1
|
Attarian F, Hatamian G, Nosrati S, Akbari Oryani M, Javid H, Hashemzadeh A, Tarin M. Role of liposomes in chemoimmunotherapy of breast cancer. J Drug Target 2025; 33:887-915. [PMID: 39967479 DOI: 10.1080/1061186x.2025.2467139] [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: 11/09/2024] [Revised: 02/01/2025] [Accepted: 02/08/2025] [Indexed: 02/20/2025]
Abstract
In the dynamic arena of cancer therapeutics, chemoimmunotherapy has shown tremendous promise, especially for aggressive forms of breast cancer like triple-negative breast cancer (TNBC). This review delves into the significant role of liposomes in enhancing the effectiveness of chemoimmunotherapy by leveraging breast cancer-specific mechanisms such as the induction of immunogenic cell death (ICD), reprogramming the tumour microenvironment (TME), and enabling sequential drug release. We examine innovative dual-targeting liposomes that capitalise on tumour heterogeneity, as well as pH-sensitive formulations that offer improved control over drug delivery. Unlike prior analyses, this review directly links advancements in preclinical research-such as PAMAM dendrimer-based nanoplatforms and RGD-decorated liposomes-to clinical trial results, highlighting their potential to revolutionise TNBC treatment strategies. Additionally, we address ongoing challenges related to scalability, toxicity, and regulatory compliance, and propose future directions for personalised, immune-focused nanomedicine. This work not only synthesises the latest research but also offers a framework for translating liposomal chemoimmunotherapy from laboratory research to clinical practice.
Collapse
Affiliation(s)
- Fatemeh Attarian
- Department of Biology, Islamic Azad University, Mashhad Branch, Mashhad, Iran
| | - Ghazaleh Hatamian
- Department of Microbiology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Shamim Nosrati
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahsa Akbari Oryani
- Department of Pathology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Javid
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Hashemzadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojtaba Tarin
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
2
|
Wang A, Mizejewski GJ, Zhang C. Growth inhibitory peptides: a potential novel therapeutic approach to cancer treatment. Eur J Pharmacol 2025; 996:177554. [PMID: 40147579 DOI: 10.1016/j.ejphar.2025.177554] [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: 01/02/2025] [Revised: 03/02/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Cancer remains a major global public health concern, with considerable interest in exploring biological molecules for cancer treatment and prevention. Growth inhibitory peptide (GIP), a promising new class of biological therapeutics, has drawn attention for its distinct anti-tumor properties. Derived from human alpha-fetoprotein (HAFP), this synthetic 34-amino-acid peptide has demonstrated substantial anti-tumor effects across various cancer cell lines, effectively inhibiting tumor cell proliferation, migration, and metastasis. Studies reveal that GIP mediates its effects through a range of mechanisms, including interactions with G protein-coupled receptors (GPCRs), anti-cell adhesion activities, inhibition of cell spreading and metastatic processes, morphological alterations, platelet aggregation inhibition, immune enhancement, cell membrane disruption, ion channel blockade, and cell cycle arrest. While GIP has exhibited promising anti-tumor activity in both in vitro and in vivo models, further investigation is essential to advance its development as a therapeutic drug, particularly regarding pharmacokinetics, safety profiles, storage stability, and clinical efficacy.
Collapse
Affiliation(s)
- Aixin Wang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, PR China; Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - G J Mizejewski
- Division of Translational Medicine, Molecular Diagnostics Laboratory, Wadsworth Center, New York State Department of Health Biggs Laboratory, Empire State Plaza Albany, NY 12237, USA
| | - Chao Zhang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, PR China; Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China.
| |
Collapse
|
3
|
Gao Y, Zhang X, Ding M, Fu Z, Zhong L. Targeting "don't eat me" signal: breast cancer immunotherapy. Breast Cancer Res Treat 2025; 211:277-292. [PMID: 40100495 DOI: 10.1007/s10549-025-07659-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: 01/13/2025] [Accepted: 02/17/2025] [Indexed: 03/20/2025]
Abstract
PURPOSE Breast cancer ranks as the most prevalent cancer type impacting women globally, both in terms of incidence and mortality rates, making it a major health concern for females. There's an urgent requirement to delve into new cancer treatment methods to improve patient survival rates. METHODS Immunotherapy has gained recognition as a promising area of research in the treatment of breast cancer, with targeted immune checkpoint therapies demonstrating the potential to yield sustained clinical responses and improve overall survival rates. Presently, the predominant immune checkpoints identified on breast cancer cells include CD47, CD24, PD-L1, MHC-I, and STC-1, among others. Nevertheless, the specific roles of these various immune checkpoints in breast carcinogenesis, metastasis, and immune evasion have yet to be comprehensively elucidated. We conducted a comprehensive review of the existing literature pertaining to breast cancer and immune checkpoint inhibitors, providing a summary of findings and an outlook on future research directions. RESULTS This article reviews the advancements in research concerning each immune checkpoint in breast cancer and their contributions to immune evasion, while also synthesizing immunotherapy strategies informed by these mechanisms. Furthermore, it anticipates future research priorities, thereby providing a theoretical foundation to guide immunotherapy as a potential interventional approach for breast cancer treatment. CONCLUSION Knowledge of immune checkpoints will drive the creation of novel cancer therapies, and future breast cancer research will increasingly emphasize personalized treatments tailored to patients' specific tumor characteristics.
Collapse
Affiliation(s)
- Yue Gao
- Department of Breast Surgery, Sixth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoyan Zhang
- Department of Breast Surgery, Sixth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mingqiang Ding
- Department of Breast Surgery, Sixth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhenkun Fu
- Department of Immunology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China.
| | - Lei Zhong
- Department of Breast Surgery, Sixth Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
4
|
Zhang M, Yang Q, Lou J, Hu Y, Shi Y. A new strategy to HER2-specific antibody discovery through artificial intelligence-powered phage display screening based on the Trastuzumab framework. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167772. [PMID: 40056877 DOI: 10.1016/j.bbadis.2025.167772] [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: 11/21/2024] [Revised: 02/23/2025] [Accepted: 02/28/2025] [Indexed: 03/10/2025]
Abstract
Human epidermal growth factor receptor 2 (HER2) is a recognized drug target, and it serves as a critical target for various cancer treatments, necessitating the discovery of more antibodies for therapeutic and detection purposes. Here, we have developed an innovative workflow for antibody generation through Artificial Intelligence-powered Phage Display Screening (AIPDS). This workflow integrates artificial intelligence-driven antibody CDRH3 sequence design, high-throughput DNA synthesis and phage display screening. We applied AIPDS workflow to generate promising antibodies against the human epidermal growth factor receptor 2 (HER2), offering a template for streamlined antibody generation. Seven novel antibodies stood out, demonstrating promising efficacy in various functional assays. Notably, DYHER2-02 demonstrates strong performance across all experimental tests. In summary, our study introduces a novel methodology to generate new antibody variants of an existing antibody using an AI-assisted phage display approach. These new antibody variants hold potential applications in research, diagnosis, and therapeutic applications.
Collapse
Affiliation(s)
- Mancang Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Qiangzhen Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Jiangrong Lou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Yang Hu
- United Research Center for Next Generation DNA Synthesis of SJTU-Dynegene, Shanghai 201108, People's Republic of China
| | - Yongyong Shi
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China; Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China.
| |
Collapse
|
5
|
Ma C, Li Y, Zhu H, Li Z, Liu Y. Clinical applications of circulating tumor cell detection: challenges and strategies. Clin Chem Lab Med 2025; 63:1060-1068. [PMID: 39610299 DOI: 10.1515/cclm-2024-0959] [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: 08/18/2024] [Accepted: 11/15/2024] [Indexed: 11/30/2024]
Abstract
Circulating tumor cells (CTCs) are pivotal in the distant metastasis of tumors, serving as one of the primary materials for liquid biopsy. They hold significant clinical importance in assessing prognosis, predicting efficacy, evaluating therapeutic outcomes, and studying recurrence, metastasis, and resistance mechanisms in cancer patients. Nevertheless, the rareness and heterogeneity of CTC and the complexity of metastasis make the clinical application of CTC detection confront many challenges, which may need to be settled by some practical strategies. This article will review the content mentioned above.
Collapse
Affiliation(s)
- Chunhui Ma
- Faculty of Medical Imaging, Naval Medical University, Shanghai, China
| | - Yang Li
- Faculty of Medical Imaging, Naval Medical University, Shanghai, China
| | - Hai Zhu
- Faculty of Medical Imaging, Naval Medical University, Shanghai, China
| | - Zhiyong Li
- Faculty of Medical Imaging, Naval Medical University, Shanghai, China
| | - Yi Liu
- 26460 The Fifth Medical Center of Chinese PLA General Hospital , Beijing, China
| |
Collapse
|
6
|
Russo G, Scimone C, Palumbo L, Roscigno G, Sarracino C, Tomaiuolo I, Pisapia P, Pepe F, Rocco D, Gridelli C, Troncone G, Malapelle U. Biologics for novel driver altered non-small cell lung cancer: potential and pitfalls. Crit Rev Oncol Hematol 2025; 212:104748. [PMID: 40324663 DOI: 10.1016/j.critrevonc.2025.104748] [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: 03/06/2025] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 05/07/2025] Open
Abstract
Precision medicine has revolutionized clinical paradigm of lung cancer (LC) patients optimizing therapeutical options on the basis of molecular fingerprinting of tumor cells. The advent of the genomic era contributed to the widespread diffusion of sequencing technologies laying the basis for the approval of an increasing number of clinically relevant predictive biomarkers in clinical settings. In the rapidly evolving scenario of predictive biomarkers, mandatory testing genes demonstrated a statistically significant clinical benefit in LC patients elected to molecular tests, but emerging biomarkers are under investigation to raise the bar in the clinical management of LC patients. To date, promising IHC-based predictive biomarkers emerged as potentially integrative tools in the panel of clinically approved biomarkers. On this basis, genomic, transcriptomic and proteomic data are gaining ground toward "3D" biology" supporting the need of a multidimensional analysis of tumor cells to clinically stratify LC patients. Here we sought to overview the most promising biomarkers investigated in clinical trials to be integrated into diagnostic panel of predictive biomarkers tools for NSCLC patients.
Collapse
Affiliation(s)
- Gianluca Russo
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Claudia Scimone
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Lucia Palumbo
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Giuseppina Roscigno
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy
| | - Claudia Sarracino
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Ilaria Tomaiuolo
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Pasquale Pisapia
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Francesco Pepe
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Danilo Rocco
- Department of Pulmonary Oncology, AORN dei Colli Monaldi, Napoli, Italy
| | - Cesare Gridelli
- Division of Medical Oncology, 'S. G. Moscati' Hospital, Avellino, Italy
| | - Giancarlo Troncone
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Umberto Malapelle
- Department of Public Health, University Federico II of Naples, Naples, Italy.
| |
Collapse
|
7
|
Krishnamurthy S, Jazowski SA, Roberson ML, Reeder-Hayes K, Tang JJ, Dusetzina SB, Essien UR. Racial and Ethnic Disparities in Receipt of ERBB2-Targeted Therapy for Breast Cancer, 2010-2020. JAMA Netw Open 2025; 8:e258086. [PMID: 40310643 PMCID: PMC12046428 DOI: 10.1001/jamanetworkopen.2025.8086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/28/2025] [Indexed: 05/02/2025] Open
Abstract
Importance Among older women (aged ≥50 years) with ERBB2 (formerly HER2 or HER2/neu)-positive breast cancer, research has shown racial and ethnic disparities in access to ERBB2-targeted therapies, with Black women receiving treatment at lower rates than their White counterparts. Objective To examine racial and ethnic disparities in receipt of ERBB2-targeted therapies and changes in receipt over time. Design, Setting, and Participants This retrospective cohort study used Surveillance, Epidemiology, and End Results-Medicare linked data from January 1, 2010, to December 31, 2020. Beneficiaries who were diagnosed with ERBB2-positive breast cancer between 2010 and 2019, were aged 66 years or older at diagnosis, were continuously enrolled in Medicare Parts A and B in the 12 months before and after diagnosis, and had localized or regional stage disease at diagnosis were included. Data were analyzed from February through September 2024. Exposure Race and ethnicity defined as non-Hispanic Black or African American, Hispanic, or non-Hispanic White. Main Outcome and Measures The primary outcome was receipt of ERBB2-targeted therapies in the 12 months after diagnosis of ERBB2-positive breast cancer. Modified Poisson regression was used to evaluate the probability of receiving ERBB2-targeted therapy by race and ethnicity. Results Among 12 765 beneficiaries with ERBB2-positive breast cancer (median [IQR] age, 74 [69-80] years; 99.2% female), 8.1% were of Black, 6.9% Hispanic, and 85.0% White race and ethnicity, and 54.2% received ERBB2-targeted therapy. The overall proportion who received ERBB2-targeted therapies increased from 41.3% in 2010-2011 to 64.3% in 2018-2019. Compared with White patients, Black patients had a lower likelihood of receiving ERBB2-targeted therapies in 2010-2011 (adjusted risk ratio [ARR], 0.81; 95% confidence limit [CL], 0.68-0.97), as did Hispanic patients (ARR, 0.75; 95% CL, 0.62-0.92). Racial and ethnic disparities in receipt of ERBB2-targeted therapies narrowed over time, with no significant differences observed across racial and ethnic groups in 2018-2019 for Black patients (ARR, 0.97; 95% CL, 0.87-1.08) and Hispanic patients (ARR, 1.05; 95% CL, 0.95-1.16). Conclusions and Relevance These findings suggest a narrowing of racial and ethnic disparities in receipt of ERBB2-targeted therapies over time among older Medicare beneficiaries with ERBB2-positive breast cancer. Future research is needed to understand the practices that contributed to the narrowing of racial and ethnic disparities and to develop implementation strategies to effectively improve the quality and equity of breast cancer care.
Collapse
Affiliation(s)
- Sudarshan Krishnamurthy
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Shelley A. Jazowski
- Department of Health Policy, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Social Sciences and Health Policy, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Mya L. Roberson
- Department of Health Policy and Management, Gillings School of Global Public Health, University of North Carolina at Chapel Hill
| | - Katherine Reeder-Hayes
- Division of Oncology, Department of Medicine, UNC School of Medicine, Chapel Hill, North Carolina
| | - Jasmyn J. Tang
- Division of General Internal Medicine and Health Services Research, David Geffen School of Medicine, University of California, Los Angeles
| | - Stacie B. Dusetzina
- Department of Health Policy, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Utibe R. Essien
- Division of General Internal Medicine and Health Services Research, David Geffen School of Medicine, University of California, Los Angeles
- Center for the Study of Healthcare Innovation, Implementation and Policy, Greater Los Angeles VA Healthcare System, Los Angeles, California
- Associate Editor, JAMA Network Open
| |
Collapse
|
8
|
Lorini L, Gili R, Resteghini C, Gerosa R, Cecchi L, Gurizzan C, Zambelli A, Zucali PA, Bossi P. Precision medicine in Salivary Gland Carcinoma: Insights from breast and prostate cancer. Oral Oncol 2025; 164:107296. [PMID: 40233547 DOI: 10.1016/j.oraloncology.2025.107296] [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: 01/03/2025] [Revised: 03/31/2025] [Accepted: 04/08/2025] [Indexed: 04/17/2025]
Abstract
Salivary Gland Carcinomas (SGC) are a heterogeneous group of diseases with varied histologies, biology, clinical behaviors, and therapeutic approaches. The World Health Organization classifies SGCs into Low Aggression and High Aggression categories. Due to their rarity and unique biology, managing SGCs is challenging, often requiring expert histological diagnosis and treatment based on low-level evidence. Despite recent international guidelines, several critical aspects of SGC management remain unresolved, in particular regarding systemic treatment. Recent discoveries of molecular alterations, such as HER2 amplification, and AR overexpression, have provided diagnostic, prognostic, and predictive biomarkers for alternative treatments. While some targeted treatment have corresponding EMA-approved therapies, others do not. Treatment strategies are further complicated by synchronous alterations, such as AR-positive SGCs with concomitant HER2 amplification. Open questions remain on the optimal use of these drugs, whether in early-stage disease, post-surgery, or in palliative settings. Given the rarity of the disease and the consequent lack of high quality data in literature, it is of importance a cross-fertilization process from other, more common disease such as breast and prostate cancers. In the current narrative review we analyze current evidence on the targeted treatment on salivary gland carcinomas and shared features with breast and prostate cancer.
Collapse
Affiliation(s)
- Luigi Lorini
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Riccardo Gili
- Medical Oncology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Carlo Resteghini
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy.
| | - Riccardo Gerosa
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Luigi Cecchi
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Cristina Gurizzan
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Alberto Zambelli
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy
| | - Paolo Andrea Zucali
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy
| | - Paolo Bossi
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy
| |
Collapse
|
9
|
Chang X, Qiu X, Tong X, Gan S, Yi W, Xie S, Liu X, Zuo C, Tan W. Sortilin-Mediated Rapid, Precise and Sustained Degradation of Membrane Proteins via mRNA-Encoded Lysosome-Targeting Chimera. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2501222. [PMID: 40305781 DOI: 10.1002/advs.202501222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 03/24/2025] [Indexed: 05/02/2025]
Abstract
Recent advances in lysosome-targeting degradation technologies have introduced strategies to regulate therapeutic membrane proteins (MPs), potentially transforming treatment paradigms. However, challenges persist, including limited degradation precision due to the broad distribution of lysosome-targeting receptors (LTRs), as well as the high cost and complexity of recombinant protein production or chemical synthesis. Herein, it identifies sortilin as a promising LTR, highly expressed in malignancies but minimally present in healthy tissues outside the nervous system. Using AlphaFold-Multimer, it screened for a specific non-endogenous protein binder to sortilin and developed a modular, mRNA-encoded lysosomal targeting chimera (MedTAC) strategy, enabling rapid design and precise degradation of oncogenic MPs. In a breast cancer-bearing mouse model, a single low dose of MedTACPTK7 (0.5 mg kg-1) reduced protein tyrosine kinase-7 (PTK7) levels by up to 80% within 24 h, with sustained degradation of 44% at 72 h, demonstrating excellent pharmacokinetics. MedTACPTK7 significantly extended survival to over 50 days without systemic toxicity, compared to 20-30 days in controls. This MedTAC strategy establishes sortilin as a selective and efficient shuttle for targeted protein degradation, offering a scalable, rapidly producible platform for biochemical research and precise therapeutic applications.
Collapse
Affiliation(s)
- Xin Chang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha, Hunan, 410082, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Xinyu Qiu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China
| | - Xiaoning Tong
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Shaoju Gan
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Weicheng Yi
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Sitao Xie
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Xiangsheng Liu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Chao Zuo
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha, Hunan, 410082, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| |
Collapse
|
10
|
Xue J, Ma H, Zhang X, Wang S, Wang J, Li Z, Wu X, Yang T, Zhang C, Luo G. An Optimized Droplet Digital PCR Assay for HER2 Copy Number Variation in Breast Cancer Based on Multi-reference Genes. Appl Biochem Biotechnol 2025:10.1007/s12010-025-05233-0. [PMID: 40304992 DOI: 10.1007/s12010-025-05233-0] [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] [Accepted: 04/21/2025] [Indexed: 05/02/2025]
Abstract
Targeted therapy is essential for the 15-30% invasive breast cancer patients with human epidermal growth factor receptor 2 (HER2) over-expression. However, current HER2 diagnosing methods rely on complex manual works and highly subjective interpretations. To more accurately and objectively assess the HER2 amplification status of formalin fixed paraffin embedded (FFPE) samples, a droplet digital PCR (ddPCR) assay based on multi-reference genes was developed. We established a four-fluorescence ddPCR assay using breast cancer cell lines (T-47D and SK-BR-3) and validated it on 101 clinical breast cancer FFPE samples. Compared to clinicopathological results, the ddPCR assay based on two out of three reference genes demonstrated superior sensitivity (82.6%), specificity (98.7%), and consistency (95.0%) in determining HER2 status over assays using single or three reference genes. Whole genome sequencing of the abnormal cases further confirmed that the ddPCR assay outperformed clinical immunohistochemistry (IHC), fluorescence in situ hybridization (FISH), and quantitative PCR (qPCR) in accuracy. Our findings demonstrate that the multi-reference gene ddPCR assay significantly improves the accuracy of HER2 status detection and reduces errors associated with chromosome 17 abnormalities. This method holds promise as a complementary or alternative approach to conventional IHC and FISH testing in tissue biopsies and is also feasible for liquid biopsies.
Collapse
Affiliation(s)
- Jinbing Xue
- Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
| | - Houshi Ma
- Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
| | - Xiaoliang Zhang
- Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
| | - Shun Wang
- Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
| | - Jinxian Wang
- Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
- Suzhou ZhongKe Medical Device Industry Development Co., Ltd, Suzhou, 215163, China
| | - Zeqin Li
- Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
| | - Xi Wu
- Department of Geriatrics, The General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Tianhang Yang
- Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China.
- Jinan Guoke Medical Technology Development Co., Ltd, Jinan, 250013, China.
| | - Changsong Zhang
- Suzhou Research Center of Medical School, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215153, China.
| | - Gangyin Luo
- Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China.
- Suzhou ZhongKe Medical Device Industry Development Co., Ltd, Suzhou, 215163, China.
| |
Collapse
|
11
|
Dai W, Navolotskaia O, Fine JL, Harinath L, Motanagh SA, Villatoro TM, Bhargava R, Clark BZ, Yu J. Not All HER2-Positive Breast Cancers Are the Same: Intratumoral Heterogeneity, Low-Level HER2 Amplification, and Their Impact on Neoadjuvant Therapy Response. Mod Pathol 2025:100785. [PMID: 40311762 DOI: 10.1016/j.modpat.2025.100785] [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: 02/26/2025] [Revised: 04/15/2025] [Accepted: 04/21/2025] [Indexed: 05/03/2025]
Abstract
HER2 positive breast cancers are frequently treated with neoadjuvant anti-HER2 and chemotherapy (NACT). However, treatment response varies, with a subset of tumors showing high residual cancer burden (RCB). This study investigates the relationship between HER2 immunohistochemical (IHC) intratumoral heterogeneity (ITH), low level HER2 amplification, and tumor response to NACT. A total of 205 post-NACT HER2 positive breast carcinomas with available RCB results were classified into five HER2 groups: IHC 3+ (HER2 IHC positive, no FISH performed), Group 1-High (FISH HER2 copies >8 or HER2/CEP17 ratio >4), Group 1-Intermediate (HER2 copies > 6 to 8 or ratio > 3 to 4), Group 1-Low (HER2 copies 4 to 6 and ratio 2 to 3), and Group 3 (HER2 copies ≥ 6 and ratio < 2). Low level HER2 amplification, collectively designated as HER2 copies 4-8 or HER2/CEP17 ratio <4, was associated with reduced response to HER2-targeted therapy and higher RCB post-NACT. HER2 IHC ITH, defined as the presence of at least three distinct staining intensities with at least 10% of tumor cells exhibiting weak or no staining, was significantly more prevalent in low level HER2 amplification groups (Gp1-Int: 93.3%, Gp1-Low: 87.5%, Gp3: 80.0%) compared to high level amplification groups (IHC 3+: 24.7%, Gp1-High: 28.6%) (p<.001). Both low level HER2 amplification and HER2 IHC ITH, regardless of hormone receptor status, were independently associated with poor treatment response, and tumors demonstrating both features had the highest likelihood of low therapeutic efficiency. These findings suggest that both low level HER2 amplification and HER2 IHC ITH contribute to poor NACT response and may warrant alternative therapeutic strategies. Further prospective studies are needed to refine the clinical significance of low level HER2 amplification and IHC ITH, particularly in the context of novel HER2-targeted therapies such as antibody-drug conjugates.
Collapse
Affiliation(s)
- Wenli Dai
- Department of Pathology, University of Pittsburgh School of Medicine
| | - Olga Navolotskaia
- Department of Pathology, University of Pittsburgh School of Medicine; University of Pittsburgh Medical Center Magee-Womens Hospital, Pittsburgh, Pennsylvania
| | - Jeffrey L Fine
- Department of Pathology, University of Pittsburgh School of Medicine; University of Pittsburgh Medical Center Magee-Womens Hospital, Pittsburgh, Pennsylvania
| | - Lakshmi Harinath
- Department of Pathology, University of Pittsburgh School of Medicine; University of Pittsburgh Medical Center Magee-Womens Hospital, Pittsburgh, Pennsylvania
| | - Samaneh A Motanagh
- Department of Pathology, University of Pittsburgh School of Medicine; University of Pittsburgh Medical Center Magee-Womens Hospital, Pittsburgh, Pennsylvania
| | - Tatiana M Villatoro
- Department of Pathology, University of Pittsburgh School of Medicine; University of Pittsburgh Medical Center Magee-Womens Hospital, Pittsburgh, Pennsylvania
| | - Rohit Bhargava
- Department of Pathology, University of Pittsburgh School of Medicine; University of Pittsburgh Medical Center Magee-Womens Hospital, Pittsburgh, Pennsylvania
| | - Beth Z Clark
- Department of Pathology, University of Pittsburgh School of Medicine; University of Pittsburgh Medical Center Magee-Womens Hospital, Pittsburgh, Pennsylvania.
| | - Jing Yu
- Department of Pathology, University of Pittsburgh School of Medicine; University of Pittsburgh Medical Center Magee-Womens Hospital, Pittsburgh, Pennsylvania.
| |
Collapse
|
12
|
Maurer C, Agostinetto E, Ameye L, Lambertini M, Martel S, Ponde N, Brandão M, Poggio F, Ferreira A, Schiff R, De Angelis C, Gelber RD, Dent S, Thomssen C, Piccart M, de Azambuja E. Association of statin use on survival outcomes of patients with early-stage HER2-positive breast cancer in the APHINITY trial. Breast Cancer Res Treat 2025:10.1007/s10549-025-07699-2. [PMID: 40293644 DOI: 10.1007/s10549-025-07699-2] [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: 02/11/2025] [Accepted: 04/04/2025] [Indexed: 04/30/2025]
Abstract
PURPOSE There is evidence that statins might improve the outcome of patients with breast cancer. The role of statins in patients with early HER2-positive breast cancer is unknown. Therefore, we explored the association between statin use and survival outcomes in early HER2-positive breast cancer patients in the phase III APHINITY trial (adjuvant pertuzumab/trastuzumab). METHODS All patients (intent-to-treat population, n = 4804) were included (6.2 years median follow-up database). The primary objective was to investigate the association of statin use on invasive disease-free survival (IDFS), distant relapse-free interval (DRFI), and overall survival (OS). Patients who received statins at baseline, or started statins within 1 year from randomization were considered statin users. Survival curves were estimated using the Kaplan-Meier method. We used a Cox proportional hazards model for multivariate analysis. RESULTS Overall, 423 (8.8%) patients were classified as statin users. They were older, more often postmenopausal, had a higher body mass index, more often diabetes, hypertension, coronary heart disease and hyperlipidemia, had smaller sized tumors, were treated more often with breast conserving surgery, and less often with anthracycline-containing regimens. Overall, 508 IDFS events (12.8% among statin users and 10.4% among non-statin users) and 272 deaths (8.5% and 5.4%, respectively) occurred. In multivariate analysis, statin use was not associated with IDFS (HR, 1.11; 95% CI, 0.80-1.52), DRFI (HR, 1.21; 95% CI, 0.81-1.81) nor OS (HR, 1.16; 95% CI, 0.78-1.73). CONCLUSION In APHINITY, statin use was not associated with improved survival outcomes. These results must be interpreted with caution due to the exploratory nature of the analysis and the associated limitations.
Collapse
Affiliation(s)
- Christian Maurer
- National Center for Tumor Diseases (NCT) Heidelberg, University Hospital and German Cancer Research Center Heidelberg, Im Neuenheimer Feld 460, 69120, Heidelberg, Germany.
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
| | - Elisa Agostinetto
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Rue Meylemeersch 90, 1070, Brussels, Belgium
| | - Lieveke Ameye
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Rue Meylemeersch 90, 1070, Brussels, Belgium
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa, Italy
- Department of Medical Oncology, U.O. Clinical Di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Samuel Martel
- Specialised Medicine Department, CISSS Montérégie-Centre/Hôpital Charles-Le Moyne, Greenfield Park, Québec, Canada
- Université of Sherbrooke, Sherbrooke, Québec, Canada
| | - Noam Ponde
- Clinical Development Department, Daiichi Sankyo, Morristown, NJ, USA
| | - Mariana Brandão
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Rue Meylemeersch 90, 1070, Brussels, Belgium
| | - Francesca Poggio
- Department of Medical Oncology, U.O. Clinical Di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Arlindo Ferreira
- Católica Medical School, Universidade Católica Portuguesa, Lisbon, Portugal
| | - Rachel Schiff
- Lester and Sue Smith Breast Center, Departments of Medicine and of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Carmine De Angelis
- Medical Oncology Unit, Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Richard D Gelber
- Harvard Medical School, Harvard TH Chan School of Public Health, Dana-Farber Cancer Institute, Frontier Science Foundation, Boston, MA, USA
| | - Susan Dent
- Wilmot Cancer Institute, Department of Medicine, University of Rochester, Rochester, NY, USA
| | - Christoph Thomssen
- Department of Gynaecology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Martine Piccart
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Rue Meylemeersch 90, 1070, Brussels, Belgium
| | - Evandro de Azambuja
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Rue Meylemeersch 90, 1070, Brussels, Belgium
| |
Collapse
|
13
|
Röder J, Alekseeva T, Kiefer A, Kühnel I, Prüfer M, Zhang C, Bodden M, Rosigkeit S, Waldmann A, Tonn T, Bockamp E, Stein S, Wels WS. ErbB2/HER2-targeted CAR-NK cells eliminate breast cancer cells in an organoid model that recapitulates tumor progression. Mol Ther 2025:S1525-0016(25)00312-0. [PMID: 40285353 DOI: 10.1016/j.ymthe.2025.04.033] [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: 12/12/2024] [Revised: 03/21/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025] Open
Abstract
Chimeric antigen receptor-engineered NK cells hold promise for adoptive cancer immunotherapy. In one such approach, the ErbB2 (HER2)-specific CAR-NK cell line NK-92/5.28.z is under investigation as an off-the-shelf therapy in a phase I trial in glioblastoma patients. To evaluate activity of NK-92/5.28.z cells against ErbB2-positive breast cancer, here we developed an organoid model derived from CKP mice that allows conditional activation of oncogenic driver mutations. Expression of ErbB2 and Cre recombinase in CKP mammary epithelial cells induced malignant transformation, with the resulting EC-CKP cells characterized by neoplastic morphology, loss of p53, and constitutive activation of the MAP kinase pathway. NK-92/5.28.z cells demonstrated potent CAR-mediated cytotoxicity against EC-CKP organoids, with tumor cell lysis dependent on exposure time and organoid size. In vivo passaging of EC-CKP organoids revealed cellular plasticity and induced an EMT phenotype associated with increased resistance to standard therapies. Importantly, NK-92/5.28.z cells retained high and specific cytotoxicity against these breast cancer cells in vitro and in an aggressive organoid-based in vivo mouse model that reflects advanced-stage disease. Our data highlight the therapeutic potential of NK-92/5.28.z cells against ErbB2-positive breast cancer, supporting their further development toward clinical application.
Collapse
Affiliation(s)
- Jasmin Röder
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt, Germany; Frankfurt Cancer Institute, Goethe University, 60596 Frankfurt, Germany
| | - Tijna Alekseeva
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt, Germany
| | - Anne Kiefer
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt, Germany
| | - Ines Kühnel
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt, Germany
| | - Maren Prüfer
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt, Germany
| | - Congcong Zhang
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt, Germany
| | - Malena Bodden
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt, Germany
| | - Sebastian Rosigkeit
- Institute of Translational Immunology and Research Centre for Immunotherapy, Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Anja Waldmann
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt, Germany
| | - Torsten Tonn
- Frankfurt Cancer Institute, Goethe University, 60596 Frankfurt, Germany; Institute for Transfusion Medicine and Immunohematology, Goethe University, Frankfurt and Red Cross Blood Donation Service Baden-Württemberg-Hessen, 60528 Frankfurt, Germany
| | - Ernesto Bockamp
- Institute of Translational Immunology and Research Centre for Immunotherapy, Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Stefan Stein
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt, Germany
| | - Winfried S Wels
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt, Germany; Frankfurt Cancer Institute, Goethe University, 60596 Frankfurt, Germany; German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, 60596 Frankfurt, Germany.
| |
Collapse
|
14
|
He Y, Liu Q, Luo Z, Hu Q, Wang L, Guo Z. Role of Tumor-Associated Macrophages in Breast Cancer Immunotherapy. FRONT BIOSCI-LANDMRK 2025; 30:26995. [PMID: 40302326 DOI: 10.31083/fbl26995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/04/2024] [Accepted: 12/11/2024] [Indexed: 05/02/2025]
Abstract
Breast cancer (BC) is the second leading cause of death among women worldwide. Immunotherapy has become an effective treatment for BC patients due to the rapid development of medical technology. Considerable breakthroughs have been made in research, marking the beginning of a new era in cancer treatment. Among them, various cancer immunotherapies such as immune checkpoint inhibitors (ICIs), cancer vaccines, and adoptive cell transfer are effective and have good prospects. The tumor microenvironment (TME) plays a crucial role in determining the outcomes of tumor immunotherapy. Tumor-associated macrophages (TAMs) are a key component of the TME, with an immunomodulatory effect closely related to the immune evasion of tumor cells, thereby affecting malignant progression. TAMs also significantly affect the therapeutic effect of ICIs (such as programmed death 1/programmed death ligand 1 (PD-1/PD-L1) inhibitors). TAMs are composed of multiple heterogeneous subpopulations, including M1 phenotypes macrophages (M1) and M2 phenotypes macrophages (M2). Furthermore, they mainly play an M2-like role and moderate a variety of harmful consequences such as angiogenesis, immunosuppression, and metastasis. Therefore, TAMs have become a key area of focus in the development of tumor therapies. However, several tumor immunotherapy studies demonstrated that ICIs are effective only in a small number of solid cancers, and tumor immunotherapy still faces relevant challenges in the treatment of solid tumors. This review explores the role of TAMs in BC immunotherapy, summarizing their involvement in BC development. It also explains the classification and functions of TAMs, outlines current tumor immunotherapy approaches and combination therapies, and discusses the challenges and potential strategies for TAMs in immuno-oncology treatments.
Collapse
Affiliation(s)
- Yan He
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, 421001 Hengyang, Hunan, China
| | - Quan Liu
- Department of Laboratory Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen University, 518052 Shenzhen, Guangdong, China
| | - Zhihao Luo
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, 421001 Hengyang, Hunan, China
| | - Qian Hu
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, 421001 Hengyang, Hunan, China
| | - Li Wang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, 421001 Hengyang, Hunan, China
| | - Zifen Guo
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, 421001 Hengyang, Hunan, China
| |
Collapse
|
15
|
Zhou G, Zhang Y, Gu Y, Cao B, Fan X, Yu D, Fu S, Liu M, Assaraf YG, Li Y, Ouyang D, Zhao Q. Enhanced antibody-mediated cellular cytotoxicity of germline-like anti-HER2 antibodies through a point mutation in complementarity-determining regions. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf063. [PMID: 40267400 DOI: 10.1093/jimmun/vkaf063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 02/24/2025] [Indexed: 04/25/2025]
Abstract
Human epidermal growth factor receptor 2 (HER2) is highly overexpressed on the surface of breast cancer cells, presenting an attractive target for therapeutic antibodies. Antibody-dependent cellular cytotoxicity (ADCC) is a central mechanism underlying the antitumor activities of anti-HER2 monoclonal antibodies. Here, we engineered Ab5, a germline-like anti-HER2 monoclonal antibody derived from a naïve human antibody library. Our aim was to enhance the ADCC properties of Ab5 by affinity maturation. Through an in silico aided mutagenesis analysis, we identified an Ab5 mutant termed Ab5m, exhibiting enhanced affinity compared with the parental Ab5. Computational modeling predicted that a crucial interacting residue, aspartic acid 31 on the complementarity-determining region 1 of the heavy chain involved in the important charged interactions with HER2 domain II. Substitution of aspartic acid with glutamic acid decreased the interaction energies, resulting in a remarkable affinity enhancement of Ab5m-scFv (KD = 0.2 nM) compared with parental Ab5-scFv (KD = 1.5 nM). This affinity maturation translated into the obvious improvement in ADCC and notable enhancement of tumor ablation in vivo, either alone or in combination with anti-B7-H3 antibodies. These findings suggest that the potential of affinity optimization as a strategy to enhance the ADCC properties of human germline antibodies.
Collapse
Affiliation(s)
- Guangyu Zhou
- MoE Frontiers Science Center for Precision Oncology, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Yunsen Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Yuheng Gu
- MoE Frontiers Science Center for Precision Oncology, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Bihui Cao
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xin Fan
- MoE Frontiers Science Center for Precision Oncology, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Di Yu
- MoE Frontiers Science Center for Precision Oncology, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Shengyu Fu
- MoE Frontiers Science Center for Precision Oncology, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Manting Liu
- Department of Radiology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yehuda G Assaraf
- Fred Wyszkowski Cancer Research Laboratory, Faculty of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Yaping Li
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Defang Ouyang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qi Zhao
- MoE Frontiers Science Center for Precision Oncology, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| |
Collapse
|
16
|
Zhao X, Li Y, Zhang H, Cai Y, Wang X, Liu Y, Li T, Xu C, Teng Y, Li D, Li F. PAK5 promotes the trastuzumab resistance by increasing HER2 nuclear accumulation in HER2-positive breast cancer. Cell Death Dis 2025; 16:323. [PMID: 40258843 PMCID: PMC12012021 DOI: 10.1038/s41419-025-07657-2] [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: 11/08/2024] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 04/23/2025]
Abstract
Nuclear HER2 (N-HER2) predicts resistance to HER2-targeted therapy and poor prognosis of breast cancer patients, and the underlying mechanisms remain unclear. Here, we show that high expression of p21-activated kinase 5 (PAK5) is associated with HER2-targeted therapy resistance and poor outcomes of breast cancer patients. Excitingly, we find an increase in N-HER2 protein expression in patients with high PAK5 expression, who demonstrate resistance to trastuzumab treatment. PAK5 phosphorylates methyltransferase METTL14 on serine 399 to enhance m6A modification of lncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), leading to increased MALAT1 stability. The stabilized MALAT1 inhibits ubiquitin-proteasomal degradation of the N-HER2 by affecting the interaction of deubiquitinase USP8 and N-HER2, thereby promoting N-HER2 accumulation. Moreover, HER2 upregulates the expression of PAK5 and MALAT1, activating the HER2-MALAT1 positive feedback loop. Importantly, PAK5 promotes the therapeutic resistance of HER2-positive breast cancer cells by increasing N-HER2 protein both in vitro and vivo. These findings highlight PAK5 as a therapeutic target for combating trastuzumab resistance in HER2-positive breast cancer.
Collapse
MESH Headings
- Humans
- p21-Activated Kinases/metabolism
- p21-Activated Kinases/genetics
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Breast Neoplasms/metabolism
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-2/genetics
- Trastuzumab/pharmacology
- Trastuzumab/therapeutic use
- Female
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- RNA, Long Noncoding/metabolism
- RNA, Long Noncoding/genetics
- Animals
- Cell Line, Tumor
- Mice
- Mice, Nude
- Gene Expression Regulation, Neoplastic/drug effects
- Cell Nucleus/metabolism
- Cell Nucleus/drug effects
- Mice, Inbred BALB C
Collapse
Affiliation(s)
- Xin Zhao
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, China
- Department of Pediatric Orthopaedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yang Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, China.
| | - Hongyan Zhang
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, China
| | - Yihang Cai
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, China
| | - Xu Wang
- Department of Breast Surgery, Department of Surgical Oncology, Research Unit of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yidu Liu
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, China
| | - Tingting Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, China
| | - Chendong Xu
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, China
| | - Yuee Teng
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.
| | - Danni Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.
| | - Feng Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
17
|
Du J, Shi J, Sun D, Wang Y, Liu G, Chen J, Wang W, Zhou W, Zheng Y, Wu H. Machine learning prediction of HER2-low expression in breast cancers based on hematoxylin-eosin-stained slides. Breast Cancer Res 2025; 27:57. [PMID: 40251691 PMCID: PMC12008878 DOI: 10.1186/s13058-025-01998-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/10/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Treatment with HER2-targeted therapies is recommended for HER2-positive breast cancer patients with HER2 gene amplification or protein overexpression. Interestingly, recent clinical trials of novel HER2-targeted therapies demonstrated promising efficacy in HER2-low breast cancers, raising the prospect of including a HER2-low category (immunohistochemistry, IHC) score of 1 + or 2 + with non-amplified in-situ hybridization for HER2-targeted treatments, which necessitated the accurate detection and evaluation of HER2 expression in tumors. Traditionally, HER2 protein levels are routinely assessed by IHC in clinical practice, which not only requires significant time consumption and financial investment but is also technically challenging for many basic hospitals in developing countries. Therefore, directly predicting HER2 expression by hematoxylin-eosin (HE) staining should be of significant clinical values, and machine learning may be a potent technology to achieve this goal. METHODS In this study, we developed an artificial intelligence (AI) classification model using whole slide image of HE-stained slides to automatically assess HER2 status. RESULTS A publicly available TCGA-BRCA dataset and an in-house USTC-BC dataset were applied to evaluate our AI model and the state-of-the-art method SlideGraph + in terms of accuracy (ACC), the area under the receiver operating characteristic curve (AUC), and F1 score. Overall, our AI model achieved the superior performance in HER2 scoring in both datasets with AUC of 0.795 ± 0.028 and 0.688 ± 0.008 on the USCT-BC and TCGA-BRCA datasets, respectively. In addition, we visualized the results generated from our AI model by attention heatmaps, which proved that our AI model had strong interpretability. CONCLUSION Our AI model is able to directly predict HER2 expression through HE images with strong interpretability, and has a better ACC particularly in HER2-low breast cancers, which provides a method for AI evaluation of HER2 status and helps to perform HER2 evaluation economically and efficiently. It has the potential to assist pathologists to improve diagnosis and assess biomarkers for companion diagnostics.
Collapse
Affiliation(s)
- Jun Du
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China
- Intelligent Pathology Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China
| | - Jun Shi
- School of Software, Hefei University of Technology, Hefei, 230601, China
| | - Dongdong Sun
- School of Computer Science and Information Engineering, Hefei University of Technology, Hefei, 230601, Anhui, China
| | - Yifei Wang
- School of Computer Science and Information Engineering, Hefei University of Technology, Hefei, 230601, Anhui, China
| | - Guanfeng Liu
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Jingru Chen
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Wei Wang
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China
- Intelligent Pathology Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China
| | - Wenchao Zhou
- Intelligent Pathology Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China.
- Department of Pathology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Yushan Zheng
- School of Engineering Medicine, Beijing Advanced Innovation Center on Biomedical Engineering, Beihang University, Beijing, 100191, China.
| | - Haibo Wu
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China.
- Intelligent Pathology Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China.
- Department of Pathology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| |
Collapse
|
18
|
Zhao Q, Wu J, Yao J, Liu Z, Li C, Sun Q. Simultaneously induce apoptosis and inhibiting metastasis of Triple negative breast cancer Enabled by a highly potent NIR fluorescent inhibitor. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 339:126183. [PMID: 40294573 DOI: 10.1016/j.saa.2025.126183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 03/31/2025] [Accepted: 04/05/2025] [Indexed: 04/30/2025]
Abstract
Triple-negative breast cancer (TNBC) is a cancerous tumor that poses a global threat to women's lives and health. Currently, chemotherapy is the preferred treatment for advanced TNBC. In this work, we have identified a novel NIR fluorescent inhibitor, SWMU690, which can effectively inhibit the proliferation of TNBC at low doses. The half-maximal inhibitory concentration (IC50) values of SWMU690 against the MDA-MB-231, 4 T1, and T47D cell lines were determined to be 104 nM, 509.4 nM, and 206 nM, respectively. We found that SWMU690 specifically localized in mitochondria to trigger mitochondria-mediated intrinsic apoptotic pathways. Furthermore, SWMU690 can suppress the production of ATP to inhibit TNBC metastasis. This study demonstrated the advantage of disrupting the function of mitochondria in inhibiting the proliferation and metastasis of TNBC, and may have other applications in the treatment of other malignant tumors.
Collapse
Affiliation(s)
- Qixin Zhao
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000 Sichuan, China.
| | - Jiao Wu
- Chengdu University of Traditional Chinese Medicine, Chengdu 610000 Sichuan, China.
| | - Jiale Yao
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000 Sichuan, China.
| | - Zengjin Liu
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000 Sichuan, China; Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou 646000 Sichuan, China.
| | - Changqiang Li
- Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China.
| | - Qin Sun
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000 Sichuan, China; Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou 646000 Sichuan, China.
| |
Collapse
|
19
|
Chen B, Huang L, Gui M, Torres-de la Roche LA, De Wilde RL, Shi W, Liu H, Gong Z. High Expression of Complement 3 Enhances the Efficacy of Neoadjuvant Chemotherapy Prior to Oncoplastic Surgery for HER2-Positive Breast Cancer. Cancer Biother Radiopharm 2025. [PMID: 40242863 DOI: 10.1089/cbr.2025.0038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025] Open
Abstract
Background: Neoadjuvant chemotherapy for breast cancer (BC) improves patient prognosis, but its efficacy is hindered by the disease's high heterogeneity. This study enhances effectiveness of targeted therapy to improve clinical outcomes. Methods: This study enrolled 335 patients from three centers. Differentially expressed genes were identified using DESeq2, and Venn analysis was applied to identify hub Complement genes. Hub gene expression was validated through public databases and IHC in real-world samples. In addition, associations between these genes and clinical factors were evaluated. Survival analysis, using the log-rank test, assessed overall survival (OS), disease-specific survival (DSS), and progression-free interval (PFI) as end points. The authors also locate hub Complement 3 gene position by immunofluorescence. Results: The study identified C3 as a hub Complement gene associated with trastuzumab sensitivity. C3 shows higher expression in normal than tumor tissues. C3 was highly expressed in HER2-negative and early-stage BC, but showed no differences in lymph node or metastasis subgroups. High C3 expression correlated with better OS, DSS, and PFI, particularly in HER2+ patients. IHC analysis confirmed higher C3 expression in normal tissues with the lowest in triple-negative BC. Immunofluorescence findings suggest that C3 recruits complement receptor 2 to enhance trastuzumab efficacy in HER2+ patients. Conclusions: This finding highlights the potential of complement 3 to improve therapeutic outcomes and pave the way for more personalized treatment strategies in BC.
Collapse
Affiliation(s)
- Bo Chen
- Department of Medical Aesthetic Surgery, The First Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Lifen Huang
- Clinicopathological Diagnosis & Research Center, the Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- Key Laboratory of Tumor Molecular Pathology of Guangxi Higher Education Institutes, Baise, China
| | - Morui Gui
- Department of Medical Aesthetic Surgery, The First Affiliated Hospital of Guilin Medical University, Guilin, China
| | | | - Rudy Leon De Wilde
- Pius-Hospital, University Hospital for Gynecology, University Medicine Oldenburg, Oldenburg, Germany
| | - Wenjie Shi
- Medical Faculty and University Hospital Magdeburg, Molecular and Experimental Surgery, Clinic for General, Visceral, Vascular and Transplantation Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | - Hui Liu
- School of Public Health, Guilin Medical University, Guilin, China
| | - Zhenyu Gong
- Department of Medical Aesthetic Surgery, The First Affiliated Hospital of Guilin Medical University, Guilin, China
| |
Collapse
|
20
|
Freyer G, Rigault C, Guéroult Accolas L, Barea A, Radu N, Ouamer A, Saghatchian M. HERmione: Understanding the Needs of Patients Living with Metastatic HER2-Positive Breast Cancer Through a Cross-Sectional Survey in Parallel with Patients and Oncologists. Cancers (Basel) 2025; 17:1349. [PMID: 40282524 PMCID: PMC12025477 DOI: 10.3390/cancers17081349] [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: 01/16/2025] [Revised: 03/04/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES HER2-positive metastatic breast cancer continues to have a significant impact on patients' lives. The HERmione project was conducted in France to identify patients' needs for support and information, understand services offered, and identify differences in the perception of burden between patients and oncologists. METHODS Between July and October 2022, 273 patients with HER2-positive metastatic breast cancer and 40 oncologists were surveyed. The mean age of patients was 52 years, with most receiving treatment at specialized cancer centers (38%) or other public hospitals (34%). RESULTS The survey revealed a substantial burden of the disease and treatment in patients, perceptions that differed from those of oncologists. Both the physical and mental well-being of the patients were below average. Despite the burden of the disease, patients lacked access to many types of support, particularly support with sexual well-being. Additionally, 60% of patients did not have access to nursing support. Patients had high expectations regarding access to information but often did not know where to access this information. Despite this, they still exhibited treatment preferences. CONCLUSIONS These findings suggest that enhanced communication is critical to ensure that patients receive adequate support. Nursing support could improve patient-oncologist communication and thereby enhance patient well-being. Finally, to meet patient expectations regarding information access, a broader array of support tools should be offered.
Collapse
Affiliation(s)
- Gilles Freyer
- CHU Lyon-Sud, Institut de Cancérologie et Université de Lyon, 69002 Lyon, France
| | | | | | | | | | | | - Mahasti Saghatchian
- Hôpital Américain de Paris, Neuilly sur Seine, 92200 Neuilly-sur-Seine, France
| |
Collapse
|
21
|
Cheng TC, Huang BM, Liao YC, Chang HS, Tu SH, Ho YS, Chen LC. Fibroblast growth factor receptor four inhibitor FGF401 improves the efficacy of trastuzumab in FGFR4-overexpressing breast cancer cells. Int J Cancer 2025; 156:1606-1620. [PMID: 39679940 DOI: 10.1002/ijc.35271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 09/27/2024] [Accepted: 10/24/2024] [Indexed: 12/17/2024]
Abstract
Breast cancer is the most common cancer among women. Among them, human epidermal growth factor receptor-positive (HER2+) breast cancer is more malignant. Fortunately, many anti-HER2 drugs are currently used in clinical treatments to increase patient survival. However, some HER2+ patients (~15%) still develop drug resistance after receiving trastuzumab treatment, leading to treatment failure. Using CCLE and METABRIC database analyses, we found that fibroblast growth factor receptor 4 (FGFR4) mRNA was highly detected in tumors from HER2+ breast cancer patients (p < .001) and was associated with poorer survival in breast cancer patients. Through retrospective immunohistochemical staining analysis, we detected higher expression of FGFR4 protein in breast cancer tissues collected from patients who were resistant to trastuzumab therapy compared with breast cancer patients who responded to treatment. An FGFR4 inhibitor (FGF401) effectively inhibits tumor growth in trastuzumab-insensitive patient-derived xenograft (PDX) tumor-bearing mice. For molecular mechanism studies, we demonstrated that HER2/FGFR4 protein complexes were detected on the cell membrane of the tumor tissues in these trastuzumab-insensitive PDX tumor tissues. After trastuzumab treatment in these drug-resistant breast cancer cells, FGFR4 translocates and enters the nucleus. However, trastuzumab-induced nuclear translocation of FGFR4/HER2-intracellular domain protein complex in trastuzumab-resistant cancer cells is blocked by FGF401 treatment. We believe that FGFR4 overexpression and complex formation with HER2 can serve as molecular markers to assist clinicians in identifying trastuzumab-resistant tumors. Our results suggest that FGF401 combined with trastuzumab as adjuvant therapy for patients with trastuzumab-resistant breast cancer may be a potential new treatment strategy.
Collapse
Affiliation(s)
- Tzu-Chun Cheng
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, China Medical University, Taichung, Taiwan
| | - Bu-Miin Huang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - You-Cheng Liao
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Han-Sheng Chang
- Department of Biological Science & Technology, College of Life Sciences, China Medical University, Taichung, Taiwan
| | - Shih-Hsin Tu
- Department of Surgery, Taipei Medical University Hospital, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yuan-Soon Ho
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, China Medical University, Taichung, Taiwan
| | - Li-Ching Chen
- Department of Biological Science & Technology, College of Life Sciences, China Medical University, Taichung, Taiwan
| |
Collapse
|
22
|
Rizk M, Mokbel K. Letter re: A critical appraisal of the DATA trial analysis on the prognostic and predictive value of the luminal-like subtype. ESMO Open 2025; 10:105067. [PMID: 40239368 PMCID: PMC12020829 DOI: 10.1016/j.esmoop.2025.105067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Affiliation(s)
- M Rizk
- The London Breast Institute, Princess Grace Hospital, London, UK.
| | - K Mokbel
- The London Breast Institute, Princess Grace Hospital, London, UK
| |
Collapse
|
23
|
Akay S, Emiroglu M, Kelten Talu C, Unal OU. Comparison of Clinicopathological Characteristics for HER2-Null, HER2-Ultralow and HER2-Low Breast Cancer: A Single-Center Study. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:719. [PMID: 40283010 PMCID: PMC12028604 DOI: 10.3390/medicina61040719] [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: 03/16/2025] [Revised: 04/04/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025]
Abstract
Background and Objectives: A recent clinical trial has demonstrated that breast cancer with low-HER2 expression levels responds to trastuzumab deruxtecan treatment. This has prompted a re-evaluation of HER2-targeted therapies in the HER2-negative group. Further research is required in the form of more detailed information about HER2-negative breast cancers with HER2-null, HER2-ultralow, and HER2-low subgroups. This study represents a novel approach to this field. Materials and Methods: HER2-negative breast cancer patients were classified into three groups as HER2-null, HER2-ultralow, and HER2-low. A comparison of clinicopathological features was analyzed retrospectively. Results: Of 722 patients, 22.3% were HER2-null, 23.7% were HER2-ultralow, 54.0% were HER2-low. While two-thirds of all the patients were evaluated as having T2 tumors, T4 tumors constituted 2.4%. Among HER2-negative cases, 11.8% were triple-negative and 88.2% were hormone-positive. The mean tumor diameter was 0.57 cm larger in the HER2-ultralow group than in the HER2-null group and 0.34 cm larger in the HER2-low group than in the HER2-null group. HER2-null tumors tend to be smaller. The HER2-low group was more likely to relapse than the HER2-null group. There were no significant differences in the distribution of hormone positivity or negativity (TNBC) among the groups; they accounted for 89.2% and 10.8% of all cases, respectively. Conclusions: HER2-negative breast cancer is a heterogeneous disease and deserves a detailed review in terms of diagnosis and treatment. HER2-ultralow tumors are larger in size and have a prognosis comparable to HER2-null tumors. HER2-low tumors tend to recur much more frequently and with poorer outcomes. In this field, new therapeutic approaches may result in better outcomes.
Collapse
Affiliation(s)
- Seval Akay
- Department of Medical Oncology, Izmir City Hospital, Izmir 35540, Turkey
| | - Mumin Emiroglu
- Department of Pathology, Izmir Tepecik Education and Research Hospital, Izmir 35020, Turkey;
| | - Canan Kelten Talu
- Department of Pathology, UHS Izmir Faculty of Medicine, Izmir Tepecik Education and Research Hospital, Izmir 35020, Turkey;
| | - Olcun Umit Unal
- Department of Medical Oncology, UHS Faculty of Medicine, Izmir City Hospital, Izmir 35540, Turkey;
| |
Collapse
|
24
|
Chen L, Chow A, Ma W, Coker C, Gu Y, Canoll P, Kandpal M, Hibshoosh H, Biswas AK, Acharyya S. A new, immunocompetent brain-metastatic mouse model of HER2-positive breast cancer. Clin Exp Metastasis 2025; 42:25. [PMID: 40220135 DOI: 10.1007/s10585-025-10343-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 03/26/2025] [Indexed: 04/14/2025]
Abstract
Brain metastasis is a common and devastating complication of cancer that affects over 50% of HER2-positive (HER2+) breast cancer patients. The lack of effective long-term treatment options for brain metastasis significantly increases morbidity and mortality among these patients. Therefore, understanding the underlying mechanisms that drive brain metastasis is critically important for developing new strategies to treat it effectively. Genetically engineered mouse models (GEMMs) of HER2+ breast cancer have been instrumental in understanding the development and progression of HER2+ breast cancer. However, the GEMM models for HER2+ breast cancer do not develop brain metastasis and are not suitable for the study of brain metastasis. We therefore developed a fully immunocompetent mouse model of experimental brain metastasis in HER2+ breast cancer by injecting a murine HER2/neu-expressing mammary-tumor-cell line into the arterial circulation of syngeneic FVB/N mice followed by isolation of brain-metastatic derivatives through in-vivo selection. By this in-vivo serial passaging process, we selected highly brain-metastatic (BrM) derivatives known as neu-BrM. Notably, after intracardiac injection, neu-BrM cells generated brain metastasis in 100% of the mice, allowing us to study the later stages of metastatic progression, including cancer-cell extravasation and outgrowth in the brain. Analogous to human brain metastasis, we observed reactive gliosis and significant immune infiltration in the brain tissue of mice injected with neu-BrM cells. We further confirmed that brain-metastatic lesions in the neu-BrM model express HER2. Consistently, we found that the brain-metastatic burden in these mice can be significantly reduced but not eliminated with tucatinib, an FDA-approved, blood-brain-barrier-penetrant HER2 inhibitor. Therefore, the neu-BrM HER2+ breast cancer model can be used to investigate the roles of innate and adaptive immune-system components during brain-metastatic progression and the mechanisms of HER2-therapy response and resistance.
Collapse
Affiliation(s)
- Leran Chen
- Institute for Cancer Genetics, Columbia University Irving Medical Center, 1130 St Nicholas Avenue, New York, NY, 10032, USA
| | - Angela Chow
- Institute for Cancer Genetics, Columbia University Irving Medical Center, 1130 St Nicholas Avenue, New York, NY, 10032, USA
| | - Wanchao Ma
- Institute for Cancer Genetics, Columbia University Irving Medical Center, 1130 St Nicholas Avenue, New York, NY, 10032, USA
| | - Courtney Coker
- Institute for Cancer Genetics, Columbia University Irving Medical Center, 1130 St Nicholas Avenue, New York, NY, 10032, USA
| | - Yifan Gu
- Institute for Cancer Genetics, Columbia University Irving Medical Center, 1130 St Nicholas Avenue, New York, NY, 10032, USA
- , 111 Biological Science Building, 484 W, 12th Avenue, Columbus, OH, 43210, USA
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 630 W 168th St, New York, NY, 10032, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 St. Nicholas Ave, New York, NY, 10032, USA
| | - Manoj Kandpal
- Center for Clinical and Translational Science, Rockefeller University Hospital, 1230 York Ave, New York, NY, 10065, USA
| | - Hanina Hibshoosh
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 630 W 168th St, New York, NY, 10032, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 St. Nicholas Ave, New York, NY, 10032, USA
| | - Anup K Biswas
- Institute for Cancer Genetics, Columbia University Irving Medical Center, 1130 St Nicholas Avenue, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 630 W 168th St, New York, NY, 10032, USA
| | - Swarnali Acharyya
- Institute for Cancer Genetics, Columbia University Irving Medical Center, 1130 St Nicholas Avenue, New York, NY, 10032, USA.
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 630 W 168th St, New York, NY, 10032, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 St. Nicholas Ave, New York, NY, 10032, USA.
| |
Collapse
|
25
|
Chai S, Cui J, Sun Y, Wang X, Cai C. Exploring Novel Therapeutic Targets in Breast Cancer via Comprehensive Omics Profiling and Experimental Verification. BIOLOGY 2025; 14:405. [PMID: 40282270 PMCID: PMC12025194 DOI: 10.3390/biology14040405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Breast cancer is the leading cause of cancer-related deaths among women worldwide. Deciphering the molecular mechanisms of breast cancer is crucial for developing targeted therapeutic approaches. METHODS This study analyzed gene expression profiles from the Gene Expression Omnibus (GEO) database to identify differentially expressed genes (DEGs) in breast cancer. Mendelian randomization (MR) analysis was then employed using publicly available eQTL databases to evaluate potential causal relationships between these DEGs and breast cancer. Enrichment analyses were further conducted to explore their functional significance. Furthermore, external validation of co-expressed genes was conducted using The Cancer Genome Atlas (TCGA) database. In vitro functional assays and drug sensitivity analyses were performed on selected target genes to validate their roles in breast cancer pathogenesis and treatment. RESULTS A total of 1052 upregulated and 1380 downregulated genes were identified in breast cancer. Additionally, MR analysis revealed 12 significant co-expressed genes potentially contributing to breast cancer pathogenesis. These genes were primarily enriched in lipid metabolism and immune responses via regulating microRNA functions and AMPK signaling. Validation through the TCGA database confirmed differential expression of these genes in breast cancer tissues. Strikingly, functional assays of the less-reported genes DNASE2 and ATOH8 demonstrated their involvement in breast cancer pathogenesis through modulating proliferation, migration, and invasion of cancer cells. Notably, several commonly used clinical drugs for breast cancer management, such as 5-Fluorouracil, exhibited dramatically increased sensitivity to DNASE2 and ATOH8 expression. CONCLUSIONS Our study provides novel insights into the molecular basis of breast cancer pathogenesis and identifies promising therapeutic strategies for this condition.
Collapse
Affiliation(s)
- Shengjun Chai
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining 810008, China; (S.C.); (J.C.); (Y.S.)
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University Medical College, Xining 810008, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Qinghai University Medical College, Xining 810008, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University Medical College, Xining 810008, China
- Department of Surgical Oncology, The Affiliated Hospital of Qinghai University, Xining 810001, China
| | - Jiayong Cui
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining 810008, China; (S.C.); (J.C.); (Y.S.)
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University Medical College, Xining 810008, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Qinghai University Medical College, Xining 810008, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University Medical College, Xining 810008, China
| | - Yinuo Sun
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining 810008, China; (S.C.); (J.C.); (Y.S.)
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University Medical College, Xining 810008, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Qinghai University Medical College, Xining 810008, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University Medical College, Xining 810008, China
- Department of Economics, School of Finance and Economics, Qinghai University, Xining 810016, China
| | - Xiaowu Wang
- Department of Surgical Oncology, The Affiliated Hospital of Qinghai University, Xining 810001, China
| | - Chunmei Cai
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining 810008, China; (S.C.); (J.C.); (Y.S.)
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University Medical College, Xining 810008, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Qinghai University Medical College, Xining 810008, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University Medical College, Xining 810008, China
| |
Collapse
|
26
|
Chen IP, Henning S, Bender M, Degenhardt S, Mhamdi Ghodbani M, Bergmann AK, Volkmer B, Brockhoff G, Wege AK, Greinert R. Detection of Human Circulating and Extracellular Vesicle-Derived miRNAs in Serum of Humanized Mice Transplanted with Human Breast Cancer (HER2 + and TNBC) Cells-A Proof of Principle Investigation. Int J Mol Sci 2025; 26:3629. [PMID: 40332177 PMCID: PMC12026515 DOI: 10.3390/ijms26083629] [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: 02/10/2025] [Revised: 04/03/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
Humanized tumor mice (HTM) allow for preclinical cancer treatment studies of breast cancer (BC) under human-like conditions. This study utilized HTM for the first time to investigate potential miRNA biomarker candidates for treatment response in sera and extracellular vesicles (EVs), following X-irradiation and atezolizumab (anti-PD-L1) treatment. We identified the changes of human-specific miRNAs (miR-23b-3p and miR-155-5p) after irradiation and anti-PD-L1 treatment in HTMs with human epidermal growth factor receptor 2 positive (HER2+ BC) and triple-negative breast cancer (TNBC). The high degree of conserved, circulating free miRNA in mice and men represents a challenge of our assay; however, miRNAs with ≥2 nucleotide mismatches can be employed for human-specific analysis, and even conserved miRNAs may be utilized under clearly defined conditions of human tumor growth in HTM. A comparative analysis of extracellular vesicle miRNA cargo and free-circulating serum miRNAs revealed several exosome-specific miRNAs (miR-29b-3p, miR-34c-5p, miR-203a-3p, miR-378g, and miR-382-5p) in HTMs, which are known to play roles in BC. Our findings demonstrate that HTMs are a suitable model to identify treatment-induced changes in free-circulating and exosomal miRNAs that influence tumor progression and immunological tumor defense, both locally and at distant sites. This study presents a proof-of-principle approach to analyzing cell-free nucleotides and exosomes in a human-like, preclinical in vivo setting. Further refinements are necessary to enhance the sensitivity and the specificity of the HTM-based approach.
Collapse
Affiliation(s)
- I-Peng Chen
- Department of Molecular Cell Biology, Skin Cancer Center Buxtehude, Elbekliniken Stade-Buxtehude, 21614 Buxtehude, Germany; (I.-P.C.); (S.H.); (M.B.); (M.M.G.); (B.V.)
| | - Stefan Henning
- Department of Molecular Cell Biology, Skin Cancer Center Buxtehude, Elbekliniken Stade-Buxtehude, 21614 Buxtehude, Germany; (I.-P.C.); (S.H.); (M.B.); (M.M.G.); (B.V.)
| | - Marc Bender
- Department of Molecular Cell Biology, Skin Cancer Center Buxtehude, Elbekliniken Stade-Buxtehude, 21614 Buxtehude, Germany; (I.-P.C.); (S.H.); (M.B.); (M.M.G.); (B.V.)
| | - Sarah Degenhardt
- Department of Molecular Cell Biology, Skin Cancer Center Buxtehude, Elbekliniken Stade-Buxtehude, 21614 Buxtehude, Germany; (I.-P.C.); (S.H.); (M.B.); (M.M.G.); (B.V.)
| | - Mouna Mhamdi Ghodbani
- Department of Molecular Cell Biology, Skin Cancer Center Buxtehude, Elbekliniken Stade-Buxtehude, 21614 Buxtehude, Germany; (I.-P.C.); (S.H.); (M.B.); (M.M.G.); (B.V.)
| | - Ann Kathrin Bergmann
- Core Facility of Electron Microscopy, University Clinics Duesseldorf, 40225 Duesseldorf, Germany;
| | - Beate Volkmer
- Department of Molecular Cell Biology, Skin Cancer Center Buxtehude, Elbekliniken Stade-Buxtehude, 21614 Buxtehude, Germany; (I.-P.C.); (S.H.); (M.B.); (M.M.G.); (B.V.)
| | - Gero Brockhoff
- Department of Gynecology and Obstetrics, Medical Center Regensburg, 93053 Regensburg, Germany; (G.B.); (A.K.W.)
- Bavarian Cancer Research Center (BZKF), 93053 Regensburg, Germany
| | - Anja K. Wege
- Department of Gynecology and Obstetrics, Medical Center Regensburg, 93053 Regensburg, Germany; (G.B.); (A.K.W.)
- Bavarian Cancer Research Center (BZKF), 93053 Regensburg, Germany
| | - Rüdiger Greinert
- Department of Molecular Cell Biology, Skin Cancer Center Buxtehude, Elbekliniken Stade-Buxtehude, 21614 Buxtehude, Germany; (I.-P.C.); (S.H.); (M.B.); (M.M.G.); (B.V.)
| |
Collapse
|
27
|
Justiz-Vaillant A, Pandit BR, Unakal C, Vuma S, Akpaka PE. A Comprehensive Review About the Use of Monoclonal Antibodies in Cancer Therapy. Antibodies (Basel) 2025; 14:35. [PMID: 40265416 PMCID: PMC12015915 DOI: 10.3390/antib14020035] [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: 01/27/2025] [Revised: 03/14/2025] [Accepted: 04/09/2025] [Indexed: 04/24/2025] Open
Abstract
Monoclonal antibodies (mAbs) targeting various pathways in cancer therapy play crucial roles in enhancing the immune system's ability to recognise and eliminate tumour cells. These therapies are designed to either block inhibitory immune checkpoint pathways or to target specific tumour cell markers for direct destruction. Additionally, mAbs can modulate the tumour microenvironment, enhance antibody-dependent cellular cytotoxicity, and inhibit angiogenesis, further amplifying their therapeutic impact. Below is a summary of monoclonal antibodies targeting key pathways, along with their indications and mechanisms of action, which are reviewed based on therapeutic mechanisms.
Collapse
Affiliation(s)
| | | | | | | | - Patrick Eberechi Akpaka
- Department of Pathology/Microbiology & Pharmacology, The University of the West Indies, St. Augustine Campus, St. Augustine 330912, Trinidad and Tobago; (A.J.-V.); (B.R.P.); (C.U.); (S.V.)
| |
Collapse
|
28
|
Ahn D, Park SH, Lee YG, Jeong MS, Gone GB, Cho Y, Chung SJ. Enhanced Targeted Drug Delivery System to Control Avidity and Drug Encapsulation Using E2 Nanocages and SpyTag/SpyCatcher. ACS Biomater Sci Eng 2025. [PMID: 40209085 DOI: 10.1021/acsbiomaterials.5c00224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2025]
Abstract
Although antibody-drug conjugates offer advanced targeted anticancer therapy that overcomes the limitations of conventional chemotherapy and therapeutic antibodies, they are restricted in their capacity to carry multiple hydrophobic payloads. Protein nanocages have emerged as versatile therapeutic platforms for targeted drug delivery, offering advantages like precise molecular assembly, biocompatibility, and multivalent targeting. This study presents the development of engineered E2 nanocages functionalized with anti-HER2 single-chain variable fragments (scFv) using the SpyTag/SpyCatcher ligation system to achieve controlled scFv display valency. The results demonstrate that increasing anti-HER2 scFv valency enhances HER2 binding affinity via avidity effects, with the highest valency nanocages showing the highest binding avidity. Furthermore, cysteine residues were introduced into the E2 nanocages to enable conjugation with monomethyl auristatin E (MMAE) through maleimide chemistry, achieving efficient drug loading. The resulting MMAE-conjugated nanocages displayed potent, subnanomolar cytotoxicity in HER2-positive SKBR3 and BT-474 cell lines while sparing HER2-negative MDA-MB-231 cells at concentrations up to 1 nM. These results underscore the critical role of scFv valency in enhancing HER2 targeting and highlight the potential of E2 protein nanocages as specific, potent platforms for targeted cancer therapy. In this study, we developed an enhanced targeted drug delivery system using E2 nanocages and scFv with SpyCatcher/SpyTag ligation to regulate binding avidity and encapsulate hydrophobic drugs. The modular design and pH-sensitive dissociation of these nanocages establish a foundation for next-generation precision medicine strategies.
Collapse
Affiliation(s)
- Dohee Ahn
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sun Hee Park
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
- AbTis Co., Ltd., Yongin 17073, Republic of Korea
| | - Yeong Geun Lee
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
- AbTis Co., Ltd., Yongin 17073, Republic of Korea
| | - Myeong Seon Jeong
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon 24341, Korea
| | - Geetanjali B Gone
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Younghun Cho
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, South Korea
| | - Sang J Chung
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
- AbTis Co., Ltd., Yongin 17073, Republic of Korea
| |
Collapse
|
29
|
Liu J, Liu Z, Hu J, Fan B, Zhang S, Chang K, Mao X, Huang G, Liu Z, Ma L. Anti-breast cancer activity of a novel genetically engineered fusion protein composed of HER2 affibody and proapoptotic peptide R8-KLA. Med Oncol 2025; 42:155. [PMID: 40205290 DOI: 10.1007/s12032-025-02707-3] [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: 11/27/2024] [Accepted: 03/29/2025] [Indexed: 04/11/2025]
Abstract
HER2-positive breast cancer is an aggressive subtype with unfavorable prognoses. Although HER2-targeted agents represented by monoclonal antibodies have achieved remarkable success in the clinic, there are still a substantial number of patients with disease relapse. Recently, multifunctional fusion proteins obtained via genetic engineering technology have received much attention in targeted tumor therapy, especially in breast cancer. In this study, we genetically engineered a novel recombinant fusion protein, named HMK, which was designed as a bifunctional construct including the HER2-specific affibody ZHER2:342 for targeted receptor recognition, and a proapoptotic module featuring a cell-penetrating octa-arginine (R8) motif conjugated to an antimicrobial peptide KLA. High-purity HMK proteins were successfully obtained using E. coli expression system and Ni-Nitrilotriacetic acid affinity purification method. HMK exhibited higher cytotoxicity in HER2-positive breast cancer cells SK-BR-3 (IC50 of 8.36 ± 0.62 μM) compared to normal breast epithelial cells MCF-10A (IC50 of 32.40 ± 2.93 μM), demonstrating favorable selectivity. HMK induced apoptosis in SK-BR-3 cells via activating both endogenous and exogenous apoptotic pathways, as evidenced by the cleavage of Caspase 8, Caspase 9, Caspase 3, and PARP. Caspase inhibitor Z-VAD significantly reversed the function of HMK in SK-BR-3 cells, suggesting that caspase-dependent apoptosis was crucial for the anti-breast cancer activity of HMK. Our results suggested that HMK protein may have the potential to become a candidate molecule for HER2-positive breast cancer treatment.
Collapse
Affiliation(s)
- Jian Liu
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China
| | - Zi Liu
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China.
| | - Junfeng Hu
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China
| | - Binru Fan
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China
| | - Shizhun Zhang
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China
| | - Kaili Chang
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China
| | - Xiuping Mao
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China
| | - Guozheng Huang
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China
| | - Zhi Liu
- Department of Pathology, Ma'anshan Municipal People's Hospital, Ma'anshan, Anhui, 243000, China
| | - Liang Ma
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China.
| |
Collapse
|
30
|
Robbins CJ, Bates KM, Rimm DL. HER2 testing: evolution and update for a companion diagnostic assay. Nat Rev Clin Oncol 2025:10.1038/s41571-025-01016-y. [PMID: 40195456 DOI: 10.1038/s41571-025-01016-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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
|
31
|
Jenkins JW, Peña A, Castro SA, Hansen MJ, Van Keulen VP, Foster ST, Rios-Cruz PE, Yakubov J, Hinson DT, Olivier SM, Pavelko KD, Felts SJ, Johnson AJ, Pease LR. MHC class II-mediated spontaneous rejection of breast carcinomas expressing model neoantigens. J Immunother Cancer 2025; 13:e010434. [PMID: 40187751 PMCID: PMC11973762 DOI: 10.1136/jitc-2024-010434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 03/06/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Cancers persist despite expression of immunogenic neoantigens and ongoing antitumor immune responses. While some occult tumors likely are cleared by effective antitumor immune responses, the targeted antigens are not easily identifiable as those tumors spontaneously disappear. METHODS We used mouse models with a defined antigenic protein mimicking tumor-specific neoantigens to address the nature of these spontaneous anti-tumor immune responses. RESULTS BALB/c (H-2d ) mice challenged with BALB/c breast tumors expressing the rat-erbB2 oncoprotein succumb to their tumors despite ongoing immune responses targeting tumor-specific model antigens. Meanwhile, congenic BALB.B (H-2b ) and H-2d/H-2b F1 hybrid mice spontaneously eliminate genetically matched tumors in a major histocompatibility complex (MHC)-II dependent manner. Adoptive transfer and immune cell depletion strategies revealed CD4+ T cells and CD20+ B cells are crucial mediators of the protective response in H-2b mice. Furthermore, passive transfer of immune serum from mice rejecting their tumors confers resistance in tumor antigen-tolerant animals with an inversely proportional relationship between tumor outgrowth and the amount of rat-erbB2 specific antibody present in tumor-bearing mice. Introduction of the rat-erb2 ectodomain into other H-2b tumor models also promotes their spontaneous tumor rejection. Notably, the tumor microenvironments differ in rat-erbB2+ tumor-bearing BALB.B and BALB/c mice at the time of fate decision in the models reflecting the differences between effective and ineffective tumor immune responses. CONCLUSIONS We find that the effective antitumor immunity targeting neoantigens in these breast cancer models is determined by MHC-II-restricted presentation of optimal cancer-associated antigens. These responses are dependent on CD4+ T cells, B cells, and antigen-specific antibodies.
Collapse
Affiliation(s)
| | - Alvaro Peña
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sarah A Castro
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael J Hansen
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Sean T Foster
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Joshua Yakubov
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Destin T Hinson
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Samuel M Olivier
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Kevin D Pavelko
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sara J Felts
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Aaron J Johnson
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Larry R Pease
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
32
|
Mulliqi E, Khelwatty S, Bagwan I, Kamaludin A, Morgan A, Long N, Ashkan K, Modjtahedi H. The Co-Expression and Cellular Location of HER Family Members, EGFRvIII, Putative Cancer Stem Cell Biomarkers CD44 and CD109 in Patients with Glioblastoma, and Their Impacts on Prognosis. Cancers (Basel) 2025; 17:1221. [PMID: 40227788 PMCID: PMC11987930 DOI: 10.3390/cancers17071221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/26/2025] [Accepted: 03/30/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES The aberrant expression and activation of HER family members is a known major oncogenic pathway for the proliferation, progression, and metastasis of a wide range of human malignancies. In this study, our aim was to examine the relative expression and prognostic significance of all members of the HER family, the type III EGFR mutant (EGFRvIII), and the putative stem cell markers CD44 and CD109 in patients with glioblastoma. METHODS The expression levels of wild-type EGFR (wtEGFR), HER2, HER3, HER4, EGFRvIII, CD44, and CD109 were determined in tumour specimens from 80 patients by immunohistochemistry. The staining was scored based on the percentage of positive tumour cells, the intensity, and the cellular location of immunostaining. The association between the expression level of the biomarkers and patient overall survival was evaluated using Chi-squared, Kaplan-Meier survival curves, and log-rank tests. RESULTS At a cut-off value of ≥5% with positive staining, 46% (wtEGFR), 75% (HER2), 19% (HER3), 71% (HER4), 85% (EGFRvIII), 95% (CD44), and 16% (CD109) of the cases were positive for these biomarkers. Interestingly, at the same cut-off value, the expression of wtEGFR in these patients was accompanied by co-expression with HER2 (35%), HER3 (0%), HER4 (30%), EGFRvIII (36%), CD44 (44%), HER2/EGFRvIII (28%), HER2/CD44 (31%), and EGFRvIII/CD44 (36%). In addition, the expression of EGFRvIII was accompanied by co-expression with HER2 (65%), HER3 (15%), HER4 (63%), CD44 (83%), CD109 (16%), wtEGFR/HER2 (28%), and 55% of the cases had co-expression of EGFRvIII/HER2/HER4/CD44. With the exception of HER2 expression, at cut-off values of ≥5% of tumour cells with positive staining, which was associated with better overall survival [HR = 0.57 (p = 0.038), HR = 0.56 (p = 0.034)], there was no significant association between the expression of other members of the HER family, EGFRvIII, CD44, and CD109 on the overall survival in both univariate and multivariate analysis. Conclusions Our results suggest that the co-expression of different members of the HER family, with EGFRvIII, CD44, and CD109, occurs in patients with glioblastoma. As the results of therapy with EGFR inhibitors have not been encouraging in patients with a brain tumour, further investigation should determine whether the co-expression of such biomarkers can be of predictive value for the response to the therapy with various types of HER inhibitors and their potential as therapeutic targets for co-targeted therapy.
Collapse
Affiliation(s)
- Ermira Mulliqi
- School of Life Science, Pharmacy and Chemistry, Faculty of Health, Science, Social Care and Education, Kingston University London, Kingston-upon-Thames KT1 2EE, UK; (E.M.); (S.K.); (I.B.); (A.M.)
| | - Said Khelwatty
- School of Life Science, Pharmacy and Chemistry, Faculty of Health, Science, Social Care and Education, Kingston University London, Kingston-upon-Thames KT1 2EE, UK; (E.M.); (S.K.); (I.B.); (A.M.)
| | - Izhar Bagwan
- School of Life Science, Pharmacy and Chemistry, Faculty of Health, Science, Social Care and Education, Kingston University London, Kingston-upon-Thames KT1 2EE, UK; (E.M.); (S.K.); (I.B.); (A.M.)
- Berkshire Surrey Pathology Services, Royal Surrey Hospital, Guildford GU2 7XX, UK
| | - Ahmad Kamaludin
- Department of Neurosurgery, Kings College Hospital, Denmark Hill, London SE5 9RS, UK; (A.K.); (N.L.); (K.A.)
| | - Anna Morgan
- School of Life Science, Pharmacy and Chemistry, Faculty of Health, Science, Social Care and Education, Kingston University London, Kingston-upon-Thames KT1 2EE, UK; (E.M.); (S.K.); (I.B.); (A.M.)
| | - Natalie Long
- Department of Neurosurgery, Kings College Hospital, Denmark Hill, London SE5 9RS, UK; (A.K.); (N.L.); (K.A.)
| | - Keyoumars Ashkan
- Department of Neurosurgery, Kings College Hospital, Denmark Hill, London SE5 9RS, UK; (A.K.); (N.L.); (K.A.)
| | - Helmout Modjtahedi
- School of Life Science, Pharmacy and Chemistry, Faculty of Health, Science, Social Care and Education, Kingston University London, Kingston-upon-Thames KT1 2EE, UK; (E.M.); (S.K.); (I.B.); (A.M.)
| |
Collapse
|
33
|
Habibi S, Bahramian S, Saeedeh ZJ, Mehri S, Ababzadeh S, Kavianpour M. Novel strategies in breast cancer management: From treatment to long-term remission. Crit Rev Oncol Hematol 2025; 211:104715. [PMID: 40187709 DOI: 10.1016/j.critrevonc.2025.104715] [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: 03/02/2025] [Revised: 03/22/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
Breast cancer (BC) is the most common malignancy among women and a leading cause of cancer-related mortality worldwide. Although improvements in early detection and therapy have been made, metastatic breast cancer (mBC) continues to be an incurable disease. Although existing treatments can prolong survival and enhance quality of life, they do not provide a definitive cure. Targeted therapies have significantly improved outcomes, particularly for subtypes such as human epidermal growth factor receptor 2 (HER2)-positive and hormone receptor (HR)-positive (HR+) BC. Key innovations include antibodydrug conjugates (ADCs) and next-generation endocrine therapies. ADCs combine monoclonal antibodies with cytotoxic agents, allowing targeted delivery to tumor cells while minimizing systemic toxicity. Immunotherapy is emerging as a promising approach for aggressive subtypes, such as triple-negative breast cancer (TNBC). Strategies under investigation include chimeric antigen receptor T-cell (CAR-T) therapy, tumor-infiltrating lymphocyte (TIL) therapies, and natural killer (NK) cell treatments, all aimed at enhancing the ability of the immune system to target and eliminate resistant tumor cells. Tissue engineering, particularly hydrogel-based delivery systems, offers the potential for localized treatment. These systems enable the controlled release of therapeutic agents or immune cells directly to the tumor site, supporting tissue regeneration and enhancing immune surveillance to reduce recurrence. Despite these advancements, challenges remain, including treatment resistance, the immunosuppressive tumor microenvironment, and high costs. Overcoming these barriers requires further innovation in drug delivery systems and a deeper understanding of tumor biology.
Collapse
Affiliation(s)
- Sina Habibi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shabbou Bahramian
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Zare Jalise Saeedeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Sara Mehri
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Guilan, Iran
| | - Shima Ababzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran; Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Maria Kavianpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran; Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
| |
Collapse
|
34
|
Uematsu M, Nakajima H, Miyake H, Wakabayashi M, Funasaka C, Kondoh C, Harano K, Matsubara N, Hosono A, Naito Y, Sakamoto N, Kojima M, Onishi T, Ishii G, Mukohara T. Digitally quantified area of residual tumor after neoadjuvant chemotherapy in HER2-positive breast cancer. Breast Cancer 2025:10.1007/s12282-025-01694-7. [PMID: 40172786 DOI: 10.1007/s12282-025-01694-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 03/17/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND The area of residual tumor (ART) is a quantitative method for assessing tumors after neoadjuvant chemotherapy (NAC). This study evaluated whether ART can identify a favorable prognosis group in patients with HER2-positive surgically resected breast cancer and residual tumors post-NAC. METHODS We retrospectively reviewed patients with HER2-positive who underwent surgery after NAC, including trastuzumab, from 2005 to 2022 at our institution. ART was assessed at the maximum cut surface of the residual primary tumor using digital pathology images. Receiver operating characteristic curve analysis determined ART-Low and ART-High cutoffs, excluding ART-0 (0 mm2) patients. RESULTS Of the 219 patients, 82 had ART greater than 0 mm2. The median follow-up was 90.2 months. The number of patients in the ART-0, ART-Low (0 < ART ≤ 4.0 mm2), and ART-High (> 4.0 mm2) groups were 137, 39, and 43, respectively. The ART-Low group showed significantly shorter event-free survival compared to the ART-0 group (HR 3.50, 95% CI 1.52-8.06), and the ART-High group also tended toward poorer prognosis (HR 2.31, 95% CI 0.89-5.97). However, there was no significant difference in prognosis between the ART-Low and ART-High groups. CONCLUSIONS The current study suggests that even minimal residual tumor cells in the primary site can significantly impact on prognosis in HER2-positive early breast cancer.
Collapse
Affiliation(s)
- Mao Uematsu
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
- Course of Advanced Clinical Research of Cancer, Juntendo University Graduate School of Medicine, Bunkyō, Japan
| | - Hiromichi Nakajima
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan.
- Department of Experimental Therapeutics, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan.
- Department of General Internal Medicine, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan.
| | - Hirohiko Miyake
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Masashi Wakabayashi
- Biostatistics Division, Center for Research Administration and Support, National Cancer Center, Chuo-Ku, Japan
| | - Chikako Funasaka
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
- Department of Experimental Therapeutics, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Chihiro Kondoh
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Kenichi Harano
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
- Department of Experimental Therapeutics, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Nobuaki Matsubara
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Ako Hosono
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
- Department of Pediatric Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Yoichi Naito
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
- Department of Experimental Therapeutics, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
- Department of General Internal Medicine, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Naoya Sakamoto
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
- Exploratory Oncology Research and Clinical Trial Center, Division of Pathology, Chuo-Ku, Japan
| | - Motohiro Kojima
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Tatsuya Onishi
- Department of Breast Surgery, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Genichiro Ishii
- Course of Advanced Clinical Research of Cancer, Juntendo University Graduate School of Medicine, Bunkyō, Japan
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Toru Mukohara
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
- Course of Advanced Clinical Research of Cancer, Juntendo University Graduate School of Medicine, Bunkyō, Japan
| |
Collapse
|
35
|
Alam R, Reva A, Edwards DG, Lege BM, Munoz-Arcos LS, Reduzzi C, Singh S, Hao X, Wu YH, Tian Z, Natalee LM, Damle G, Demircioglu D, Wang Y, Wu L, Molteni E, Hasson D, Lim B, Gugala Z, Chipuk JE, Lang JE, Sparano JA, Cheng C, Cristofanilli M, Xiao H, Zhang XHF, Bado IL. Bone-Induced HER2 Promotes Secondary Metastasis in HR+/HER2- Breast Cancer. Cancer Discov 2025; 15:818-837. [PMID: 39835789 PMCID: PMC11964846 DOI: 10.1158/2159-8290.cd-23-0543] [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: 05/19/2023] [Revised: 06/12/2024] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
SIGNIFICANCE Given the urgent need for alternative strategies to block metastasis progression, we demonstrate that blocking HER2-mediated secondary metastasis improves clinical outcome and establish HER2 as a biomarker for bone metastasis in patients with initial HR+/HER2- breast cancer, which represents ∼70% of all cases.
Collapse
Affiliation(s)
- Rahat Alam
- Icahn School of Medicine, Mount Sinai, New York, NY 10029, USA
- Tisch Cancer Institute, Mount Sinai, New York, NY 10029, USA
- Department of Oncological Sciences, Mount Sinai, New York, NY 10029, USA
| | - Anna Reva
- Icahn School of Medicine, Mount Sinai, New York, NY 10029, USA
- Tisch Cancer Institute, Mount Sinai, New York, NY 10029, USA
- Department of Oncological Sciences, Mount Sinai, New York, NY 10029, USA
| | - David G. Edwards
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bree M. Lege
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Laura S. Munoz-Arcos
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Carolina Reduzzi
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Swarnima Singh
- Icahn School of Medicine, Mount Sinai, New York, NY 10029, USA
- Tisch Cancer Institute, Mount Sinai, New York, NY 10029, USA
- Department of Oncological Sciences, Mount Sinai, New York, NY 10029, USA
| | - Xiaoxin Hao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yi-Hsuan Wu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zeru Tian
- Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005 USA
| | - Laura M. Natalee
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Gargi Damle
- Icahn School of Medicine, Mount Sinai, New York, NY 10029, USA
- Tisch Cancer Institute, Mount Sinai, New York, NY 10029, USA
| | - Deniz Demircioglu
- Icahn School of Medicine, Mount Sinai, New York, NY 10029, USA
- Tisch Cancer Institute, Mount Sinai, New York, NY 10029, USA
| | - Yixian Wang
- Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005 USA
| | - Ling Wu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Elisabetta Molteni
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Dan Hasson
- Icahn School of Medicine, Mount Sinai, New York, NY 10029, USA
- Tisch Cancer Institute, Mount Sinai, New York, NY 10029, USA
| | - Bora Lim
- Department of Breast Medical Oncology, Division of Cancer Medicine, MD Anderson, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Zbigniew Gugala
- Department of Orthopedic Surgery and Rehabilitation, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA
| | - Jerry E. Chipuk
- Icahn School of Medicine, Mount Sinai, New York, NY 10029, USA
- Tisch Cancer Institute, Mount Sinai, New York, NY 10029, USA
- Department of Oncological Sciences, Mount Sinai, New York, NY 10029, USA
| | - Julie E. Lang
- Department of Cancer Biology, Division of Breast Cancer, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Joseph A. Sparano
- Icahn School of Medicine, Mount Sinai, New York, NY 10029, USA
- Tisch Cancer Institute, Mount Sinai, New York, NY 10029, USA
- Department of Oncological Sciences, Mount Sinai, New York, NY 10029, USA
| | - Chonghui Cheng
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Massimo Cristofanilli
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Han Xiao
- Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005 USA
| | - Xiang H.-F. Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- McNair Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Igor L. Bado
- Icahn School of Medicine, Mount Sinai, New York, NY 10029, USA
- Tisch Cancer Institute, Mount Sinai, New York, NY 10029, USA
- Department of Oncological Sciences, Mount Sinai, New York, NY 10029, USA
- Lead contact
| |
Collapse
|
36
|
Lavender V, Tanay M, Gomm L. The Transformation of Cancer Nursing Propelled by Novel SACT. Semin Oncol Nurs 2025; 41:151843. [PMID: 39952818 DOI: 10.1016/j.soncn.2025.151843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/15/2025] [Accepted: 01/23/2025] [Indexed: 02/17/2025]
Affiliation(s)
- Verna Lavender
- Head of Nursing for Research and Education, Cancer and Surgery, Guy's and St Thomas' NHS Foundation Trust, London, UK.
| | - Mary Tanay
- Nurse Consultant (Genomics), Berkshire Cancer Centre, Royal Berkshire NHS Foundation Trust, London, UK
| | - Linda Gomm
- Advanced Nurse Practitioner (Cell Therapy & Early Phase Trials), Oncology and Haematology Clinical Trials, Guy's and St Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
37
|
Hou R, Wu X, Wang C, Fan H, Zhang Y, Wu H, Wang H, Ding J, Jiang H, Xu J. Tumor‑associated neutrophils: Critical regulators in cancer progression and therapeutic resistance (Review). Int J Oncol 2025; 66:28. [PMID: 40017131 PMCID: PMC11900975 DOI: 10.3892/ijo.2025.5734] [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: 11/21/2024] [Accepted: 02/11/2025] [Indexed: 03/01/2025] Open
Abstract
Cancer is the second leading cause of death among humans worldwide. Despite remarkable improvements in cancer therapies, drug resistance remains a significant challenge. The tumor microenvironment (TME) is intimately associated with therapeutic resistance. Tumor‑associated neutrophils (TANs) are a crucial component of the TME, which, along with other immune cells, play a role in tumorigenesis, development and metastasis. In the current review, the roles of TANs in the TME, as well as the mechanisms of neutrophil‑mediated resistance to cancer therapy, including immunotherapy, chemotherapy, radiotherapy and targeted therapy, were summarized. Furthermore, strategies for neutrophil therapy were discussed and TANs were explored as potential targets for cancer treatment. In conclusion, the need to explore the precise roles, recruitment pathways and mechanisms of action of TANs was highlighted for the purpose of developing therapies that precisely target TANs and reverse drug resistance.
Collapse
Affiliation(s)
- Rui Hou
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Xi Wu
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Cenzhu Wang
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Hanfang Fan
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Yuhan Zhang
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Hanchi Wu
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Huiyu Wang
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Junli Ding
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Huning Jiang
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Junying Xu
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| |
Collapse
|
38
|
Marhold M, Vaz Batista M, Blancas I, Morales C, Saura-Manich C, Saavedra C, Ruíz-Borrego M, Cortez P, Slebe F, Campolier M, Santos JC, Guerrero-Martínez JA, Jiménez-Cortegana C, Rottenmanner B, Forstner H, Bartsch R, Preusser M. TUXEDO-4: phase II study of trastuzumab-deruxtecan in HER2-low breast cancer with new or progressing brain metastases. Future Oncol 2025; 21:1065-1073. [PMID: 40018758 PMCID: PMC11988270 DOI: 10.1080/14796694.2025.2470604] [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: 09/27/2024] [Accepted: 02/19/2025] [Indexed: 03/01/2025] Open
Abstract
CLINICAL TRIAL REGISTRATION NCT06048718 (clinicaltrials.gov); 2023 -506,702-39-00 (EudraCT number).
Collapse
Affiliation(s)
- Maximilian Marhold
- Department of Medicine 1, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Marta Vaz Batista
- Department of Oncology, Hospital Professor Doutor Fernando Fonseca EPE, Lisbon, Portugal
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
| | - Isabel Blancas
- Department of Oncology, Hospital Universitario Clínico San Cecilio, Granada, Spain
- Medicine Department, University of Granada, Granada, Spain
- Área de Oncología Instituto de Investigación Biosanitaria de Granada, Granada, Spain
| | - Cristina Morales
- Servicio de Oncología Médica, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Cristina Saura-Manich
- Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, Barcelona, Spain
| | - Cristina Saavedra
- Departamento de Oncología Médica, Hospital Universitario Ramón y Cajal, Madrid, Spain
- IOB Madrid, Institute of Oncology, Hospital Beata María Ana, Madrid, Spain
| | | | - Patricia Cortez
- IOB Madrid, Institute of Oncology, Hospital Beata María Ana, Madrid, Spain
| | - Felipe Slebe
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
| | - Marta Campolier
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
| | | | | | | | - Beate Rottenmanner
- Department of Medicine 1, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Heidrun Forstner
- Department of Medicine 1, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Rupert Bartsch
- Department of Medicine 1, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Matthias Preusser
- Department of Medicine 1, Division of Oncology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
39
|
Huang H, Zhou Y, Shang C, Zhang Y, Shen Y. A novel anti-HER2/EGFR bispecific antibody-drug conjugate demonstrates promising antitumor efficacy and overcomes resistance to HER2- or EGFR-targeted ADCs. Invest New Drugs 2025; 43:262-275. [PMID: 39982632 DOI: 10.1007/s10637-025-01507-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 01/17/2025] [Indexed: 02/22/2025]
Abstract
HER2 and EGFR are frequently co-expressed in various tumors. While antibody-drug conjugates (ADCs) targeting HER2, such as T-DM1 and T-Dxd, have shown remarkable antitumor effects in clinical responses, their effectiveness is constrained by drug resistance. EGFR amplification or high expression is one of the factors that lead to resistance against HER2-targeted ADCs. Likewise, the amplification of HER2 may lead to the development of resistance to EGFR-targeted therapies. To overcome these challenges, we, therefore, developed a bispecific antibody (B2C4) that targets HER2 and EGFR. B2C4 exhibited strong binding affinity and internalization activity in tumor cells with high expression of HER2 and EGFR, as well as in those with high expression of either target. B2C4 was then conjugated with vc-MMAE to create a bispecific ADC (B2C4-MMAE) with an average DAR of 4.05. By effectively engaging both arms of the bispecific ADC, B2C4-MMAE demonstrated significant antitumor activity in tumor cells and animal models that were unresponsive HER2- or EGFR-targeted ADCs. B2C4-MMAE could serve as an alternative therapeutic option for tumors that are resistant to single-target treatments. Additionally, B2C4-MMAE exhibited potential in treating tumors resistant to T-Dxd, underscoring its promise as a treatment for challenging cases.
Collapse
Affiliation(s)
- Huoying Huang
- Anhui University of Chinese Medicine, Hefei, People's Republic of China
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, People's Republic of China
- Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong, People's Republic of China
| | - Yuxin Zhou
- Anhui University of Chinese Medicine, Hefei, People's Republic of China
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, People's Republic of China
- Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong, People's Republic of China
| | - Chengzhang Shang
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, People's Republic of China
| | - Yifu Zhang
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, People's Republic of China
| | - Yuelei Shen
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, People's Republic of China.
- Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong, People's Republic of China.
| |
Collapse
|
40
|
Chen O, Fu L, Wang Y, Li J, Liu J, Wen Y. Targeting HSP90AA1 to overcome multiple drug resistance in breast cancer using magnetic nanoparticles loaded with salicylic acid. Int J Biol Macromol 2025; 298:139443. [PMID: 39756742 DOI: 10.1016/j.ijbiomac.2024.139443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/31/2024] [Accepted: 12/31/2024] [Indexed: 01/07/2025]
Abstract
Multiple drug resistance (MDR) remains a major obstacle in effective breast cancer chemotherapy. This study explores the role of HSP90AA1 in driving MDR and evaluates the potential of magnetic nanoparticles (Fe3O4@SA) loaded with salicylic acid (SA) to counteract drug resistance. A comprehensive screening of 200 SA-related target genes identified nine core genes, including HSP90AA1. Pharmacophore analysis revealed that SA interacts with HSP90AA1, a key regulator of mitochondrial K+ channels. Fe3O4@SA nanoparticles demonstrated efficient cellular uptake and lysosomal escape, markedly improving the chemosensitivity of resistant breast cancer cells and promoting apoptosis. In vivo experiments further confirmed the anticancer efficacy of Fe3O4@SA, highlighting its potential as a promising therapeutic strategy to overcome MDR in breast cancer.
Collapse
Affiliation(s)
- Ou Chen
- Department of clinical laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Linlin Fu
- Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yu Wang
- Department of Pharmacy, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Jinggui Li
- Liaoning Jiahe Hospital of Traditional Chinese Medicine, Medical Imaging Center, Shenyang, China
| | - Jun Liu
- Department of cardiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China.
| | - Yanqing Wen
- Department of Pharmacy, The Fourth Affiliated Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
41
|
Zhang Q, Wang X, Shao Z, Zhang Y, Zhang L, Chen M, Zhou X, Zhu H, Zhou Y, Lu X, Li P, Chi W, Li L, Shao Z, Huang S, Xue J, Chi Y, Wu J, Xiu B. LINC01235 Promotes Clonal Evolution through DNA Replication Licensing-Induced Chromosomal Instability in Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413527. [PMID: 39950924 PMCID: PMC11984920 DOI: 10.1002/advs.202413527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/19/2025] [Indexed: 04/12/2025]
Abstract
Despite the development of HER2-targeting drugs such as trastuzumab and T-DXd, treatment resistance is a substantial challenge, often leading to relapse and distant metastasis. Tumor heterogeneity in HER2-positive breast cancer drives the evolution of resistant clones following therapeutic stress. However, the targetable drivers of anti-HER2 treatment resistance are not thoroughly identified. This study aims to use neoadjuvant-targeted therapy cohorts and a patient-derived organoid in vitro treatment model to uncover the potential targetable drivers of anti-HER2 treatment resistance. it is found that LINC01235 significantly enhances DNA replication licensing and chromosomal instability, fostering clonal expansion and evolution, and ultimately increasing resistance to therapeutic interventions. LINC01235 regulates global H3K27ac, H3K9ac, and H3K36me3 modifications, promotes H2A.Z expression in regulatory regions, and increases the accessibility of DNA licensing factors to their promoter regions. XRCC5 is identified as a key component for maintaining genomic stability, crucial for LINC01235's role in replication licensing. Furthermore, therapeutic strategies targeting LINC01235, including the use of antisense oligonucleotides or ATR inhibitors, which showed promise in overcoming treatment resistance are explored. These findings underscore the pivotal role of LINC01235 in driving resistance mechanisms and highlight novel avenues for targeted therapies to improve the outcomes of patients with HER2-positive breast cancer.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Xuliren Wang
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Zhibo Shao
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Yi Zhang
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Liyi Zhang
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Ming Chen
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Xujie Zhou
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Han Zhu
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
| | - Yue Zhou
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Xinya Lu
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Pei Li
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Weiru Chi
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Lun Li
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
- Department of General SurgeryThe Second Xiangya HospitalCentral South UniversityHunan410011China
| | - Zhi‐Ming Shao
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Shenglin Huang
- Fudan University Shanghai Cancer CenterKey Laboratory of Medical Epigenetics and MetabolismInstitutes of Biomedical SciencesFudan UniversityShanghai200032China
| | - Jingyan Xue
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Yayun Chi
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
- Pathology CenterShanghai General HospitalShanghai Jiaotong University School of MedicineShanghai200080China
| | - Jiong Wu
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
- Collaborative Innovation Center for Cancer MedicineShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Bingqiu Xiu
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| |
Collapse
|
42
|
Feng K, Yi Z, Xu B. Artificial Intelligence and Breast Cancer Management: From Data to the Clinic. CANCER INNOVATION 2025; 4:e159. [PMID: 39981497 PMCID: PMC11840326 DOI: 10.1002/cai2.159] [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: 07/07/2024] [Revised: 10/10/2024] [Accepted: 10/22/2024] [Indexed: 02/22/2025]
Abstract
Breast cancer (BC) remains a significant threat to women's health worldwide. The oncology field had an exponential growth in the abundance of medical images, clinical information, and genomic data. With its continuous advancement and refinement, artificial intelligence (AI) has demonstrated exceptional capabilities in processing intricate multidimensional BC-related data. AI has proven advantageous in various facets of BC management, encompassing efficient screening and diagnosis, precise prognosis assessment, and personalized treatment planning. However, the implementation of AI into precision medicine and clinical practice presents ongoing challenges that necessitate enhanced regulation, transparency, fairness, and integration of multiple clinical pathways. In this review, we provide a comprehensive overview of the current research related to AI in BC, highlighting its extensive applications throughout the whole BC cycle management and its potential for innovative impact. Furthermore, this article emphasizes the significance of constructing patient-oriented AI algorithms. Additionally, we explore the opportunities and potential research directions within this burgeoning field.
Collapse
Affiliation(s)
- Kaixiang Feng
- Department of Breast and Thyroid Surgery, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study CenterZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study CenterZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
| | - Zongbi Yi
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study CenterZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
43
|
Shaik R, Mounika V, Begum S, Rajkumar A, Mallikarjun B, Sri Harshini V, Kolure R, Sreevani B, Thakur S. Monoclonal Antibodies in Clinical Trials for Breast Cancer Treatment. Monoclon Antib Immunodiagn Immunother 2025; 44:17-39. [PMID: 40171653 DOI: 10.1089/mab.2024.0018] [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: 04/04/2025] Open
Abstract
One of the most potent therapeutic and diagnostic agents in contemporary medicine is the monoclonal antibody (mAb). mAbs can perform a variety of tasks in breast cancer (BC), including identifying and delivering therapeutic medications to targets, preventing cell development, and suppressing immune system inhibitors including directly attacking cancer cells. mAbs are one of the most effective therapeutic options, particularly for HER2, but they have not been well studied for their use in treating other forms of BC, particularly triple negative breast tumors. Bispecific and trispecific mAbs have created new opportunities for more targeted specific efficacy, which has a positive impact on the viability of antigen specificity. They are more versatile and effective than other forms of treatment, emerging as most popular option for treating BC. However, mAbs have a limit in treatment due to certain adverse effects, including fever, shaking, exhaustion, headache, nausea, and vomiting, as well as rashes, bleeding, and difficulty breathing. To examine the current and prospective future capacities of mAbs with regard to the detection and treatment of BC, the present review highlights advantages and disadvantages of mAb approach.
Collapse
Affiliation(s)
- Rahaman Shaik
- School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| | - Varikuppala Mounika
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| | - Shireen Begum
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| | - Agolapu Rajkumar
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| | - Bathurasi Mallikarjun
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| | - Vollala Sri Harshini
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| | - Rajini Kolure
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| | | | - Sneha Thakur
- Department of Pharmacognosy, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| |
Collapse
|
44
|
Iacovacci J, Brough R, Moughari FA, Alexander J, Kemp H, Tutt ANJ, Natrajan R, Lord CJ, Haider S. Proteogenomic discovery of RB1-defective phenocopy in cancer predicts disease outcome, response to treatment, and therapeutic targets. SCIENCE ADVANCES 2025; 11:eadq9495. [PMID: 40138429 PMCID: PMC11939072 DOI: 10.1126/sciadv.adq9495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 02/11/2025] [Indexed: 03/29/2025]
Abstract
Genomic defects caused by truncating mutations or deletions in the Retinoblastoma tumor suppressor gene (RB1) are frequently observed in many cancer types leading to dysregulation of the RB pathway. Here, we propose an integrative proteogenomic approach that predicts cancers with dysregulation in the RB pathway. A subset of these cancers, which we term as "RBness," lack RB1 genomic defects and yet phenocopy the transcriptional profile of RB1-defective cancers. We report RBness as a pan-cancer phenomenon, associated with patient outcome and chemotherapy response in multiple cancer types, and predictive of CDK4/6 inhibitor response in estrogen-positive breast cancer. Using RNA interference and a CRISPR-Cas9 screen in isogenic models, we find that RBness cancers also phenocopy synthetic lethal vulnerabilities of cells with RB1 genomic defects. In summary, our findings suggest that dysregulation of the RB pathway in cancers lacking RB1 genomic defects provides a molecular rationale for how these cancers could be treated.
Collapse
Affiliation(s)
- Jacopo Iacovacci
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
- Data Science Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano 20133, Italy
| | - Rachel Brough
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
- CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Fatemeh Ahmadi Moughari
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - John Alexander
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Harriet Kemp
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Andrew N. J. Tutt
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Rachael Natrajan
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Christopher J. Lord
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
- CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Syed Haider
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| |
Collapse
|
45
|
Zhang Y, Xu Y, Zhang Y, Wang S, Zhao M. The multiple functions and mechanisms of long non-coding RNAs in regulating breast cancer progression. Front Pharmacol 2025; 16:1559408. [PMID: 40223929 PMCID: PMC11985786 DOI: 10.3389/fphar.2025.1559408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 03/14/2025] [Indexed: 04/15/2025] Open
Abstract
Breast cancer (BC) is a malignant tumor that has the highest morbidity and mortality rates in the female population, and its high tendency to metastasize is the main cause of poor clinical prognosis. Long non-coding RNAs (lncRNAs) have been extensively documented to exhibit aberrant expression in various cancers and influence tumor progression via multiple molecular pathways. These lncRNAs not only modulate numerous aspects of gene expression in cancer cells, such as transcription, translation, and post-translational modifications, but also play a crucial role in the reprogramming of energy metabolism by regulating metabolic regulators, which is particularly significant in advanced BC. This review examines the characteristics and mechanisms of lncRNAs in regulating BC cells, both intracellularly (e.g., cell cycle, autophagy) and extracellularly (e.g., tumor microenvironment). Furthermore, we explore the potential of specific lncRNAs and their regulatory factors as molecular markers and therapeutic targets. Lastly, we summarize the application of lncRNAs in the treatment of advanced BC, aiming to offer novel personalized therapeutic options for patients.
Collapse
Affiliation(s)
- Yongsheng Zhang
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
- Department of Anesthesia and Perioperative Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Yanjiao Xu
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yanping Zhang
- Department of Anesthesia and Perioperative Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Shoushi Wang
- Department of Anesthesia and Perioperative Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Mingqiang Zhao
- Department of Anesthesia and Perioperative Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| |
Collapse
|
46
|
Kannappan P, Kaniyur Chandrasekaran M, Vani Raju M, Gopalakrishnan S, Dhamodharan P, Muthaiyan Ahalliya R, Palanisamy CP, Raju B, Velliyur Kanniappan G. Exploring the anticancer potential of Jerantinine A from Tabernaemontana coronaria against prostate, breast, and ovarian cancers: a computational approach. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2025:jcim-2024-0443. [PMID: 40147045 DOI: 10.1515/jcim-2024-0443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/28/2025] [Indexed: 03/29/2025]
Abstract
OBJECTIVES Cancer remains a significant global health challenge, with prostate, breast, and ovarian cancers ranking among the leading causes of morbidity and mortality. Natural products, particularly those derived from medicinal plants, have gained attention for their potential in alternative cancer therapies. The main objective of the study was an isolation and characterization of Jerantinine A, a bioactive alkaloid from Tabernaemontana coronaria, and its interactions with key protein targets involved in cancer progression. METHODS Jerantinine A was isolated through column chromatography and characterized using spectroscopic techniques, including UV-visible spectroscopy, FTIR, and NMR. Molecular docking studies were performed to assess its binding affinities with six critical protein targets: PTEN and androgen receptor for prostate cancer, CXCR4 and HER2 for ovarian cancer, and CDK1 and NEK2 for breast cancer. RESULTS Molecular docking analyses revealed that Jerantinine A exhibits strong binding affinities with all six protein targets, suggesting its potential to inhibit cancer cell proliferation by interfering with key signaling pathways. CONCLUSIONS These findings underscore the therapeutic potential of T. coronaria, particularly through Jerantinine A, as a promising candidate for cancer treatment. By targeting pivotal proteins associated with prostate, breast, and ovarian cancers, Jerantinine A offers a foundation for further research and development as a novel anti-cancer agent.
Collapse
Affiliation(s)
- Poornima Kannappan
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
| | | | - Manikandan Vani Raju
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
| | - Shankari Gopalakrishnan
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
- Centre for Bioinformatics, Karpagam Academy of Higher Education, Coimbatore, India
| | - Prabhu Dhamodharan
- Department of Biotechnology, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
- Centre for Bioinformatics, Karpagam Academy of Higher Education, Coimbatore, India
| | - Rathi Muthaiyan Ahalliya
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
- Department of Biotechnology, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
| | - Chella Perumal Palanisamy
- Centre for Global Health Research, Saveetha Medical College & Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai, Tamil Nadu, India
| | - Balaji Raju
- Centre for Global Health Research, Saveetha Medical College & Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai, Tamil Nadu, India
| | - Gopalakrishnan Velliyur Kanniappan
- Department of Physiology, 194347 Saveetha Medical College & Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS) , Thandalam, Chennai, Tamil Nadu, India
| |
Collapse
|
47
|
Carvalho E, Canberk S, Schmitt F, Vale N. Molecular Subtypes and Mechanisms of Breast Cancer: Precision Medicine Approaches for Targeted Therapies. Cancers (Basel) 2025; 17:1102. [PMID: 40227634 PMCID: PMC11987866 DOI: 10.3390/cancers17071102] [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: 02/18/2025] [Revised: 03/20/2025] [Accepted: 03/23/2025] [Indexed: 04/15/2025] Open
Abstract
Breast cancer remains one of the most prevalent diseases worldwide, primarily affecting women. Its heterogeneous nature poses a significant challenge in the development of effective and targeted treatments. Molecular characterization has enabled breast cancer to be classified into four main subtypes: luminal A, luminal B, HER2-positive, and triple-negative breast cancer, based on hormone receptor expression and HER2 status. A deeper understanding of these molecular markers and their associated signaling pathways, such as MAPK and PI3K/AKT, is essential for improving prognosis and optimizing treatment strategies. Currently, several therapeutic agents are utilized in neoadjuvant and adjuvant therapies, often in combination with surgical interventions. However, emerging evidence highlights the growing challenge of drug resistance, which significantly limits the efficacy of existing treatments. Addressing this issue may require innovative approaches, including combination therapies and precision medicine strategies, tailored to the molecular profile of each patient. Therefore, a comprehensive understanding of the pathophysiologic mechanisms driving breast cancer progression and resistance is crucial for the development of advanced targeted therapies with greater precision and efficacy. This review aims to explore recent advancements in molecular research related to breast cancer subtypes and provide a critical analysis of current therapeutic approaches within the framework of precision medicine.
Collapse
Affiliation(s)
- Eduarda Carvalho
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; (E.C.); (S.C.); (F.S.)
| | - Sule Canberk
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; (E.C.); (S.C.); (F.S.)
- RISE-Health, Department of Pathology, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Fernando Schmitt
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; (E.C.); (S.C.); (F.S.)
- RISE-Health, Department of Pathology, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Nuno Vale
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; (E.C.); (S.C.); (F.S.)
- RISE-Health, Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|
48
|
Lian J, Yao R, Pang S, Ren X, Pan B, Zhou Y. Comprehensive analysis of HER2 Low Breast Cancer Response to Neoadjuvant Chemotherapy, a Retrospective Cohort Study. Clin Breast Cancer 2025:S1526-8209(25)00081-3. [PMID: 40254501 DOI: 10.1016/j.clbc.2025.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/08/2025] [Accepted: 03/20/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND This study aimed to identify the response and survival outcomes of HER2-low patients following neoadjuvant chemotherapy (NAC). METHOD A retrospective cohort of patients who received NAC in the Department of Breast Surgery at Peking Union Medical College Hospital (PUMCH) from September 27, 2016, to January 23, 2024, was reviewed. Multivariate logistic regression and Cox proportional hazards models were used to identify factors associated with pathological complete response (pCR) and survival outcomes. The Kaplan-Meier method was applied to compare survival outcomes between HER2-zero and HER2-low patients. RESULT Four hundred and twenty-one patients meeting the inclusion criteria. The pCR rate of HER2-zero patients was significantly higher than that of HER2-low patients after adjusting for confounders (OR = 2.83, 95% CI: 1.44-5.65, adjusted P = .003). HER2-low patients demonstrated the most stable status after NAC within the hormone receptor (HR)-positive subset (80.82%) and the most unstable status within the HR-negative subset (57.69%). HER2-zero status was associated with worse disease-free survival (DFS) (HR = 1.94, 95% CI: 1.13-3.32, adjusted P = .02), but not with overall survival (OS) (HR = 1.18, 95% CI: 0.59-2.37, P = .65). HER2-zero patients had significantly worse DFS than HER2-low patients in the entire cohort (P = .014) and the HR-positive subset (P < .001). pCR could serve as a surrogate endpoint of favorable survival outcomes for HER2-zero patients, but not for HER2-low patients. CONCLUSION HER2-low patients might exhibit distinct characteristics compared with HER2-zero patients following NAC. Further multicenter, prospective studies are warranted to validate the conclusions of this exploratory research and assess whether HER2-low expression could serve as a new clinical subtype for evaluating NAC treatment outcomes.
Collapse
Affiliation(s)
- Jie Lian
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ru Yao
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Siyuan Pang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xinyu Ren
- Department of Pathology and Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Bo Pan
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Yidong Zhou
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
49
|
Sun ZD, Zhang Y, Yang YS, Liu CY, Pei MQ, Fu WD, He HH. Construction and validation of a novel nomogram for prediction of lymph node metastasis in HER2-positive breast cancer: based on the optimal number of examined lymph nodes for accurate nodal staging. BMC Womens Health 2025; 25:132. [PMID: 40121485 PMCID: PMC11929165 DOI: 10.1186/s12905-025-03663-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: 02/20/2024] [Accepted: 03/07/2025] [Indexed: 03/25/2025] Open
Abstract
PURPOSE This study aimed to construct and validate a novel nomogram for prediction of lymph node metastasis in HER2-positive breast cancer based on the optimal number of examined lymph nodes (ELNs) for accurate nodal staging. METHODS We included 4,040 patients diagnosed with HER2-positive breast cancer from the SEER database, randomly allocating them into training and validation cohorts in a 7:3 ratio. The optimal number of ELNs was identified via piecewise linear regression. The association of ELNs count with nodal migration was evaluated through Logistic Regression (LR) analysis and Random Forest (RF). The nomogram was constructed, and its' performance was evaluated by the receiver operating characteristic curves, calibration curve and Decision curve analysis curves. RESULTS The optimal number of ELNs was 13. LR and RF identified the optimal number of ELNs, radiotherapy status, chemotherapy status, T stage, and grade as independent predictive variables for node metastasis, which were used in the nomogram's construction. And the area under the curve values for the nomogram were 0.829 (95% confidence interval (CI): 0.813-0.845) and 0.833 (95% CI:0.808-0.858) in the training and test split respectively, surpassing those of the optimal number of ELNs (0.649, 95% CI: 0.631-0.667 and 0.676, 95% CI:0.648-0.704). Calibration plots exhibited low Brier scores (0.150 for training split, 0.145 for test split). CONCLUSION This study developed a novel nomogram that integrates the optimal number of ELNs with other independent risk factors, facilitating individualized prediction of lymph node metastasis in patients with HER2-positive breast cancer.
Collapse
Affiliation(s)
- Zhen-Dong Sun
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, Fujian Province, 362000, China
| | - Yan Zhang
- Department of Anesthesiology, Zhuzhou Central Hospital (Zhuzhou Hospital Affiliated to Xiangya School of Medicine), No. 118 South Changjiang Road, Zhuzhou, Hunan Province, 412000, China
| | - Yu-Shen Yang
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, Fujian Province, 362000, China
| | - Chu-Yun Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, Fujian Province, 362000, China
| | - Meng-Qin Pei
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, Fujian Province, 362000, China
| | - Wei-Dong Fu
- Department of Anesthesiology, Zhuzhou Central Hospital (Zhuzhou Hospital Affiliated to Xiangya School of Medicine), No. 118 South Changjiang Road, Zhuzhou, Hunan Province, 412000, China.
| | - He-Han He
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, Fujian Province, 362000, China.
| |
Collapse
|
50
|
Iaboni M, Crivellin F, Arena F, La Cava F, Cordaro A, Stummo F, Faletto D, Huet S, Candela L, Pedrault J, Zanella ER, Bertotti A, Blasi F, Maiocchi A, Poggi L, Reitano E. Complete preclinical evaluation of the novel antibody mimetic Nanofitin-IRDye800CW for diverse non-invasive diagnostic applications in the management of HER-2 positive tumors. Sci Rep 2025; 15:9832. [PMID: 40118987 PMCID: PMC11928573 DOI: 10.1038/s41598-025-93696-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 03/10/2025] [Indexed: 03/24/2025] Open
Abstract
There are well-known limitations associated to the use of antibodies in the non-invasive detection of HER-2 expression. In fact, current procedures recommended for diagnostic purposes of HER-2 status are still invasive techniques. Here, a novel, smaller diagnostic probe, the anti-HER-2 Nanofitin conjugated to the fluorophore IRDye800CW (NF-800), underwent an in vitro/in vivo proof of concept study by Optical Imaging. NF-800 showed high affinity and specificity for the cellular target, and the ability to internalize into HER-2 positive cells. By ex vivo analysis, NF-800 showed a selective tumor accumulation in xenograft tumor models, and also a good tumor targeting efficacy in translational preclinical setups, such as orthotopic and patient-derived xenograft (PDX) models. In the latter, NF-800 was compared to the anti-HER-2 antibody Trastuzumab, displaying a large diagnostic advantage. Interestingly, NF-800 did not seem to share the same binding site with Trastuzumab and Pertuzumab, opening specific theragnostic opportunities for NF-800 in combination with standard-of-care antibodies.
Collapse
Affiliation(s)
- Margherita Iaboni
- Bracco Research Centre, Bracco Imaging Spa, Via Ribes 5, 10010, Colleretto Giacosa, Turin, Italy.
| | - Federico Crivellin
- Bracco Research Centre, Bracco Imaging Spa, Via Ribes 5, 10010, Colleretto Giacosa, Turin, Italy
| | - Francesca Arena
- Bracco Research Centre, Bracco Imaging Spa, Via Ribes 5, 10010, Colleretto Giacosa, Turin, Italy
| | - Francesca La Cava
- Bracco Research Centre, Bracco Imaging Spa, Via Ribes 5, 10010, Colleretto Giacosa, Turin, Italy
| | - Alessia Cordaro
- Bracco Research Centre, Bracco Imaging Spa, Via Ribes 5, 10010, Colleretto Giacosa, Turin, Italy
| | - Francesco Stummo
- Bracco Research Centre, Bracco Imaging Spa, Via Ribes 5, 10010, Colleretto Giacosa, Turin, Italy
| | - Daniele Faletto
- Bracco Research Centre, Bracco Imaging Spa, Via Ribes 5, 10010, Colleretto Giacosa, Turin, Italy
| | - Simon Huet
- Affilogic SAS, 24 Rue de La Rainière, 44300, Nantes, France
| | - Leo Candela
- Affilogic SAS, 24 Rue de La Rainière, 44300, Nantes, France
| | - Jessy Pedrault
- Affilogic SAS, 24 Rue de La Rainière, 44300, Nantes, France
| | - Eugenia R Zanella
- Candiolo Cancer Institute - FPO IRCCS, Strada Provinciale 142, 10060, Candiolo, Turin, Italy
| | - Andrea Bertotti
- Candiolo Cancer Institute - FPO IRCCS, Strada Provinciale 142, 10060, Candiolo, Turin, Italy
- Department of Oncology, University of Turin, Candiolo, Turin, Italy
| | - Francesco Blasi
- Bracco Research Centre, Bracco Imaging Spa, Via Ribes 5, 10010, Colleretto Giacosa, Turin, Italy
| | - Alessandro Maiocchi
- Bracco Research Centre, Bracco Imaging Spa, Via Ribes 5, 10010, Colleretto Giacosa, Turin, Italy
| | - Luisa Poggi
- Bracco Research Centre, Bracco Imaging Spa, Via Ribes 5, 10010, Colleretto Giacosa, Turin, Italy
| | - Erika Reitano
- Bracco Research Centre, Bracco Imaging Spa, Via Ribes 5, 10010, Colleretto Giacosa, Turin, Italy
| |
Collapse
|