorogov
- Cytokine Receptor Laboratory, Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, South Australia, Australia
| | - Angel F Lopez
- Cytokine Receptor Laboratory, Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, South Australia, Australia
- Discipline of Medicine, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Michael W Parker
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria, Australia
- Australian Cancer Research Foundation Rational Drug Discovery Centre, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| |
Collapse
|
39
|
Chen XC, Jiao DC, Qiao JH, Wang CZ, Sun XF, Lu ZD, Li LF, Zhang CJ, Yan M, Wei Y, Chen B, Feng YQ, Deng M, Ma MD, Plichta JK, He YW, Liu ZZ. De-escalated neoadjuvant weekly nab-paclitaxel with trastuzumab and pertuzumab versus docetaxel, carboplatin, trastuzumab, and pertuzumab in patients with HER2-positive early breast cancer (HELEN-006): a multicentre, randomised, phase 3 trial. Lancet Oncol 2025; 26:27-36. [PMID: 39612919 DOI: 10.1016/s1470-2045(24)00581-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/02/2024] [Accepted: 10/09/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND A previous phase 2 trial showed promising outcomes for patients with HER2-positive early-stage breast cancer using neoadjuvant de-escalation chemotherapy with paclitaxel, trastuzumab, and pertuzumab. We aimed to evaluate the efficacy of weekly nab-paclitaxel compared with the standard regimen of docetaxel plus carboplatin, both with trastuzumab and pertuzumab, as neoadjuvant therapies for patients with HER2-positive breast cancer. METHODS HELEN-006 was a multicentre, randomised, phase 3 trial done at six hospitals in China. We enrolled patients aged 18-70 years with untreated, histologically confirmed stage II-III invasive HER2-positive breast cancer and an Eastern Cooperative Oncology Group performance status of 0 or 1. Using an interactive response system, patients were randomly assigned (1:1) under a permuted block randomisation scheme (block size of four), stratified by tumour stage, nodal status, and hormone receptor status. Patients received either intravenous nab-paclitaxel (125 mg/m2 on days 1, 8, and 15) for six 3-week cycles, or intravenous docetaxel (75 mg/m2 on day 1) plus intravenous carboplatin (area under the concentration-time curve 6 mg/mL per min on day 1) for six 3-week cycles. Both groups also received concurrent intravenous trastuzumab, with an initial loading dose of 8 mg/kg and a maintenance dose of 6 mg/kg on day 1, as well as intravenous pertuzumab with a loading dose of 840 mg and a maintenance dose of 420 mg on day 1. This report is the final analysis of the primary endpoint, pathological complete response (ypT0/is ypN0), analysed in all patients who started treatment (modified intention to treat). The trial is registered with ClinicalTrials.gov, NCT04547907, and follow-up of the adjuvant phase is ongoing. FINDINGS Between Sept 20, 2020, and March 1, 2023, 789 patients were screened for eligibility, 689 of whom were randomly assigned (343 to the nab-paclitaxel group and 346 to the docetaxel plus carboplatin group). All 689 patients were Asian women. 669 patients received at least one dose of the study treatment and were included in the full analysis set (332 in the nab-paclitaxel group and 337 in the docetaxel plus carboplatin group). Median age of the patients was 50 years (IQR 43-55). Median follow-up time was 26 months (IQR 19-32). 220 (66·3% [95% CI 61·2-71·4]) patients in the nab-paclitaxel group had a pathological complete response, compared with 194 (57·6% [52·3-62·9]) in the docetaxel plus carboplatin group (combined odds ratio 1·54 [95% CI 1·10-2·14]; stratified p=0·011). 100 (30%) patients in the nab-paclitaxel group and 128 (38%) in the docetaxel plus carboplatin group had grade 3-4 adverse events. The most common grade 3-4 adverse events were nausea (22 [7%] in the nab-paclitaxel group vs 76 [23%] in the docetaxel plus carboplatin group), diarrhoea (25 [8%] vs 55 [16%]), and neuropathy (43 [13%] vs eight [2%]). Serious drug-related adverse events were reported in three (1%) patients in the nab-paclitaxel group and five (2%) in the docetaxel plus carboplatin group. No treatment-related deaths were reported in either group. INTERPRETATION These findings might suggest a potential advantage of nab-paclitaxel combined with trastuzumab and pertuzumab compared with the standard regimen in neoadjuvant therapy for patients with HER2-positive early breast cancer, suggesting that this new combination might establish a new standard for neoadjuvant treatment in this patient population. FUNDING National Natural Science Foundation of China, and Science and Technology Research Projects of Henan Province, China. TRANSLATION For the Chinese translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Xiu-Chun Chen
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - De-Chuang Jiao
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Jiang-Hua Qiao
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Cheng-Zheng Wang
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Xian-Fu Sun
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Zhen-Duo Lu
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Lian-Fang Li
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Chong-Jian Zhang
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Min Yan
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Ya Wei
- Department of Breast Surgery, Anyang Tumor Hospital, Anyang, China
| | - Bo Chen
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Yue-Qing Feng
- Department of Breast Surgery, Xinxiang Central Hospital, Xinxiang, China
| | - Miao Deng
- Department of Breast Surgery, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Ming-De Ma
- Department of Thyroid and Breast Surgery, Huaihe Hospital of Henan University, Kaifeng, China
| | - Jennifer K Plichta
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - You-Wen He
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
| | - Zhen-Zhen Liu
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China.
| |
Collapse
|
40
|
Wang X, Shang Y, Zhang J, Liu J, Fang Z, Liu Y, Cheng W, Duan Y, Hu A, Zhang J, Li M, Li Y, Zhang H, Rong Z, S Shakila S, Kong F, Guo B. Survival outcomes of neoadjuvant versus adjuvant therapy in patients with T1c, node-negative, human epidermal growth factor receptor 2-positive breast cancer: A Surveillance, Epidemiology, and End Results population-based study. Cancer 2025; 131:e35581. [PMID: 39306696 DOI: 10.1002/cncr.35581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 07/26/2024] [Accepted: 08/19/2024] [Indexed: 01/03/2025]
Abstract
BACKGROUND Persistent debates exist regarding the superiority of neoadjuvant therapy (NAT) over adjuvant therapy (AT) for patients with T1c, node-negative, human epidermal growth factor receptor 2-positive (HER2+) breast cancer, and relevant guidelines for these patients are lacking. METHODS Data on patients with T1cN0M0-stage HER2+ breast cancer who received chemotherapy and surgery were extracted from 2010 to 2020 from the Surveillance, Epidemiology, and End Results database. Propensity score matching (PSM) was used to create well-balanced cohorts for the NAT and AT groups. Kaplan-Meier (KM) analysis and Cox proportional hazards models were used to assess the differences between NAT and AT in terms of overall survival (OS) and breast cancer-specific survival (BCSS). Additionally, logistic regression models were used to explore factors associated with response to NAT. RESULTS After PSM, 2140 patient pairs were successfully matched, which achieved a balanced distribution between the NAT and AT groups. KM curves revealed similar OS and BCSS between patients receiving NAT and those undergoing AT. A multivariate Cox model identified achieving pathological complete response (pCR) after NAT, compared with AT, as a protective prognostic factor for OS (hazard ratio, 0.52; 95% CI, 0.35-0.77; p < .001) and BCSS (hazard ratio, 0.60; 95% CI, 0.37-0.98; p = .041). A logistic regression model revealed that White race and hormone receptor-negative status independently predicted pCR. CONCLUSIONS For patients with T1cN0M0-stage HER2+ breast cancer, NAT demonstrated comparable OS and BCSS to AT. Patients who achieved pCR after NAT exhibited significantly better survival outcomes compared with those who received AT.
Collapse
Affiliation(s)
- Xuelian Wang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuhang Shang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiayang Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jiangwei Liu
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhengbo Fang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yansong Liu
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Weilun Cheng
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yunqiang Duan
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Anbang Hu
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiarui Zhang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mingcui Li
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanling Li
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hanyu Zhang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhiyuan Rong
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Suborna S Shakila
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fanjing Kong
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Baoliang Guo
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
41
|
Zhang S, Guo L, Tao R, Liu S. Ferroptosis-targeting drugs in breast cancer. J Drug Target 2025; 33:42-59. [PMID: 39225187 DOI: 10.1080/1061186x.2024.2399181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 07/07/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
In 2020, breast cancer surpassed lung cancer as the most common cancer in the world for the first time. Due to the resistance of some breast cancer cell lines to apoptosis, the therapeutic effect of anti-breast cancer drugs is limited. According to recent report, the susceptibility of breast cancer cells to ferroptosis affects the progress, prognosis and drug resistance of breast cancer. For instance, roblitinib induces ferroptosis of trastuzumab-resistant human epidermal growth factor receptor 2 (HER2)-positive breast cancer cells by diminishing fibroblast growth factor receptor 4 (FGFR4) expression, thereby augmenting the susceptibility of these cells to HER2-targeted therapies. In tamoxifen-resistant breast cancer cells, Fascin exacerbates their resistance by repressing solute carrier family 7 member 11 (SLC7A11) expression, which in turn heightens their responsiveness to tamoxifen. In recent years, Chinese herbs extracts and therapeutic drugs have been demonstrated to elicit ferroptosis in breast cancer cells by modulating a spectrum of regulatory factors pertinent to ferroptosis, including SLC7A11, glutathione peroxidase 4 (GPX4), acyl-CoA synthetase long chain family member 4 (ACSL4), and haem oxygenase 1 (HO-1). Here, we review the roles and mechanisms of Chinese herbal extracts and therapeutic drugs in regulating ferroptosis in breast cancer, providing potential therapeutic options for anti-breast cancer.
Collapse
Affiliation(s)
- Shuxian Zhang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, China
| | - Lijuan Guo
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, China
| | - Ran Tao
- Department of Anatomy, Medical College, Dalian University, Dalian, China
| | - Shuangping Liu
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, China
| |
Collapse
|
42
|
Li J, Wang X, Zhang H, Hu X, Peng X, Jiang W, Zhuo L, Peng Y, Zeng G, Wang Z. Fenamates: Forgotten treasure for cancer treatment and prevention: Mechanisms of action, structural modification, and bright future. Med Res Rev 2025; 45:164-213. [PMID: 39171404 DOI: 10.1002/med.22079] [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: 12/13/2022] [Revised: 08/03/2024] [Accepted: 08/08/2024] [Indexed: 08/23/2024]
Abstract
Fenamates as classical nonsteroidal anti-inflammatory agents are widely used for relieving pain. Preclinical studies and epidemiological data highlight their chemo-preventive and chemotherapeutic potential for cancer. However, comprehensive reviews of fenamates in cancer are limited. To accelerate the repurposing of fenamates, this review summarizes the results of fenamates alone or in combination with existing chemotherapeutic agents. This paper also explores targets of fenamates in cancer therapy, including COX, AKR family, AR, gap junction, FTO, TEAD, DHODH, TAS2R14, ion channels, and DNA. Besides, this paper discusses other mechanisms, such as regulating Wnt/β-catenin, TGF-β, p38 MAPK, and NF-κB pathway, and the regulation of the expressions of Sp, EGR-1, NAG-1, ATF-3, ErbB2, AR, as well as the modulation of the tumor immune microenvironment. Furthermore, this paper outlined the structural modifications of fenamates, highlighting their potential as promising leads for anticancer drugs.
Collapse
Affiliation(s)
- Junfang Li
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaodong Wang
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Honghua Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaoling Hu
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Xue Peng
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Weifan Jiang
- Postdoctoral Station for Basic Medicine, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Linsheng Zhuo
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Postdoctoral Station for Basic Medicine, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yan Peng
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Guo Zeng
- Postdoctoral Station for Basic Medicine, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhen Wang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Postdoctoral Station for Basic Medicine, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
43
|
Li Y, Feng T, Wang Q, Wu Y, Wang J, Zhang W, Kong Q. High expression of SULF1 is associated with adverse prognosis in breast cancer brain metastasis. Animal Model Exp Med 2025; 8:162-170. [PMID: 38590118 PMCID: PMC11798736 DOI: 10.1002/ame2.12406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/24/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND Breast cancer is the most common cancer in women, and in advanced stages, it often metastasizes to the brain. However, research on the biological mechanisms of breast cancer brain metastasis and potential therapeutic targets are limited. METHODS Differential gene expression analysis (DEGs) for the datasets GSE43837 and GSE125989 from the GEO database was performed using online analysis tools such as GEO2R and Sangerbox. Further investigation related to SULF1 was conducted using online databases such as Kaplan-Meier Plotter and cBioPortal. Thus, expression levels, variations, associations with HER2, biological processes, and pathways involving SULF1 could be analyzed using UALCAN, cBioPortal, GEPIA2, and LinkedOmics databases. Moreover, the sensitivity of SULF1 to existing drugs was explored using drug databases such as RNAactDrug and CADSP. RESULTS High expression of SULF1 was associated with poor prognosis in advanced breast cancer brain metastasis and was positively correlated with the expression of HER2. In the metastatic breast cancer population, SULF1 ranked top among the 16 DEGs with the highest mutation rate, reaching 11%, primarily due to amplification. KEGG and GSEA analyses revealed that the genes co-expressed with SULF1 were positively enriched in the 'ECM-receptor interaction' gene set and negatively enriched in the 'Ribosome' gene set. Currently, docetaxel and vinorelbine can act as treatment options if the expression of SULF1 is high. CONCLUSIONS This study, through bioinformatics analysis, unveiled SULF1 as a potential target for treating breast cancer brain metastasis (BM).
Collapse
Affiliation(s)
- Yitong Li
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Comparative Medicine Center, Peking Union Medical College (PUMC)BeijingChina
| | - Tingting Feng
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Comparative Medicine Center, Peking Union Medical College (PUMC)BeijingChina
| | - Qinghong Wang
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Comparative Medicine Center, Peking Union Medical College (PUMC)BeijingChina
| | - Yue Wu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Comparative Medicine Center, Peking Union Medical College (PUMC)BeijingChina
| | - Jue Wang
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Comparative Medicine Center, Peking Union Medical College (PUMC)BeijingChina
| | - Wenlong Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Comparative Medicine Center, Peking Union Medical College (PUMC)BeijingChina
| | - Qi Kong
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Comparative Medicine Center, Peking Union Medical College (PUMC)BeijingChina
| |
Collapse
|
44
|
Zhao Z, Zhu L, Luo Y, Xu H, Zhang Y. Collateral lethality: A unique type of synthetic lethality in cancers. Pharmacol Ther 2025; 265:108755. [PMID: 39581504 DOI: 10.1016/j.pharmthera.2024.108755] [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] [Revised: 10/31/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024]
Abstract
Genetic interactions play crucial roles in cell-essential functions. Intrinsic genetic defects in tumors typically involve gain-of- and loss-of-function mutations in tumor suppressor genes (TSGs) and oncogenes, respectively, providing potential antitumor vulnerabilities. Moreover, tumor cells with TSG deficiencies exhibit heightened sensitivity to the inhibition of compensatory pathways. Synthetic and collateral lethality are two strategies used for exploiting novel drug targets in multiple types of cancer. Collateral lethality is a unique type of synthetic lethality that occurs when passenger genes are co-deleted in neighboring TSGs. Although synthetic lethality has already been successfully demonstrated in clinical practice, antitumor therapeutics based on collateral lethality are predominantly still in the preclinical phase. Therefore, screening for potential genetic interactions within the cancer genome has emerged as a promising approach for drug development. Here, the two conceptual therapeutic strategies that involve the deletion or inactivation of cancer-specific TSGs are discussed. Moreover, existing approaches for screening and identifying potential gene partners are also discussed. Particularly, this review highlights the current advances of "collateral lethality" in the preclinical phase and addresses the challenges involved in translating them into therapeutic applications. This review provides insights into these strategies as new opportunities for the development of personalized antitumor therapies.
Collapse
Affiliation(s)
- Zichen Zhao
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China; Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Lingling Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China; Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Luo
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Heng Xu
- Institute of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Laboratory Medicine/Research Center of Clinical Laboratory Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Zhang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China; Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
45
|
Zhu JY, Jiang RY, Zhang HP, Fang ZR, Zhou HH, Wei Q, Wang X. Advancements in research and clinical management of interstitial lung injury associated with ADC drugs administration in breast cancer. Discov Oncol 2024; 15:843. [PMID: 39729236 DOI: 10.1007/s12672-024-01705-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024] Open
Abstract
Antibody-drug conjugates (ADCs) represent a novel class of targeted anti-tumor medications that utilize the covalent linkage between monoclonal antibodies and cytotoxic agents. This unique mechanism combines the cytotoxic potency of drugs with the targeting specificity conferred by antigen recognition. However, it is essential to recognize that many ADCs still face challenges related to off-target toxicity akin to cytotoxic payloads, as well as targeted toxicity and other potential life-threatening adverse effects, such as treatment-induced interstitial lung injury. Currently, of the four approved ADC drugs for breast cancer, several reports have documented post-treatment lung injury-related fatalities. As a result, treatment-induced interstitial lung injury due to ADC drugs has become a clinical concern. In this review article, we delve into the factors associated with ADC-induced interstitial lung injury in patients with advanced-stage breast cancer and highlight strategies expected to decrease the incidence of ADC-related interstitial lung injury in the years ahead. These efforts are directed at enhancing treatment outcomes in both advanced and early-stage cancer patients while also providing insights into the development and innovation of ADC drugs and bolstering clinicians' understanding of the diagnosis and management of ADC-associated interstitial lung injury.
Collapse
Affiliation(s)
- Jia-Yu Zhu
- Department of Graduate Student, Zhejiang Chinese Medical University, NO.548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Rui-Yuan Jiang
- Department of Graduate Student, Zhejiang Chinese Medical University, NO.548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Huan-Ping Zhang
- Department of Graduate Student, Zhejiang Chinese Medical University, NO.548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China
- Department of Graduate Student, Wenzhou Medical University, No. 270, Xueyuan West Road, Lucheng District, Wenzhou, 325027, Zhejiang, China
| | - Zi-Ru Fang
- Department of Graduate Student, Zhejiang Chinese Medical University, NO.548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Huan-Huan Zhou
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Qing Wei
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| | - Xiaojia Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
46
|
Beňačka R, Szabóová D, Guľašová Z, Hertelyová Z. Non-Coding RNAs in Breast Cancer: Diagnostic and Therapeutic Implications. Int J Mol Sci 2024; 26:127. [PMID: 39795985 PMCID: PMC11719911 DOI: 10.3390/ijms26010127] [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: 11/24/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Breast cancer (BC) is one of the most prevalent forms of cancer globally, and has recently become the leading cause of cancer-related mortality in women. BC is a heterogeneous disease comprising various histopathological and molecular subtypes with differing levels of malignancy, and each patient has an individual prognosis. Etiology and pathogenesis are complex and involve a considerable number of genetic alterations and dozens of alterations in non-coding RNA expression. Non-coding RNAs are part of an abundant family of single-stranded RNA molecules acting as key regulators in DNA replication, mRNA processing and translation, cell differentiation, growth, and overall genomic stability. In the context of breast cancer, non-coding RNAs are involved in cell cycle control and tumor cell migration and invasion, as well as treatment resistance. Alterations in non-coding RNA expression may contribute to the development and progression of breast cancer, making them promising biomarkers and targets for novel therapeutic approaches. Currently, the use of non-coding RNAs has not yet been applied to routine practice; however, their potential has been very well studied. The present review is a literature overview of current knowledge and its objective is to delineate the function of diverse classes of non-coding RNAs in breast cancer, with a particular emphasis on their potential utility as diagnostic and prognostic markers or as therapeutic targets and tools.
Collapse
Affiliation(s)
- Roman Beňačka
- Department of Pathophysiology, Medical Faculty, Pavol Jozef Šafarik University, 04011 Košice, Slovakia;
| | - Daniela Szabóová
- Department of Pathophysiology, Medical Faculty, Pavol Jozef Šafarik University, 04011 Košice, Slovakia;
| | - Zuzana Guľašová
- Center of Clinical and Preclinical Research MEDIPARK, Pavol Jozef Šafarik University, 04011 Košice, Slovakia; (Z.G.); (Z.H.)
| | - Zdenka Hertelyová
- Center of Clinical and Preclinical Research MEDIPARK, Pavol Jozef Šafarik University, 04011 Košice, Slovakia; (Z.G.); (Z.H.)
| |
Collapse
|
47
|
Hu J, Chen S, Jia H, Cai N, Hu R, Chen X, Guan Y. Cardiovascular adverse events in patients with HER2-positive breast cancer treated with trastuzumab-drug conjugates : a Bayesian disproportional real world study for signal detection leveraging the FDA Adverse Event Reporting System (FAERS). Expert Opin Drug Saf 2024:1-10. [PMID: 39675974 DOI: 10.1080/14740338.2024.2442513] [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: 06/18/2024] [Revised: 11/22/2024] [Accepted: 11/29/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND The cardiovascular risk profiles of trastuzumab drug conjugates (T-DCs) in the treatment of Human Epidermal Growth Factor Receptor 2 (HER2)-positive breast cancer are uncertain. This study analyzed real-world adverse events data from the Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS) to investigate the association between T-DCs and major adverse cardiovascular events (MACE). METHODS We searched the FAERS database for reports of cardiovascular adverse events in patients with HER2-positive breast cancer receiving trastuzumab, ado-trastuzumab emtansine (T-DM1), and trastuzumab deruxtecan (T-Dxd). The primary endpoint was MACE and the secondary endpoint was other cardiovascular events. RESULTS Between January 2015 and April 2024, out of the 135,226 drug-adverse event pairs analyzed, 8,497 were cardiovascular events, with 4,457 classified as MACE and 4,040 as other cardiovascular events. Trastuzumab reached pharmacovigilance for MACE (ROR = 1.39, 95%CI: 1.30-1.49, IC025 = 0.25), while neither T-DM1 (ROR = 0.78, 95%CI: 0.69-0.88, IC025=-0.51) nor T-Dxd (ROR = 0.63, 95%CI: 0.52-0.76, IC025=-0.93) reached MACE-related pharmacovigilance. Among MACE, stroke linked to T-DM1 (ROR = 1.86, 95%CI: 1.42-2.43, IC025 = 0.35) and T-Dxd (ROR = 2.44, 95%CI: 1.68-3.56, IC025 = 0.57) both reached pharmacovigilance in younger patients (<65 years). Proportionate analysis showed age and weight were the two key factors contributing to the occurrence of T-DCs induced MACE. CONCLUSION HER2-positive breast cancer patients receiving T-DCs require additional cardiac monitoring, particularly for stroke in younger patients.
Collapse
Affiliation(s)
- Jinfang Hu
- Guangdong Provincial Biotechnology Research Institute (Guangdong Provincial Laboratory Animals Monitoring Center), Guangzhou, Guangdong, China
| | - Siyu Chen
- Guangdong Provincial Biotechnology Research Institute (Guangdong Provincial Laboratory Animals Monitoring Center), Guangzhou, Guangdong, China
| | - Huanhuan Jia
- Guangdong Provincial Biotechnology Research Institute (Guangdong Provincial Laboratory Animals Monitoring Center), Guangzhou, Guangdong, China
| | - Nvlue Cai
- Guangdong Provincial Biotechnology Research Institute (Guangdong Provincial Laboratory Animals Monitoring Center), Guangzhou, Guangdong, China
| | - Runlong Hu
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Xi Chen
- School of Health, Brooks College (Sunnyvale), California, the United States. Department of Epidemiology and Statistics, School of Public Health, Medical College, Zhejiang University, Hangzhou, China
| | - Yezhi Guan
- Guangdong Provincial Biotechnology Research Institute (Guangdong Provincial Laboratory Animals Monitoring Center), Guangzhou, Guangdong, China
| |
Collapse
|
48
|
de Bruijn A, Schipper RJ, Voogd AC, Pullens MJJ, Bloemen JG, de Munck L, van Riet YE, Siesling S, Vriens BEP, Nieuwenhuijzen GAP. Adding Anti-HER2 Therapy to Neoadjuvant Endocrine Therapy Seems Effective in Hormone Receptor and HER2 Positive Breast Cancer Patients Unfit for Chemotherapy: A Nationwide Population-Based Cohort Study. Cancers (Basel) 2024; 16:4188. [PMID: 39766087 PMCID: PMC11674293 DOI: 10.3390/cancers16244188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/08/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Introduction: Data are lacking on the optimal neoadjuvant systemic treatment (NST) for women with hormone receptor-positive (HR+) human epidermal growth factor receptor 2-positive (HER2+) breast cancer if they are unfit to receive the combination of chemotherapy and anti-HER2 therapy. The aim of this study was to determine whether the rates of ypT0 and ypN0 differ between patients treated with neoadjuvant endocrine therapy (NET) versus NET combined with anti-HER2 therapy (NET+aHER2). Materials and Methods: Data from the Netherlands Cancer Registry were analysed to identify women diagnosed with primary HR+/HER2+ breast cancer between 2008 and 2019, treated with either NET or NET+aHER2. The ypT0 and ypN0 rates were analysed (using uni- and multivariable logistic regression analyses, as applicable) in relation to the characteristics of the patient, the tumour, and the treatment. Results: Of the 190 patients identified (median age 77), 150 had been treated with NET and 40 with NET+aHER2. Patients with clinically node-positive disease (cN+) were significantly more likely to have been treated with NET+aHER2 (p = 0.029). The ypT0 rate was significantly higher after NET+aHER2, with 10.0% (4/40) versus 1.3% (2/150) following NET (p = 0.019). The ypN0 rate was significantly higher after NET+aHER2, with 25.0% (6/24) versus 5.5% (3/55) following NET in the cN+ patients (p = 0.020) and 81.3% (13/16) versus 55.8% (53/95) after NET in the cN- patients (p = 0.047). In the cN- patients, the ypN0 status was independently associated with age (p = 0.008) and the administration of NET+aHER2 (p = 0.016). Conclusions: The rates of ypT0 and ypN0 in women with HR+/HER2+ breast cancer treated with NST was significantly higher following NET+aHER2 than after NET.
Collapse
Affiliation(s)
- Anne de Bruijn
- Department of Surgery, Catharina Hospital Eindhoven, 5623 EJ Eindhoven, The Netherlands; (R.-J.S.)
| | - Robert-Jan Schipper
- Department of Surgery, Catharina Hospital Eindhoven, 5623 EJ Eindhoven, The Netherlands; (R.-J.S.)
| | - Adri C. Voogd
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), 3511 LC Utrecht, The Netherlands
- Maastricht University Medical Center, GROW—School for Oncology and Developmental Biology, 6229 HX Maastricht, The Netherlands
| | - Marleen J. J. Pullens
- Master Advanced Nursing Practice, Faculty of People and Health, Fontys University of Applied Sciences, 5022 DM Tilburg, The Netherlands
| | - Johanne G. Bloemen
- Department of Surgery, Catharina Hospital Eindhoven, 5623 EJ Eindhoven, The Netherlands; (R.-J.S.)
| | - Linda de Munck
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), 3511 LC Utrecht, The Netherlands
| | - Yvonne E. van Riet
- Department of Surgery, Catharina Hospital Eindhoven, 5623 EJ Eindhoven, The Netherlands; (R.-J.S.)
| | - Sabine Siesling
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), 3511 LC Utrecht, The Netherlands
- Department of Health Technology and Services Research, Technical Medical Centre, University of Twente, 7522 NB Enschede, The Netherlands
| | - Birgit E. P. Vriens
- Department of Internal Medicine, Catharina Hospital Eindhoven, 5623 EJ Eindhoven, The Netherlands
| | | |
Collapse
|
49
|
Alkhatabi HA, Alatyb HN. In Silico Design of Peptide Inhibitors Targeting HER2 for Lung Cancer Therapy. Cancers (Basel) 2024; 16:3979. [PMID: 39682166 DOI: 10.3390/cancers16233979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/16/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Human epidermal growth factor receptor 2 (HER2) is overexpressed in several malignancies, such as breast, gastric, ovarian, and lung cancers, where it promotes aggressive tumor proliferation and unfavorable prognosis. Targeting HER2 has thus emerged as a crucial therapeutic strategy, particularly for HER2-positive malignancies. The present study focusses on the design and optimization of peptide inhibitors targeting HER2, utilizing machine learning to identify and enhance peptide candidates with elevated binding affinities. The aim is to provide novel therapeutic options for malignancies linked to HER2 overexpression. METHODS This study started with the extraction and structural examination of the HER2 protein, succeeded by designing the peptide sequences derived from essential interaction residues. A machine learning technique (XGBRegressor model) was employed to predict binding affinities, identifying the top 20 peptide possibilities. The candidates underwent further screening via the FreeSASA methodology and binding free energy calculations, resulting in the selection of four primary candidates (pep-17, pep-7, pep-2, and pep-15). Density functional theory (DFT) calculations were utilized to evaluate molecular and reactivity characteristics, while molecular dynamics simulations were performed to investigate inhibitory mechanisms and selectivity effects. Advanced computational methods, such as QM/MM simulations, offered more understanding of peptide-protein interactions. RESULTS Among the four principal peptides, pep-7 exhibited the most elevated DFT values (-3386.93 kcal/mol) and the maximum dipole moment (10,761.58 Debye), whereas pep-17 had the lowest DFT value (-5788.49 kcal/mol) and the minimal dipole moment (2654.25 Debye). Molecular dynamics simulations indicated that pep-7 had a steady binding free energy of -12.88 kcal/mol and consistently bound inside the HER2 pocket during a 300 ns simulation. The QM/MM simulations showed that the overall total energy of the system, which combines both QM and MM contributions, remained around -79,000 ± 400 kcal/mol, suggesting that the entire protein-peptide complex was in a stable state, with pep-7 maintaining a strong, well-integrated binding. CONCLUSIONS Pep-7 emerged as the most promising therapeutic peptide, displaying strong binding stability, favorable binding free energy, and molecular stability in HER2-overexpressing cancer models. These findings suggest pep-7 as a viable therapeutic candidate for HER2-positive cancers, offering a potential novel treatment strategy against HER2-driven malignancies.
Collapse
Affiliation(s)
- Heba Ahmed Alkhatabi
- Faculty of Applied Medical Science, King Abdulaziz University, Jeddah 22254, Saudi Arabia
- Hematology Research Unit (HRU), King Fahd Medical Research Center (KFMRC), Jeddah 22252, Saudi Arabia
- Center of Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah 22254, Saudi Arabia
| | - Hisham N Alatyb
- Center of Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah 22254, Saudi Arabia
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 22254, Saudi Arabia
| |
Collapse
|
50
|
Liu JJ, Zhang Y, Zhang SC, Liu X, Wang SN, Liu XY, Zhang J. Analysis of factors influencing the efficacy of NAC and prognosis between HER2-zero and HER2-low HR negative breast cancer. Front Cell Dev Biol 2024; 12:1417271. [PMID: 39650721 PMCID: PMC11621093 DOI: 10.3389/fcell.2024.1417271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 11/11/2024] [Indexed: 12/11/2024] Open
Abstract
Objective: The aim of this paper was to assess the differences in clinicopathological characteristics, efficacy and prognosis of neoadjuvant chemotherapy (NAC) in human epidermal growth factor receptor2(HER2)-zero and HER2-low hormone receptor (HR)-negative breast cancer (BC) patients, and the impact of HER2-evolution on prognosis before and after NAC. Methods: 319 triple negative breast cancer (TNBC) patients who completed NAC and surgery from August 2014 to August 2018 at Tianjin Medical University Cancer Institute and Hospital were included. Clinicopathological features, efficacy of NAC and assessment of prognosis were retrospectively analysed. The evolution of HER2-zero to HER2-low after NAC is defined as HER2-gain, the evolution of HER2-low to HER2-zero after NAC is defined as HER2-loss, and HER2 unchanged after NAC is defined as HER2-stable. Results: In HR-negative BC, the pathological complete response (pCR) rate was significantly higher in HER2-zero compared with HER2-low patients, and the difference was statistically significant (38.9% vs 23.2%, p = 0.004), but there was no significant difference in the prognosis between the two groups. The overall rate of HER2-evolution after NAC was 19.7%, and there was a significant correlation between HER2-loss and histological grading, whereas HER2-gain was significantly associated with Ki-67 expression. In terms of prognosis, HER2-gain was better compared to the other two groups. Conclusion: In this study, we found that HER2-low HR-negative BC showed different clinicopathological features and response to NAC compared with HER2-zero, as well as HER2-evolution before and after NAC had a significant impact on prognosis.
Collapse
Affiliation(s)
- Jing-Jing Liu
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Yi Zhang
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Shi-Chao Zhang
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Xu Liu
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Shu-Nan Wang
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Xin-Yu Liu
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Jin Zhang
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| |
Collapse
|